1
|
Habelrih T, Augustin TL, Mauffette-Whyte F, Ferri B, Sawaya K, Côté F, Gallant M, Olson DM, Chemtob S. Inflammatory mechanisms of preterm labor and emerging anti-inflammatory interventions. Cytokine Growth Factor Rev 2024; 78:50-63. [PMID: 39048393 DOI: 10.1016/j.cytogfr.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Preterm birth is a major public health concern, requiring a deeper understanding of its underlying inflammatory mechanisms and to develop effective therapeutic strategies. This review explores the complex interaction between inflammation and preterm labor, highlighting the pivotal role of the dysregulation of inflammation in triggering premature delivery. The immunological environment of pregnancy, characterized by a fragile balance of immune tolerance and resistance, is disrupted in preterm labor, leading to a pathological inflammatory response. Feto-maternal infections, among other pro-inflammatory stimuli, trigger the activation of toll-like receptors and the production of pro-inflammatory mediators, promoting uterine contractility and cervical ripening. Emerging anti-inflammatory therapeutics offer promising approaches for the prevention of preterm birth by targeting key inflammatory pathways. From TLR-4 antagonists to chemokine and interleukin receptor antagonists, these interventions aim to modulate the inflammatory environment and prevent adverse pregnancy outcomes. In conclusion, a comprehensive understanding of the inflammatory mechanisms leading to preterm labor is crucial for the development of targeted interventions in hope of reducing the incidence of preterm birth and improving neonatal health outcomes.
Collapse
Affiliation(s)
- Tiffany Habelrih
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Thalyssa-Lyn Augustin
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Félix Mauffette-Whyte
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Béatrice Ferri
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Kevin Sawaya
- Research Center, CHU Sainte-Justine, Montreal, QC, Canada; Programmes de cycles supérieurs en sciences biomédicales, Faculté de médecine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Mathilde Gallant
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
2
|
Ji K, Chen Y, Pan X, Chen L, Wang X, Wen B, Bao J, Zhong J, Lv Z, Zheng Z, Liu H. Single-cell and spatial transcriptomics reveal alterations in trophoblasts at invasion sites and disturbed myometrial immune microenvironment in placenta accreta spectrum disorders. Biomark Res 2024; 12:55. [PMID: 38831319 PMCID: PMC11149369 DOI: 10.1186/s40364-024-00598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/04/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Placenta accreta spectrum disorders (PAS) are a severe complication characterized by abnormal trophoblast invasion into the myometrium. The underlying mechanisms of PAS involve a complex interplay of various cell types and molecular pathways. Despite its significance, both the characteristics and intricate mechanisms of this condition remain poorly understood. METHODS Spatial transcriptomics (ST) and single-cell RNA sequencing (scRNA-seq), were performed on the tissue samples from four PAS patients, including invasive tissues (ST, n = 3; scRNA-seq, n = 4), non-invasive normal placenta samples (ST, n = 1; scRNA-seq, n = 2). Three healthy term pregnant women provided normal myometrium samples (ST, n = 1; scRNA-seq, n = 2). ST analysis characterized the spatial expression landscape, and scRNA-seq was used to identify specific cellular components in PAS. Immunofluorescence staining was conducted to validate the findings. RESULTS ST slices distinctly showed the myometrium in PAS was invaded by three subpopulations of trophoblast cells, extravillous trophoblast cells, cytotrophoblasts, and syncytiotrophoblasts, especially extravillous trophoblast cells. The pathways enriched by genes in trophoblasts, smooth muscle cells (SMC), and immune cells of PAS were mainly associated with immune and inflammation. We identified elevated expression of the angiogenesis-stimulating gene PTK2, alongside the cell proliferation-enhancing gene EGFR, within the trophoblasts of PAS group. Trophoblasts mainly contributed the enhancement of HLA-G and EBI3 signaling, which is crucial in establishing immune escape. Meanwhile, SMC regions in PAS exhibited upregulation of immunomodulatory markers such as CD274, HAVCR2, and IDO1, with CD274 expression experimentally verified to be increased in the invasive SMC areas of the PAS group. CONCLUSIONS This study provided information of cellular composition and spatial organization in PAS at single-cell and spatial level. The dysregulated expression of genes in PAS revealed a complex interplay between enhanced immune escape in trophoblasts and immune tolerance in SMCs during invasion in PAS. These findings will enhance our understanding of PAS pathogenesis for developing potential therapeutic strategies.
Collapse
Affiliation(s)
- Kaiyuan Ji
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yunshan Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Xiuyu Pan
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Lina Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaodi Wang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Bolun Wen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Junjie Bao
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Junmin Zhong
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Zi Lv
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China
| | - Zheng Zheng
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China.
| | - Huishu Liu
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, China.
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Chen W, Tang C, Chen G, Li J, Li N, Zhang H, Di L, Wang R. Boosting Checkpoint Immunotherapy with Biomimetic Nanodrug Delivery Systems. Adv Healthc Mater 2024; 13:e2304284. [PMID: 38319961 DOI: 10.1002/adhm.202304284] [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: 12/04/2023] [Revised: 01/26/2024] [Indexed: 02/08/2024]
Abstract
Immune checkpoint blockade (ICB) has achieved unprecedented progress in tumor immunotherapy by blocking specific immune checkpoint molecules. However, the high biodistribution of the drug prevents it from specifically targeting tumor tissues, leading to immune-related adverse events. Biomimetic nanodrug delivery systems (BNDSs) readily applicable to ICB therapy have been widely developed at the preclinical stage to avoid immune-related adverse events. By exploiting or mimicking complex biological structures, the constructed BNDS as a novel drug delivery system has good biocompatibility and certain tumor-targeting properties. Herein, the latest findings regarding the aforementioned therapies associated with ICB therapy are highlighted. Simultaneously, prospective bioinspired engineering strategies can be designed to overcome the four-level barriers to drug entry into lesion sites. In future clinical translation, BNDS-based ICB combination therapy represents a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Wenjing Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Chenlu Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Guijin Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Hanwen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| |
Collapse
|
4
|
Fu T, Wang X, Zhao X, Jiang Y, Liu X, Zhang H, Ren Y, Li Z, Hu X. Single-cell transcriptomic analysis of decidual immune cell landscape in the occurrence of adverse pregnancy outcomes induced by Toxoplasma gondii infection. Parasit Vectors 2024; 17:213. [PMID: 38730500 PMCID: PMC11088043 DOI: 10.1186/s13071-024-06266-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: 02/13/2024] [Accepted: 03/29/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular parasite that can lead to adverse pregnancy outcomes, particularly in early pregnancy. Previous studies have illustrated the landscape of decidual immune cells. However, the landscape of decidual immune cells in the maternal-fetal microenvironment during T. gondii infection remains unknown. METHODS In this study, we employed single-cell RNA sequencing to analyze the changes in human decidual immune cells following T. gondii infection. The results of scRNA-seq were further validated with flow cytometry, reverse transcription-polymerase chain reaction, western blot, and immunofluorescence staining. RESULTS Our results showed that the proportion of 17 decidual immune cell clusters and the expression levels of 21 genes were changed after T. gondii infection. Differential gene analysis demonstrated that T. gondii infection induced the differential expression of 279, 312, and 380 genes in decidual NK cells (dNK), decidual macrophages (dMφ), and decidual T cells (dT), respectively. Our results revealed for the first time that several previously unknown molecules in decidual immune cells changed following infection. This result revealed that the function of maternal-fetal immune tolerance declined, whereas the killing ability of decidual immune cells enhanced, eventually contributing to the occurrence of adverse pregnancy outcomes. CONCLUSIONS This study provides valuable resource for uncovering several novel molecules that play an important role in the occurrence of abnormal pregnancy outcomes induced by T. gondii infection.
Collapse
Affiliation(s)
- Tianyi Fu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xiaohui Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xiaoyue Zhao
- Department of Clinical Psychology, Yantai Affiliated Hospital of Binzhou Medial University, Yantai, 264100, Shandong, People's Republic of China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yushan Ren
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of 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
|
Zhang YN, Wu Q, Deng YH. Phenotypic characterisation of regulatory T cells in patients with gestational diabetes mellitus. Sci Rep 2024; 14:4881. [PMID: 38418860 PMCID: PMC10902321 DOI: 10.1038/s41598-023-47638-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/16/2023] [Indexed: 03/02/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a common complication that occurs during pregnancy. Emerging evidence suggests that immune abnormalities play a pivotal role in the development of GDM. Specifically, regulatory T cells (Tregs) are considered a critical factor in controlling maternal-fetal immune tolerance. However, the specific characteristics and alterations of Tregs during the pathogenesis of GDM remain poorly elucidated. Therefore, this study aimed to investigate the changes in Tregs among pregnant women diagnosed with GDM compared to healthy pregnant women. A prospective study was conducted, enrolling 23 healthy pregnant women in the third trimester and 21 third-trimester women diagnosed with GDM. Participants were followed up until the postpartum period. The proportions of various Treg, including Tregs, mTregs, and nTregs, were detected in the peripheral blood of pregnant women from both groups. Additionally, the expression levels of PD-1, HLA-G, and HLA-DR on these Tregs were examined. The results revealed no significant differences in the proportions of Tregs, mTregs, and nTregs between the two groups during the third trimester and postpartum period. However, GDM patients exhibited significantly reduced levels of PD-1+ Tregs (P < 0.01) and HLA-G+ Tregs (P < 0.05) in the third trimester compared to healthy pregnant women in the third trimester. Furthermore, GDM patients demonstrated significantly lower levels of PD-1+ mTregs (P < 0.01) and HLA-G+ (P < 0.05) mTregs compared to healthy pregnant women in the third trimester. Overall, the proportion of Tregs did not exhibit significant changes during the third trimester in GDM patients compared to healthy pregnant women. Nevertheless, the observed dysregulation of immune regulation function in Tregs and mTregs may be associated with the development of GDM in pregnant women.
Collapse
Affiliation(s)
- Ya-Nan Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Qin Wu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Yi-Hui Deng
- School of Chinese Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China.
| |
Collapse
|
7
|
Akilla MA, Nchor IAA, Banyeh M, Amidu N. Immune checkpoint molecules B7-1 and B7-H1 as predictive markers of pre-eclampsia: A case-control study in a Ghana. Immun Inflamm Dis 2024; 12:e1142. [PMID: 38270323 PMCID: PMC10777883 DOI: 10.1002/iid3.1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND/AIM Immune tolerance in the fetal-maternal junction is maintained by a balance in the Th1/Th2 system. Th1-type immunity is associated with pro-inflammatory cytokines and immune checkpoint molecules (ICMs) such as B7-H1, while Th2-type immunity is characterized by anti-inflammatory cytokines and ICMs such as B7-1. Any imbalance in the Th1/Th2 immune system may lead to adverse pregnancy outcomes such as pre-eclampsia (PE). Hitherto, the potential of serum B7-1 and B7-H1 proteins as early markers of PE has not been explored in the Ghanaian population. MATERIALS AND METHODS This was a case-control study from May 2020 to April 2022 at the War Memorial and the Upper East Regional Hospitals. The study involved 291 women, including 180 (61.9%) with normotensive pregnancy and 111 (38.1%) with PE. Venous blood samples were collected and assayed for blood cell count, serum interleukins (ILs)-4, -6, -12, -18, and TNF-α as well as serum B7-1 and B7-H1 proteins. RESULTS The monocyte count (p = .007), the serum levels of IL-18 (p = .035), TNF-α (p = .001), and B7-H1 (p = .006) were significantly higher in PE than in normotensive pregnancy. In addition, the monocyte count (p = .002), the serum levels of IL-12 (p = .029), TNF-α (p = .016), and B7-1 (p = .009) levels were significantly higher in the third trimester than the second trimester PE. In predicting PE, the area under the curve of cytokines and ICMs ranged from 0.51 for IL-6 to 0.62 for TNF-α. CONCLUSION PE may be characterized by a dominant Th1-type immunity with higher levels of pro-inflammatory cytokines and B7-H1 proteins, but these variables may not be suitable for predicting PE.
Collapse
Affiliation(s)
- Martin Awe Akilla
- Department of Biomedical Laboratory Science, School of Allied Health SciencesUniversity for Development StudiesTamaleGhana
| | | | - Moses Banyeh
- Department of Biomedical Laboratory Science, School of Allied Health SciencesUniversity for Development StudiesTamaleGhana
| | - Nafiu Amidu
- Department of Biomedical Laboratory Science, School of Allied Health SciencesUniversity for Development StudiesTamaleGhana
| |
Collapse
|
8
|
Shekari N, Shanehbandi D, Kazemi T, Zarredar H, Baradaran B, Jalali SA. VISTA and its ligands: the next generation of promising therapeutic targets in immunotherapy. Cancer Cell Int 2023; 23:265. [PMID: 37936192 PMCID: PMC10631023 DOI: 10.1186/s12935-023-03116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel negative checkpoint receptor (NCR) primarily involved in maintaining immune tolerance. It has a role in the pathogenesis of autoimmune disorders and cancer and has shown promising results as a therapeutic target. However, there is still some ambiguity regarding the ligands of VISTA and their interactions with each other. While V-Set and Immunoglobulin domain containing 3 (VSIG-3) and P-selectin glycoprotein ligand-1(PSGL-1) have been extensively studied as ligands for VISTA, the others have received less attention. It seems that investigating VISTA ligands, reviewing their functions and roles, as well as outcomes related to their interactions, may allow an understanding of their full functionality and effects within the cell or the microenvironment. It could also help discover alternative approaches to target the VISTA pathway without causing related side effects. In this regard, we summarize current evidence about VISTA, its related ligands, their interactions and effects, as well as their preclinical and clinical targeting agents.
Collapse
Affiliation(s)
- Najibeh Shekari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Amir Jalali
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Yang Y, Zhu G, Yang L, Yang Y. Targeting CD24 as a novel immunotherapy for solid cancers. Cell Commun Signal 2023; 21:312. [PMID: 37919766 PMCID: PMC10623753 DOI: 10.1186/s12964-023-01315-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/13/2023] [Indexed: 11/04/2023] Open
Abstract
Cluster of differentiation 24 (CD24), a mucin-like highly glycosylated molecule has been extensively studied as a cancer stem cell marker in a variety of solid cancers. The functional role of CD24 is either fulfilled by combining with ligands or participating in signal transduction, which mediate the initiation and progression of neoplasms. Recently, CD24 was also described as an innate immune checkpoint with apparent significance in several types of solid cancers. Herein, we review the current understanding of the molecular fundamentals of CD24, the role of CD24 in tumorigenesis and cancer progression, the possibility as a promising therapeutic target and summarized different therapeutic agents or strategies targeting CD24 in solid cancers. Video Abstract.
Collapse
Affiliation(s)
- Yan Yang
- Xinxiang Engineering Technology Research Center of Tumor-Targeted Drug Development, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453000, Henan, China
| | - Guangming Zhu
- Clinical Laboratory, The First People's Hospital of Taian, Taian 271000, Shandong, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, Zhengzhou, 450052, Henan, China
| | - Yun Yang
- Xinxiang Engineering Technology Research Center of Tumor-Targeted Drug Development, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453000, Henan, China.
| |
Collapse
|
10
|
Huang L, Tu Z, Wei L, Sun W, Wang Y, Bi S, He F, Du L, Chen J, Kzhyshkowska J, Wang H, Chen D, Zhang S. Generating Functional Multicellular Organoids from Human Placenta Villi. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301565. [PMID: 37438660 PMCID: PMC10502861 DOI: 10.1002/advs.202301565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Indexed: 07/14/2023]
Abstract
The interaction between trophoblasts, stroma cells, and immune cells at the maternal-fetal interface constitutes the functional units of the placenta, which is crucial for successful pregnancy outcomes. However, the investigation of this intricate interplay is restricted due to the absence of efficient experimental models. To address this challenge, a robust, reliable methodology for generating placenta villi organoids (PVOs) from early, late, or diseased pregnancies using air-liquid surface culture is developed. PVOs contain cytotrophoblasts that can self-renew and differentiate directly, along with stromal elements that retain native immune cells. Analysis of scRNA sequencing and WES data reveals that PVOs faithfully recapitulate the cellular components and genetic alterations of the corresponding source tissue. Additionally, PVOs derived from patients with preeclampsia exhibit specific pathological features such as inflammation, antiangiogenic imbalance, and decreased syncytin expression. The PVO-based propagation of primary placenta villi should enable a deeper investigation of placenta development and exploration of the underlying pathogenesis and therapeutics of placenta-originated diseases.
Collapse
Affiliation(s)
- Lijun Huang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Zhaowei Tu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Liudan Wei
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Wei Sun
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yifan Wang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Shilei Bi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Fang He
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Lili Du
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Jingsi Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and ImmunologyMedical Faculty MannheimUniversity of Heidelberg68167MannheimGermany
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health ResearchDepartment of Obstetrics and GynecologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361005China
| | - Dunjin Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
- Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceGuangzhou510150China
- Guangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineGuangzhou510150China
- Guangdong Engineering and Technology Research Center of Maternal‐Fetal MedicineGuangzhou510150China
| | - Shuang Zhang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| |
Collapse
|
11
|
Wang R, Xiong K, Wang Z, Wu D, Hu B, Ruan J, Sun C, Ma D, Li L, Liao S. Immunodiagnosis - the promise of personalized immunotherapy. Front Immunol 2023; 14:1216901. [PMID: 37520576 PMCID: PMC10372420 DOI: 10.3389/fimmu.2023.1216901] [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: 05/04/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023] Open
Abstract
Immunotherapy showed remarkable efficacy in several cancer types. However, the majority of patients do not benefit from immunotherapy. Evaluating tumor heterogeneity and immune status before treatment is key to identifying patients that are more likely to respond to immunotherapy. Demographic characteristics (such as sex, age, and race), immune status, and specific biomarkers all contribute to response to immunotherapy. A comprehensive immunodiagnostic model integrating all these three dimensions by artificial intelligence would provide valuable information for predicting treatment response. Here, we coined the term "immunodiagnosis" to describe the blueprint of the immunodiagnostic model. We illustrated the features that should be included in immunodiagnostic model and the strategy of constructing the immunodiagnostic model. Lastly, we discussed the incorporation of this immunodiagnosis model in clinical practice in hopes of improving the prognosis of tumor immunotherapy.
Collapse
Affiliation(s)
- Renjie Wang
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kairong Xiong
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhimin Wang
- Division of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Di Wu
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bai Hu
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Ruan
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie Liao
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Zheng M, Zhang Z, Yu L, Wang Z, Dong Y, Tong A, Yang H. Immune-checkpoint protein VISTA in allergic, autoimmune disease and transplant rejection. Front Immunol 2023; 14:1194421. [PMID: 37435070 PMCID: PMC10330820 DOI: 10.3389/fimmu.2023.1194421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Negative checkpoint regulators (NCRs) reduce the T cell immune response against self-antigens and limit autoimmune disease development. V-domain Ig suppressor of T cell activation (VISTA), a novel immune checkpoint in the B7 family, has recently been identified as one of the NCRs. VISTA maintains T cell quiescence and peripheral tolerance. VISTA targeting has shown promising results in treating immune-related diseases, including cancer and autoimmune disease. In this review, we summarize and discuss the immunomodulatory role of VISTA, its therapeutic potential in allergic, autoimmune disease, and transplant rejection, as well as the current therapeutic antibodies, to present a new method for regulating immune responses and achieving durable tolerance for the treatment of autoimmune disease and transplantation.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Yu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zeng Wang
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Yijun Dong
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Chen J, Li Y, Xu L, Sang Y, Li D, Du M. Paradoxical expression of NRP1 in decidual stromal and immune cells reveals a novel inflammation balancing mechanism during early pregnancy. Inflamm Res 2023:10.1007/s00011-023-01734-y. [PMID: 37328599 DOI: 10.1007/s00011-023-01734-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 06/18/2023] Open
Abstract
OBJECTIVE AND DESIGN To investigate the balancing mechanisms between decidualization-associated inflammation and pregnancy-related immunotolerance. MATERIAL OR SUBJECTS Decidual samples from women with normal pregnancy (n = 58) or unexplained spontaneous miscarriage (n = 13), peripheral blood from normal pregnancy and endometria from non-pregnancy (n = 10) were collected. Primary endometrial stromal cells (ESCs), decidual stromal cells (DSCs), decidual immune cells (DICs) and peripheral blood mononuclear cells (PBMCs) were isolated. TREATMENT The plasmid carrying neuropilin-1 (NRP1) gene was transfected into ESC for overexpression. To induce decidualization in vitro, ESCs were treated with a combination of 10 nM estradiol, 100 nM progesterone and 0.5 mM cAMP. Anti-Sema3a and anti-NRP1 neutralizing antibodies were applied to block the ligand-receptor interactions. METHODS RNA-seq analysis was performed to identify differentially expressed genes in DSCs and DICs, and NRP1 expression was verified by Western blotting and flow cytometry. The secretion of inflammatory mediators was measured using a multifactor cytometric bead array. The effects of Sema3a-NRP1 pathway on DICs were determined by flow cytometry. Statistical differences between groups were compared using the T test and one way or two-way ANOVA. RESULTS Combined with five RNA-seq datasets, NRP1 was the only immune checkpoint changing oppositely between DSCs and DICs. The decreased expression of NRP1 in DSCs allowed intrinsic inflammatory responses required for decidualization, while its increased expression in DICs enhanced tolerant phenotypes beneficial to pregnancy maintenance. DSC-secreted Sema3a promoted immunosuppression in DICs via NRP1 binding. In women with miscarriage, NRP1 was abnormally elevated in DSCs but diminished in decidual macrophages and NK cells. CONCLUSION NRP1 is a multifunctional controller that balances the inflammatory states of DSCs and DICs in gravid uterus. Abnormal expression of NRP1 is implicated in miscarriage.
Collapse
Affiliation(s)
- Jiajia Chen
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China
| | - Yanhong Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China
| | - Ling Xu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China
| | - Yifei Sang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China.
| | - Meirong Du
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Zhao Zhou Road 413, Shanghai, 200032, China.
- Department of Obstetrics and Gynecology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, 200434, China.
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau SAR, 519020, China.
| |
Collapse
|
14
|
Chen RY, Zhu Y, Shen YY, Xu QY, Tang HY, Cui NX, Jiang L, Dai XM, Chen WQ, Lin Q, Li XZ. The role of PD-1 signaling in health and immune-related diseases. Front Immunol 2023; 14:1163633. [PMID: 37261359 PMCID: PMC10228652 DOI: 10.3389/fimmu.2023.1163633] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
Programmed cell death 1 receptor (PD-1) and its ligands constitute an inhibitory pathway to mediate the mechanism of immune tolerance and provide immune homeostasis. Significantly, the binding partners of PD-1 and its associated ligands are diverse, which facilitates immunosuppression in cooperation with other immune checkpoint proteins. Accumulating evidence has demonstrated the important immunosuppressive role of the PD-1 axis in the tumor microenvironment and in autoimmune diseases. In addition, PD-1 blockades have been approved to treat various cancers, including solid tumors and hematological malignancies. Here, we provide a comprehensive review of the PD-1 pathway, focusing on the structure and expression of PD-1, programmed cell death 1 ligand 1 (PD-L1), and programmed cell death 1 ligand 2 (PD-L2); the diverse biological functions of PD-1 signaling in health and immune-related diseases (including tumor immunity, autoimmunity, infectious immunity, transplantation immunity, allergy and immune privilege); and immune-related adverse events related to PD-1 and PD-L1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Qiang Lin
- *Correspondence: Qiang Lin, ; Xiao-Zhong Li,
| | | |
Collapse
|
15
|
Bailly C, Thuru X, Goossens L, Goossens JF. Soluble TIM-3 as a biomarker of progression and therapeutic response in cancers and other of human diseases. Biochem Pharmacol 2023; 209:115445. [PMID: 36739094 DOI: 10.1016/j.bcp.2023.115445] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Immune checkpoints inhibition is a privileged approach to combat cancers and other human diseases. The TIM-3 (T cell immunoglobulin and mucin-domain containing-3) inhibitory checkpoint expressed on different types of immune cells is actively investigated as an anticancer target, with a dozen of monoclonal antibodies in (pre)clinical development. A soluble form sTIM-3 can be found in the plasma of patients with cancer and other diseases. This active circulating protein originates from the proteolytic cleavage by two ADAM metalloproteases of the membrane receptor shared by tumor and non-tumor cells, and extracellular vesicles. In most cancers but not all, overexpression of mTIM-3 at the cell surface leads to high level of sTIM-3. Similarly, elevated levels of sTIM-3 have been reported in chronic autoimmune diseases, inflammatory gastro-intestinal diseases, certain viral and parasitic diseases, but also in cases of organ transplantation and in pregnancy-related pathologies. We have analyzed the origin of sTIM-3, its methods of dosage in blood or plasma, its presence in multiple diseases and its potential role as a biomarker to follow disease progression and/or the treatment response. In contrast to sPD-L1 generated by different classes of proteases and by alternative splicing, sTIM-3 is uniquely produced upon ADAM-dependent shedding, providing a more homogenous molecular entity and a possibly more reliable molecular marker. However, the biological functionality of sTIM-3 remains insufficiently characterized. The review shed light on pathologies associated with an altered expression of sTIM-3 in human plasma and the possibility to use sTIM-3 as a diagnostic or therapeutic marker.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Consulting Scientific Office, Lille (Wasquehal) 59290, France; University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France.
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Laurence Goossens
- University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| | - Jean-François Goossens
- University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| |
Collapse
|
16
|
Chen Y, Xiao L, Xu J, Wang J, Yu Z, Zhao K, Zhang H, Cheng S, Sharma S, Liao A, Liu C. Recent insight into autophagy and immunity at the maternal-fetal interface. J Reprod Immunol 2023; 155:103781. [PMID: 36463798 DOI: 10.1016/j.jri.2022.103781] [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: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022]
Abstract
Autophagy is a lysosomal degradation pathway that supports metabolic adaptation and energy cycling. It is essential for cell homeostasis, differentiation, development, and survival. Recent studies have shown that autophagy could influence immune responses by regulating immune cell functions. Reciprocally, immune cells strongly influence autophagy. Immune cells at the maternal-fetal interface are thought to play essential roles in pregnancy. Here, we review the induction of autophagy at the maternal-fetal interface and its role in decidualization and placental development. Additionally, we emphasize the role of autophagy in the immune microenvironment at the maternal-fetal interface, including innate immunity, adaptive immunity, and immune tolerance molecules. It also suggests new research directions and prospects.
Collapse
Affiliation(s)
- Yuanyao Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Lin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Jia Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Jingming Wang
- Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Zhiquan Yu
- Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Shibin Cheng
- Department of Pediatrics, Obstetrics and Gynecology and Pathology, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Obstetrics and Gynecology and Pathology, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aihua Liao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| |
Collapse
|
17
|
Singh K. Prenatal Interventions for the Treatment of Congenital Disorders. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
18
|
Habets DHJ, Schlütter A, van Kuijk SMJ, Spaanderman MEA, Al‐Nasiry S, Wieten L. Natural killer cell profiles in recurrent pregnancy loss: Increased expression and positive associations with TACTILE and LILRB1. Am J Reprod Immunol 2022; 88:e13612. [PMID: 36004818 PMCID: PMC9787570 DOI: 10.1111/aji.13612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/15/2022] [Accepted: 08/12/2022] [Indexed: 12/30/2022] Open
Abstract
PROBLEM NK cells are important for healthy pregnancy and aberrant phenotypes or effector functions have been associated with RPL. We compared expression of a broad panel of NK cell receptors, including immune checkpoint receptors, and investigated their clinical association with RPL as this might improve patient stratification and prediction of RPL. METHOD OF STUDY Peripheral blood mononuclear cells were isolated from 52 women with RPL and from 2 women with an uncomplicated pregnancy for flowcytometric analysis and plasma was used to determine anti-CMV IgG antibodies. RESULTS Between RPL and controls, we observed no difference in frequencies of T-, NKT or NK cells, in CD56dimCD16+ or CD56brightCD16- NK cell subsets or in the expression of KIRs, NKG2A, NKG2C, NKG2D, NKp30, NKp44, NKp46 or DNAM1. NK cells from women with RPL had a higher expression of LILRB1 and TACTILE and this was associated with the number of losses. The immune checkpoint receptors PD1, TIM3 and LAG3 were not expressed on peripheral blood NK cells. In RPL patients, there was a large variation in NKG2C expression and higher levels could be explained by CMV seropositivity. CONCLUSION Our study identified LILRB1 and TACTILE as NK cell receptors associated with RPL. Moreover, we provide first support for the potential role of CMV in RPL via its impact on the NK cell compartment. Thereby our study could guide future studies to confirm the clinical association of LILRB1, TACTILE and NKG2C with RPL in a larger cohort and to explore their functional relevance in reproductive success.
Collapse
Affiliation(s)
- Denise H. J. Habets
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtthe Netherlands,Department of Transplantation ImmunologyMaastricht University Medical CentreMaastrichtthe Netherlands,GROW school for Oncology and Developmental BiologyMaastricht UniversityMaastrichtthe Netherlands
| | - Anna Schlütter
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtthe Netherlands
| | - Sander M. J. van Kuijk
- Department of Clinical Epidemiology and Medical Technology AssessmentMaastricht University Medical CentreMaastrichtthe Netherlands
| | - Marc E. A. Spaanderman
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtthe Netherlands,GROW school for Oncology and Developmental BiologyMaastricht UniversityMaastrichtthe Netherlands,Department of Obstetrics and GynecologyRadboud University Medical CentreNijmegenthe Netherlands
| | - Salwan Al‐Nasiry
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtthe Netherlands,GROW school for Oncology and Developmental BiologyMaastricht UniversityMaastrichtthe Netherlands
| | - Lotte Wieten
- Department of Transplantation ImmunologyMaastricht University Medical CentreMaastrichtthe Netherlands,GROW school for Oncology and Developmental BiologyMaastricht UniversityMaastrichtthe Netherlands
| |
Collapse
|
19
|
Wen Z, Zhang Y, Feng J, Aimulajiang K, Aleem MT, Lu M, Xu L, Song X, Li X, Yan R. Excretory/secretory proteins inhibit host immune responses by downregulating the TLR4/NF-κB/MAPKs signaling pathway: A possible mechanism of immune evasion in parasitic nematode Haemonchus contortus. Front Immunol 2022; 13:1013159. [PMID: 36238295 PMCID: PMC9551057 DOI: 10.3389/fimmu.2022.1013159] [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: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Haemonchus contortus is an important parasitic nematode of ruminants. Previous studies showed that H. contortus escape the immunity through complex mechanisms, including releasing excretory/secretory proteins (ESPs) to modulate the host immune response. However, the detailed mechanism through which H. contortus excretory/secretory proteins (HcESPs) promote immune evasion remains unknown. In the present study, we demonstrated that HcESPs inhibit the adaptive immune response of goats including downregulation of immune cell antigen presentation, upregulation of immune checkpoint molecules, activation of the STAT3/PD-L1 pathway, and activation of immunosuppressive regulatory T (Treg) cells. Furthermore, HcESPs reversed the LPS-induced upregulation of pro-inflammatory mediators in PBMCs by inhibiting the TLR4/NF-κB/MAPKs/NLRP3 signaling pathway. Our study provides a better understanding of the evasion mechanisms for H. contortus, which could be helpful in providing an alternative way to prevent the infection of this parasite.
Collapse
Affiliation(s)
- Zhaohai Wen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yue Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiajun Feng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kalibixiati Aimulajiang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Muhammad Tahir Aleem
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Mingmin Lu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lixin Xu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaokai Song
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiangrui Li
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruofeng Yan
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Ruofeng Yan,
| |
Collapse
|
20
|
Xu F, Zou C, Gao Y, Shen J, Liu T, He Q, Li S, Xu S. Comprehensive analyses identify RIPOR2 as a genomic instability-associated immune prognostic biomarker in cervical cancer. Front Immunol 2022; 13:930488. [PMID: 36091054 PMCID: PMC9458976 DOI: 10.3389/fimmu.2022.930488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
Cervical cancer (CC) is a malignancy that tends to have a poor prognosis when detected at an advanced stage; however, there are few studies on the early detection of CC at the genetic level. The tumor microenvironment (TME) and genomic instability (GI) greatly affect the survival of tumor patients via effects on carcinogenesis, tumor growth, and resistance. It is necessary to identify biomarkers simultaneously correlated with components of the TME and with GI, as these could predict the survival of patients and the efficacy of immunotherapy. In this study, we extracted somatic mutational data and transcriptome information of CC cases from The Cancer Genome Atlas, and the GSE44001 dataset from the Gene Expression Omnibus database was downloaded for external verification. Stromal components differed most between genomic unstable and genomic stable groups. Differentially expressed genes were screened out on the basis of GI and StromalScore, using somatic mutation information and ESTIMATE methods. We obtained the intersection of GI- and StromalScore-related genes and used them to establish a four-gene signature comprising RIPOR2, CCL22, PAMR1, and FBN1 for prognostic prediction. We described immunogenomic characteristics using this risk model, with methods including CIBERSORT, gene set enrichment analysis (GSEA), and single-sample GSEA. We further explored the protective factor RIPOR2, which has a close relationship with ImmuneScore. A series of in vitro experiments, including immunohistochemistry, immunofluorescence, quantitative reverse transcription PCR, transwell assay, CCK8 assay, EdU assay, cell cycle detection, colony formation assay, and Western blotting were performed to validate RIPOR2 as an anti-tumor signature. Combined with integrative bioinformatic analyses, these experiments showed a strong relationship between RIPOR2 with tumor mutation burden, expression of genes related to DNA damage response (especially PARP1), TME-related scores, activation of immune checkpoint activation, and efficacy of immunotherapy. To summarize, RIPOR2 was successfully identified through comprehensive analyses of the TME and GI as a potential biomarker for forecasting the prognosis and immunotherapy response, which could guide clinical strategies for the treatment of CC patients.
Collapse
Affiliation(s)
- Fangfang Xu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Zou
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yueqing Gao
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiacheng Shen
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingwei Liu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Shaohua Xu, ; Shuangdi Li, ; Qizhi He,
| | - Shuangdi Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Shaohua Xu, ; Shuangdi Li, ; Qizhi He,
| | - Shaohua Xu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Shaohua Xu, ; Shuangdi Li, ; Qizhi He,
| |
Collapse
|
21
|
Affiliation(s)
- Gil Mor
- C.S Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
22
|
Isolation of Decidual Macrophages and Hofbauer Cells from Term Placenta-Comparison of the Expression of CD163 and CD80. Int J Mol Sci 2022; 23:ijms23116113. [PMID: 35682791 PMCID: PMC9181726 DOI: 10.3390/ijms23116113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: Placental immune cells are playing a very important role in a successful placentation and the prevention of pregnancy complications. Macrophages dominate in number and relevance in the maternal and the fetal part of the placenta. The evidence on the polarization state of fetal and maternal macrophages involved in both, healthy and pregnancy-associated diseases, is limited. There is no representative isolation method for the direct comparison of maternal and fetal macrophages so far. (2) Material and Methods: For the isolation of decidual macrophages and Hofbauer cells from term placenta, fresh tissue was mechanically dissected and digested with trypsin and collagenase A. Afterwards cell enrichment was increased by a Percoll gradient. CD68 is represented as pan-macrophage marker, the surface markers CD80 and CD163 were further investigated. (3) Results: The established method revealed a high cell yield and purity of the isolated macrophages and enabled the comparison between decidual macrophages and Hofbauer cells. No significant difference was observed in the percentage of single CD163+ cells in the distinct macrophage populations, by using FACS and immunofluorescence staining. A slight increase of CD80+ cells could be found in the decidual macrophages. Considering the percentage of CD80+CD163− and CD80−CD163+ cells we could not find differences. Interestingly we found an increased number of double positive cells (CD80+CD163+) in the decidual macrophage population in comparison to Hofbauer cells. (4) Conclusion: In this study we demonstrate that our established isolation method enables the investigation of decidual macrophages and Hofbauer cells in the placenta. It represents a promising method for direct cell comparison, enzyme independently, and unaffected by magnetic beads, to understand the functional subsets of placental macrophages and to identify therapeutic targets of pregnancy associated diseases.
Collapse
|
23
|
Zhang T, Shen HH, Qin XY, Li MQ. The metabolic characteristic of decidual immune cells and their unique properties in pregnancy loss. Immunol Rev 2022; 308:168-186. [PMID: 35582842 DOI: 10.1111/imr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.
Collapse
Affiliation(s)
- Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue-Yun Qin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,Shanghai Medical School, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|