1
|
Wen X, Yan X, Xiao Y, Wang J, Jiang S, Liu T, Zhou L. Characteristics in patients with unexplained recurrent spontaneous abortion and the impact on immune, coagulation, and inflammatory profiles. J Matern Fetal Neonatal Med 2025; 38:2477069. [PMID: 40089835 DOI: 10.1080/14767058.2025.2477069] [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: 10/22/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Unexplained Recurrent Spontaneous Abortion (RSA) impacts the physical and psychological well-being of women of reproductive age. This study aimed to reveal the potential mechanisms behind unexplained RSA from the perspective of glucose and lipid metabolic profiles before conception and their relationship with immune, coagulation, and inflammatory markers. METHODS A retrospective analysis was conducted on 100 patients with unexplained recurrent spontaneous abortion at our gynecology department from June 2022 to June 2023. According to whether abnormal glucose and lipid metabolism exist, patients were grouped into normal group and abnormal group. The study compared the glucose and lipid metabolic characteristics, immune status, coagulation, and inflammatory parameters between two groups. RESULTS The Abnormal Group exhibited higher fasting blood glucose, 2-hour oral glucose tolerance test, homeostatic model assessment of insulin resistance, total cholesterol, and LDL cholesterol, along with lower levels of HDL cholesterol. Additionally, alterations in immune profile parameters, including lower levels of CD4+ T cells, CD8+ T cells, B cells, NK cells, and elevated IL-6, were observed in the Abnormal Group. Regarding coagulation and inflammatory profile parameters, the Abnormal Group demonstrated prolonged prothrombin time, activated partial thromboplastin time, elevated fibrinogen and D-dimer levels, and increased platelet count, along with elevated C-reactive protein, TNF-α, and IL-1β levels. Correlation analysis revealed significant associations between the pre-pregnancy metabolic characteristics and immune, coagulation, and inflammatory profile parameters. CONCLUSION Abnormal glucose and lipid metabolism may lead to or exacerbate unexplained RSA in some patients, and therapeutic interventions targeting glucose and lipid metabolism hold promise for improving pregnancy outcomes in this population.
Collapse
Affiliation(s)
- Xi Wen
- Department of Gynecology and Obstetrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoying Yan
- Department of Gynecology and Obstetrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Xiao
- Department of Reproductive Medical Center, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyi Wang
- Sixth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shan Jiang
- Department of Gynecology and Obstetrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tong Liu
- Department of Gynecology and Obstetrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liane Zhou
- Department of Gynecology and Obstetrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Liu G, Wang H, Li X, Mi Y, Zhang C, Chen Y, Miao L, Long H, He J, Ge Q, Liu Y. Biodistribution and persistence of human umbilical cord-derived mesenchymal stem cells in NCG mice: a comparative study. Future Sci OA 2025; 11:2471723. [PMID: 40035430 PMCID: PMC11881841 DOI: 10.1080/20565623.2025.2471723] [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: 07/02/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION This study aims to investigate the biodistribution and persistence of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in NCG mice post-intravenous injection, utilizing 89Zr-PET/CT, bioluminescence imaging, multiplex immunohistochemistry (mIHC), and quantitative polymerase chain reaction (qPCR). METHODS hUC-MSCs were labeled with 89Zr-oxine (89Zr-MSCs) or transduced with luciferase gene (Luc-MSCs). Real-time tracking of 89Zr-MSCs lasted for 14-days followed by mIHC staining of hCD73. Real-time tracking of Luc-MSCs lasted for 7-days, followed by mIHC staining of hCD73 and human Alu-based qPCR. All methods adhered to ICH and other regulatory guidelines for development of cell-based drugs. RESULTS A biodistribution and persistence pattern was observed in the order of lung > liver > kidney > >spleen, although discrepancies were noted for the liver and kidney. CONCLUSION Each method exhibited strengths and weaknesses: 89Zr-PET/CT enabled long-term tracking but encountered issues with 89Zr shedding and dead cells; bioluminescence provided specific detection but was hampered by a rapid decline in signal; mIHC identified cells but relied on antigen abundance; qPCR detected minimal cell quantities but was unable to differentiate between live and dead cells. These limitations may obscure the true fate of cells in vivo, highlighting the need for more accurate and reliable assessment techniques.
Collapse
Affiliation(s)
- Guangyang Liu
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Herui Wang
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Xin Li
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Yi Mi
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Chenliang Zhang
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Yaoyao Chen
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Li Miao
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Haomiao Long
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| | - Jun He
- Centre for Safety Evaluation and Research of Drugs, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Science, Beijing, China
| | - Qinggang Ge
- Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Yongjun Liu
- Stem Cell Biology and Regenerative Medicine Apartment, Yi-Chuang Institute of Bio-Industry, Beijing, China
| |
Collapse
|
3
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
4
|
Yang SH, Yang L, Shi Y, Xu HR, Gan J, Shi JX, Zhang Y, Shen SL, Wang J, Zhang X. GDF15 promotes trophoblast invasion and pregnancy success via the BMPR1A/BMPR2/p-SMAD1 pathway: Implications for recurrent miscarriage. Life Sci 2025; 371:123586. [PMID: 40157640 DOI: 10.1016/j.lfs.2025.123586] [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: 11/25/2024] [Revised: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Insufficient invasion of extravillous trophoblasts (EVTs) is associated with adverse pregnancy outcomes, including recurrent miscarriage (RM). Dysregulated expression of growth differentiation factor 15 (GDF15) has been implicated in RM, but the underlying mechanism remains unclear. This study investigated the role of GDF15 in EVTs function and pregnancy outcomes. Spearman correlation analysis revealed a positive correlation between GDF15 and both BMPR1A and BMPR2 in EVTs. Furthermore, GDF15, BMPR1A, BMPR2, and phosphorylated SMAD1 (p-SMAD1) expression were significantly reduced in placental tissue from RM patients compared to Normal controls. Mechanistically, GDF15 activated the p-SMAD1 signaling pathway, inducing expression of its downstream targets, ID1 and Snail, and enhancing migratory and invasive activity in HTR-8/SVneo cells through interaction with the BMPR1A-BMPR2 receptor complex. Eriodictyol, a small molecule activator of BMPR2, was identified and shown to improve pregnancy outcomes in a mouse model of lipopolysaccharide (LPS)-induced early pregnancy loss (EPL). Eriodictyol can also enhance EVTs migration and invasion as well as activated the p-SMAD1 pathway by activating BMPR2. In conclusion, this study identifies BMPR1A as a receptor for GDF15 in EVTs and demonstrates that GDF15 promotes EVTs invasion and improves pregnancy outcomes via the BMPR1A/BMPR2/p-SMAD1 signaling axis. Eriodictyol, acting as a BMPR2 agonist, may offer a novel therapeutic strategy for preventing early pregnancy loss.
Collapse
Affiliation(s)
- Shu-Han Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Long Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Yan Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Hao-Ran Xu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Jie Gan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Jia-Xin Shi
- Institute of Pathology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Yu Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Shi-Long Shen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China
| | - Jian Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China.
| | - Xuan Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Medical School, Fudan University, Shanghai 200237, China.
| |
Collapse
|
5
|
Mebarki M, Moine-Picard C, Enjaume-Rauch R, Laurent-Puig A, Suissa A, Feyants V, Larghero J, Cras A. Pooling umbilical cord-mesenchymal stromal cells derived from selected multiple donors reduces donor-dependent variability and improves their immunomodulatory properties. Stem Cell Res Ther 2025; 16:252. [PMID: 40394664 PMCID: PMC12093856 DOI: 10.1186/s13287-025-04361-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Umbilical Cord-derived Mesenchymal Stromal Cells (UC-MSCs) display high immunoregulatory properties, offering new perspectives to treat severe immune and inflammatory diseases. However, the heterogeneity of their biological properties remains a challenge to predict clinical response. The aim of our study is to evaluate a strategy based on the constitution of a pool of several pre-selected donors to reduce the biological variability of UC-MSCs and improve their immunomodulatory properties. METHODS Umbilical cords were collected from 10 healthy donors. Isolated UC-MSCs were characterized in the basal state and after a pro-inflammatory priming in vitro by interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα). Proliferation, immunophenotype, the expression of activation markers and the inhibition of T cell proliferation in vitro were assessed in UC-MSCs from selected single donors and from pools. RESULTS Our study highlights the donor-dependent heterogeneity of UC-MSCs immunomodulatory functions. Based on their ability to suppress T-cell proliferation in vitro, we classified donors into three profiles: high, medium and low. Preparation of pools containing UC-MSCs derived from each profile in a 1:1:1 ratio reduced the donor-dependent variability and, most importantly, improved the lowest immunomodulatory functions. After priming with pro-inflammatory cytokines, the inhibition of T-cell expansion by the pooled UC-MSCs was significantly higher than the low donor and the theoretical mean of individual donors, and was associated with increased expression of the key immunoregulatory proteins. Interestingly, the pool did not induce a cumulative immunogenic effect: expression of HLA or costimulatory molecules between the high donor and the pool were similar. Finally, pooling UC-MSCs derived from high and low donors in a 1:2 ratio was sufficient to enhance the lowest immunomodulatory properties. CONCLUSION Overall, our results demonstrate that pooled UC-MSCs with selected high donor offers a new strategy to optimize the immunomodulatory functions of allogeneic UC-MSC-based medicinal products.
Collapse
Affiliation(s)
- Miryam Mebarki
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France.
- INSERM UMR1342, Université Paris Cité, 75010, Paris, France.
- Faculté de Pharmacie, Université Paris Cité, 75006, Paris, France.
| | - Coralie Moine-Picard
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
- INSERM UMR1342, Université Paris Cité, 75010, Paris, France
- Faculté de Pharmacie, Université Paris Cité, 75006, Paris, France
| | - Romain Enjaume-Rauch
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
| | - Antoine Laurent-Puig
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
| | - Annaelle Suissa
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
| | - Valentine Feyants
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
| | - Jérôme Larghero
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France
- INSERM UMR1342, Université Paris Cité, 75010, Paris, France
- AP-HP, Centre MEARY de Thérapie Cellulaire et Génique, Hôpital Saint-Louis, 75010, Paris, France
| | - Audrey Cras
- AP-HP, Unité de Thérapie Cellulaire, INSERM Centre d'Investigation Clinique en Biothérapies CIC-BT, Hôpital Saint-Louis, 75010, Paris, France.
- INSERM UMR1342, Université Paris Cité, 75010, Paris, France.
- Faculté de Pharmacie, Université Paris Cité, 75006, Paris, France.
| |
Collapse
|
6
|
Yang C, Feng X, Guo Y, Shaukat A, Wu Z, Chen Y, Meng L, Deng G. Bta-miR-93 regulates the maternal-fetal tolerance in dairy cows via promoting Programmed Cell Death Ligand 1 mediated M2 macrophage polarization†. Biol Reprod 2025; 112:880-894. [PMID: 40079674 DOI: 10.1093/biolre/ioaf046] [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: 09/22/2024] [Revised: 12/23/2024] [Accepted: 03/12/2025] [Indexed: 03/15/2025] Open
Abstract
Bovine embryo resorption is one of the key factors restricting the reproductive efficiency in dairy cattle, which mainly occurs in the early stage of maternal recognition of pregnancy, causing substantial economic losses to the dairy industry. Macrophages, the most numerous endometrial immune cells in cows during early pregnancy, are critical in developing maternal-fetal immune tolerance. However, the mechanism of their action on the maternal-fetal interface of dairy cows remains unknown. MicroRNAs have important roles in immune tolerance and macrophage polarization, but the underlying mechanisms still need further investigation. In previous laboratory studies, RNA-sequencing revealed that bta-miR-93 was dramatically reduced in bovine endometrial luminal epithelial cells (bEECs) upon IFN-tau stimulation. In the current study, it is revealed that the expression of bta-miR-93 was decreased substantially in the endometrium of pregnant cows and negatively correlated with that of Programmed Cell Death Ligand 1 (PD-L1). In vitro experiments displayed that suppression of bta-miR-93 promoted M2 polarization via promoting PD-L1/PD-1/AKT/mTOR signaling pathway in bEECs and bovine macrophage co-culture model, and vice versa. The 3'-untranslated region of PD-L1 is directly regulated by bta-miR-93. Furthermore, our in vivo studies revealed that overexpression of bta-miR-93 efficiently leads to embryo resorption and suppression of M2 polarization in mice.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xinyu Feng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yingfang Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
- School of Physical Education and International Equestrianism, Wuhan Business University, Wuhan, People's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Lu Meng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
7
|
Zhang X, Sun X, Luo Y, Wang X, Mao C, Xu Z, Song Y, Yan J. Endogenous sulfur dioxide deficiency impairs bone regeneration through abolishing S-sulfenylating p38 at cysteine 211. Int Immunopharmacol 2025; 158:114814. [PMID: 40347882 DOI: 10.1016/j.intimp.2025.114814] [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: 09/22/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Bone defects result in substantial medical expenses and a diminished quality of life for patients. Macrophage polarization is crucial in the bone regeneration process mediated by bone marrow-derived mesenchymal stem cells (BMSCs). macrophage-derived sulfur dioxide (SO2), the fourth endogenous gas signaling molecule, following hydrogen sulfide (H2S), has been shown to regulate macrophage chemotaxis and inflammatory responses. Nevertheless, the specific regulatory effects and mechanisms of macrophage-derived SO2 on bone regeneration are not yet fully understood. This study reveals for the first time that the absence of macrophage-derived SO2 promotes M1 macrophage polarization, whereas the administration of exogenous SO2 donors inhibits M1 polarization. The deficiency of macrophage-derived SO2 results in impaired osteogenic differentiation of BMSCs, whereas the administration of SO2 donors enhances this differentiation process. Further investigations have elucidated that p38α MAPK (p38) is crucial in mediating SO2's effects on M1 macrophage polarization and BMSCs osteogenic differentiation. Mechanistically, SO2 induces S-sulfenylation of p38 in macrophages, an effect that can be reversed by the thiol reductant dithiothreitol. Additionally, the C211S mutation in p38 abrogates the SO2-induced S-sulfenylation of p38, thereby preventing the inhibition of p38 activation and subsequently disrupting the regulation of M1 macrophage polarization and BMSCs osteogenic differentiation. In a model of mouse calvarial bone defects, we consistently observed that inhibiting SO2 production using the SO2-generating enzyme inhibitor HDX impaired bone regeneration capacity in mice, whereas the administration of an SO2 donor enhanced this capacity. In summary, macrophage-derived SO2 S-sulfenylates p38 at cysteine 211, thereby suppressing p38 activation, which inhibits M1 polarization and subsequently maintains the osteogenic differentiation of BMSCs. This study is the first to elucidate the role and mechanism of SO2 in sustaining osteogenesis, offering a novel strategy for addressing bone defect-related disorders.
Collapse
Affiliation(s)
- Xuanming Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xutao Sun
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yumeng Luo
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaoyan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Caiyun Mao
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zihang Xu
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yunjia Song
- Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Pimentel A, Gonçalves-Silva T, Jasmin, Mendez-Otero R. Isolation and characterization of canine umbilical cord mesenchymal/stromal stem cells. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01023-4. [PMID: 40325278 DOI: 10.1007/s11626-025-01023-4] [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: 11/05/2024] [Accepted: 01/30/2025] [Indexed: 05/07/2025]
Abstract
Mesenchymal stem cells (MSCs) have therapeutic potential due to their immunomodulatory and anti-inflammatory properties. In veterinary medicine, adipose tissue is the most common source of MSCs to treat canine disease, but the collection process is invasive, and the cells are influenced by the age and health conditions of the donor. These problems enhance interest in seeking alternative MSC sources, such as perinatal tissues. In this study, we developed and validated an optimized protocol for isolating canine umbilical cord MSCs for application in veterinary therapies. Umbilical cords obtained from cesarean sections were processed using three different protocols, involving combinations of mechanical and enzymatic tissue dissociation. The cells were cultured and evaluated for membrane receptors by flow cytometry to identify MSCs and assessed for their differentiation capacity. The number of cells obtained did not differ significantly between the combined protocol with trypsin and collagenase (TRIP + COL) and the collagenase protocol (COL). In in vitro culture, the combined TRIP + COL and COL yielded 12 to 14 times more cells, respectively, in the first passage than the explant (EXP) group, within fewer days of culture. Additionally, the cells obtained from these protocols showed a greater capacity for expansion over passages, and cells from both protocols showed fibroblast-like morphology and proliferation capacity up to the sixth passage. The cells obtained from these protocols were characterized by phenotype: CD45-, CD34-, CD14-, HLA-DR-, CD29+, CD44+, and CD90+, consistent with MSC identity. However, CD90 expression in the cells decreased significantly at sixth passage. Regarding differentiation, cells obtained from the COL protocol showed a capacity for commitment to the chondrogenic and osteogenic lineages. In conclusion, the COL and TRIP + COL protocols were more effective than the EXP protocol in terms of both the number and quality of isolated cells. However, due to its less-aggressive enzymatic nature, we considered the COL protocol to be the best method to obtain canine MSCs.
Collapse
Affiliation(s)
- Aline Pimentel
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro (IBCCF-UFRJ), Av. Carlos Chagas Filho, 373, Rio de Janeiro, RJ 21941-170, Brazil.
| | - Triciana Gonçalves-Silva
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (CENABIO-UFRJ), Av. Carlos Chagas Filho, 373, Rio de Janeiro, RJ 21941-170, Brazil
| | - Jasmin
- Duque de Caxias Campus, NUMPEX-Bio, Federal University of Rio de Janeiro, Estrada de Xerém, 27 - Xerém, Duque de Caxias, RJ 25245-390, Brazil
| | - Rosalia Mendez-Otero
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro (IBCCF-UFRJ), Av. Carlos Chagas Filho, 373, Rio de Janeiro, RJ 21941-170, Brazil
| |
Collapse
|
9
|
Fan J, Zhang J, Bi X, Zhu P, Wang J, Su D, Shi W, Liu Y, Liu H, Wu X. Upregulation of OTULIN Alleviated Recurrent Pregnancy Loss by Suppressing Trophoblast Dysfunction and NF-κB Signaling Pathway. Mol Reprod Dev 2025; 92:e70029. [PMID: 40372975 DOI: 10.1002/mrd.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
Recurrent pregnancy loss (RPL) is one of obstetrical diseases with no effective therapy methods. Trophoblast cell dysfunction and inflammation induce embryo implantation insufficiency, thereby resulting in RPL. OTU deubiquitinase with linear linkage specificity (OTULIN) plays a role in regulating the immune response and cell death. However, the role of OTULIN in RPL remains unclear. Spontaneous abortion mouse model and lipopolysaccharide-treated HTR-8/SVneo cells were used to investigate the role of OTULIN in RPL. OTULIN expression was downregulated in the labyrinth trophoblast of RPL mice and LPS-treated trophoblast cells. The embryonic reabsorption rate was decreased in OTULIN-overexpressed spontaneous abortion mice, accompanied with the increase in placental/fetus weight ratio. OTULIN overexpression significantly inhibited apoptosis in vivo and in vitro, as evidenced by the decrease in the activity of caspase 3. The expression of pro-inflammatory cytokines was decreased with OTULIN overexpression. Moreover, OTULIN overexpression decreased p-IκBα/IκBα and p-p65/p65 ratio. The nuclear translocation of NF-κB was suppressed via OTULIN overexpression both in vivo and in vitro. Our study suggested that OTULIN deficiency might cause inflammation and trophoblast abnormalities in RPL. The supplementation with OTULIN might alleviate the development of RPL via inhibiting NF-κB mediated inflammation response.
Collapse
Affiliation(s)
- Junmei Fan
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Junkun Zhang
- Department of Intensive Care Unit, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xingyu Bi
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Pengfei Zhu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Jinbao Wang
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Dan Su
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Wenjing Shi
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Yanling Liu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Huiping Liu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
Cheng C, Guo L, Xu Y, Xiong R, Zheng L, Peng Y, Hua R. ChIP-seq and RNA-seq Reveal the Involvement of Histone Lactylation Modification in Early Pregnancy with Subclinical Hypothyroidism. Biochem Genet 2025:10.1007/s10528-025-11095-2. [PMID: 40252140 DOI: 10.1007/s10528-025-11095-2] [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: 01/04/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025]
Abstract
Subclinical hypothyroidism (SCH) is associated with multiple adverse outcomes in early pregnancy. This study aims to explore the regulatory mechanisms underlying histone lactylation modification in early pregnancy with SCH. Peripheral blood mononuclear cells were collected from early pregnant women with or without SCH. RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses were performed to identify the transcriptional pattern and histone lactylation modification in early pregnancy with SCH. RNA-seq analysis revealed that the differentially expressed genes associated with the extracellular matrix exhibited a significant downregulation in early pregnancy with SCH (EP_SCH) compared to early pregnancy without SCH (EP), while those involved in apoptosis were significantly upregulated. In the ChIP-seq analysis, 1660 hypomodified and 766 hypermodified H3K18la-binding peaks were identified in the EP_SCH group compared to the EP group. The hypomodified genes in early pregnancy with SCH compared to its control were enriched in GO terms of apoptotic process and differentiation of immune cells. The genes with increased H3K18 lactylation in early pregnancy with SCH compared to its control were associated with the nervous system, female pregnancy, and the OXT signaling pathway. When RNA-seq data was integrated with ChIP-seq data, we found that the expression and H3K18la enrichment of KCTD7, SIPA1L2, HDAC9, BCL2L14, TXNRD1, and SGK1 were increased in early pregnancy with SCH compared to its control, which was further confirmed by RT-qPCR and ChIP-PCR analyses. This study identifies the changes in histone lactylation modification in early pregnancy with SCH. These findings provide novel insights into the regulatory mechanisms of SCH during early pregnancy.
Collapse
Affiliation(s)
- Chaofei Cheng
- Department of Gynecology and Obstetrics, Zengcheng Central Hospital, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Lizhen Guo
- Department of Gynecology and Obstetrics, Jihua Hospital, Guangzhou, 510000, Guangdong, China
| | - Yinjuan Xu
- Department of Gynecology and Obstetrics, Zengcheng Central Hospital, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Rongzhu Xiong
- Department of Gynecology and Obstetrics, Zengcheng Central Hospital, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Leirong Zheng
- Department of Gynecology and Obstetrics, Zengcheng Central Hospital, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yanmei Peng
- Department of Gynecology and Obstetrics, Zengcheng Central Hospital, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Rui Hua
- Reproductive Medicine Center, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
11
|
Ye YM, Zhao YX, Xiang LR, Zou CY, Xiao H, Lu H, Yang H, Hu JJ, Xie HQ. The Immunomodulatory mechanism and research progress of mesenchymal stem cells in the treatment of allergic rhinitis. Stem Cell Res Ther 2025; 16:188. [PMID: 40251675 PMCID: PMC12008879 DOI: 10.1186/s13287-025-04333-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Allergic rhinitis (AR) affects 10-40% of the global population, yet current therapies (drugs, immunotherapy) face limitations in efficacy and safety. Mesenchymal stem cells (MSCs) have emerged as a promising alternative due to their immunomodulatory properties. KEY FINDINGS Preclinical studies demonstrate that MSCs from adipose, bone marrow, umbilical cord, and tonsils reduce AR symptoms (sneezing, nasal inflammation) and serum IgE (Immunoglobulin E) levels by restoring Th1/Th2 immune equilibrium and enhancing Treg (Regulatory T cells) activity. MSC-derived exosomes and hydrogel-encapsulated formulations further improve targeting and safety. However, clinical translation is hindered by heterogeneous protocols and unresolved long-term risks (e.g., tumorigenicity). CLINICAL SIGNIFICANCE MSC-based therapies offer potential for durable AR remission by addressing immune dysregulation at its root. Future efforts must prioritize standardized production, phase I safety trials, and combination strategies (e.g., exosomes + hydrogels) to accelerate clinical adoption.
Collapse
Affiliation(s)
- Yu-Meng Ye
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yu-Xin Zhao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Li-Rong Xiang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Hao Xiao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Huan Lu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Otolaryngology-Head & Neck Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610213, P. R. China
| | - Hui Yang
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Juan-Juan Hu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| |
Collapse
|
12
|
Warren AJ, Liu L, O'Toole DP, Laffey JG, Masterson CH. The impact of the inflammatory pulmonary microenvironment on the behavior and function of mesenchymal stromal cells. Expert Rev Respir Med 2025:1-12. [PMID: 40223328 DOI: 10.1080/17476348.2025.2491715] [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: 11/21/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Acute respiratory distress syndrome is characterized by the dysregulation and activation of several inflammatory pathways which lead to widespread inflammation in the lungs. Presently, direct therapy is unavailable and the use of mesenchymal stromal cells as a direct therapy has been proposed, as early-phase studies have shown promise. AREAS COVERED MSCs exert various therapeutic effects on the inflammatory microenvironment, such as anti-microbial effects, restoration of the alveolar-capillary barrier, and exuding various anti-inflammatory effects. However, to exert these effects MSCs need to be submitted to specific external stimuli which can affect their immunomodulation, survival, migration and metabolic state. This review references several articles found through targeted searches in PubMed [Accessed between November 2024 and March 2025], for key terms such as 'mesenchymal stromal cells', 'inflammatory microenvironment', anti-inflammatory', 'metabolism', and 'immunomodulation'. EXPERT OPINION The advancement of MSCs therapy in the treatment of ARDS has not progressed as effectively as one might have anticipated. Several clinical findings have established patient subgroups based on inflammatory cytokine profiles and severity of ARDS. This variation in patients may influence the clinical efficacy of MSCs and instead of concluding that MSCs therapy is not worth pursuing, more research is needed to develop an appropriate therapy.
Collapse
Affiliation(s)
- Abigail Jm Warren
- Anaesthesia, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Lanzhi Liu
- Physiology, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Daniel P O'Toole
- Physiology, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland
| | - Claire H Masterson
- Physiology, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
13
|
He W, Zhao Y, Yin L, Du Q, Ren W, Mao L, Liu A, Wang D, Qian J. The transcription factor XBP1 regulates mitochondrial remodel and autophagy in spontaneous abortion. Int Immunopharmacol 2025; 152:114398. [PMID: 40068517 DOI: 10.1016/j.intimp.2025.114398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
PURPOSE Spontaneous abortion (SA) remains a clinical challenge in early pregnancy. It has been reported that endoplasmic reticulum stress (ERS) is implicated in pregnancy-related complications. However, the precise mechanistic role of ERS in SA pathogenesis remains elusive. This study aims to explore the therapeutic potential of targeting ERS-related decidual dysfunction in SA. METHODS An ERS model was established in both decidualized stromal cells (DSCs) and pregnant mice through tunicamycin (Tu) administration. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays were performed to determine the interaction between XBP1s and the transcription factor binding site (TFBS) of tumor necrosis factor receptor-associated factor 6 (TRAF6). Mitochondrial membrane potential (MMP) and mitochondrial function were assessed using JC-1 and TMRM staining following ERS induction in DSCs. The effects of XBP1s inhibitors on mitochondrial metabolism and autophagy were evaluated through RT-qPCR, Western blotting, RNA-Seq, TUNEL assays, ROS and MitoSOX detection, and histological analyses in Tu-treated DSCs and SA patients. STF-083010 (STF) or shXBP1 was utilized to assess the inhibitory effects of X-box binding protein 1 (XBP1s) on DSC function both in vitro and in vivo. RESULTS We observed significant upregulation of XBP1s in decidual tissues from SA patients and Tu-exposed DSCs. Tu exposure significantly increased the proportion of TUNEL-positive cells and upregulated pro-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18) in DSCs. XBP1s inhibition via shXBP1 or pharmacological inhibitor STF attenuated Tu-induced apoptosis and inflammatory cytokine expression. Notably, STF or shXBP1 treatment enhanced MMP and upregulated LC3-II expression in Tu-treated DSCs, indicating autophagy activation.Intriguingly, chloroquine (CQ)-mediated autophagy suppression exacerbated apoptosis in STF/Tu-co-treated DSCs, suggesting that XBP1s inhibition confers cytoprotection through autophagy induction. Mechanistically, XBP1s directly bound to the TFBS of TRAF6, a ubiquitin E3 ligase. TRAF6 overexpression exacerbated mitochondrial dysfunction and apoptosis while suppressing autophagy via inhibition of mTORC2/Akt pathway in Tu-treated DSCs. CONCLUSION XBP1s inhibition restored mitochondrial homeostasis and promoted autophagy by modulating the TRAF6/mTORC2 axis under ERS conditions, providing novel mechanistic insights into SA pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Weihua He
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yating Zhao
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lijun Yin
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiangxing Du
- Department of Obstetrics and Gynecology, Jingning She Autonomous County People's Hospital, Lishui, Zhejiang, China
| | - Wenfen Ren
- Department of Obstetrics and Gynecology, Jingning She Autonomous County People's Hospital, Lishui, Zhejiang, China
| | - Liwei Mao
- Department of Obstetrics and Gynecology, Jingning She Autonomous County People's Hospital, Lishui, Zhejiang, China
| | - Aixia Liu
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China.
| | - Dimin Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Kurniadi A, Zucha MA, Kusumanto A, Salima S, Harsono AB. Application of adult stem cells in obstetrics and gynecology: A scoping review. Eur J Obstet Gynecol Reprod Biol X 2025; 25:100367. [PMID: 39967607 PMCID: PMC11833614 DOI: 10.1016/j.eurox.2025.100367] [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: 12/10/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Background Advancements in regenerative medicine have led to the applicability of stem cell technology in various diseases. Stem cells that have self-renewable abilities may differentiate into several cell types to provide therapeutic potential. Among different stem cells, adult stem cells are considered as the safest with remarkable potential for therapeutic application. In this review, we provide current available evidence regarding the application of adult stem cells in medicine, especially in the field of obstetrics and gynecology. Objective This scoping review aims to map and describe the current research on adult stem cell application in obstetrics and gynecology. Methods We performed a systematic search on PubMed, Google Scholar, and Cochrane Library in August 2024 to identify research articles involving adult stem cells in the field of obstetrics and gynecology. We used the Deduplicate website to filter articles based on keywords that met our inclusion and exclusion criteria. The results were presented based on recommendations from the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews. Results We found 42 articles that met the inclusion criteria. Some studies were clinical studies, whereas the majority were preclinical studies. We categorized the articles into clinical and preclinical studies to understand their applicability in human subjects. Conclusions Adult stem cell therapy is a candidate treatment for several pathologies in obstetrics and gynecology. The promising results of adult stem cell therapy, especially in degenerative gynecologic diseases, may lead to further application of the technology in the near future.
Collapse
Affiliation(s)
- Andi Kurniadi
- Department of Obstetrics and Gynecology, Universitas Padjadjaran – RSUP Hasan Sadikin, Jl. Pasteur No. 38, Bandung, Indonesia
| | - Muhammad Ary Zucha
- Department of Obstetrics and Gynecology, Universitas Padjadjaran – RSUP Hasan Sadikin, Jl. Pasteur No. 38, Bandung, Indonesia
- Department of Obstetrics and Gynecology, Universitas Gadjah Mada – RSUP Sardjito, Jl. Kesehatan No. 1, Sleman, Indonesia
| | - Ardhanu Kusumanto
- Department of Obstetrics and Gynecology, Universitas Gadjah Mada – RSUP Sardjito, Jl. Kesehatan No. 1, Sleman, Indonesia
| | - Siti Salima
- Department of Obstetrics and Gynecology, Universitas Padjadjaran – RSUP Hasan Sadikin, Jl. Pasteur No. 38, Bandung, Indonesia
| | - Ali Budi Harsono
- Department of Obstetrics and Gynecology, Universitas Padjadjaran – RSUP Hasan Sadikin, Jl. Pasteur No. 38, Bandung, Indonesia
| |
Collapse
|
15
|
Meng X, Shi M, Qian J, Luo Y, Cui L, Li D, Wang S. Tacrolimus (FK506) promotes placentation and maternal-fetal tolerance through modulating FASN-CEACAM1 pathway. Front Immunol 2025; 16:1522346. [PMID: 40046057 PMCID: PMC11879939 DOI: 10.3389/fimmu.2025.1522346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
The establishment of placentation and maternal-fetal tolerance are important determinants of a successful pregnancy. Tacrolimus, also known as FK506, is a calcineurin inhibitor that has often been used for pregnant women after solid organ transplantation. Previous therapeutic interventions have shown the benefits of using the immuno-suppressive agent FK506 in improving clinical pregnancy and live birth rates and reducing the risk of spontaneous miscarriage. However, the mechanism(s) by which FK506 is involved in these processes have not been fully elucidated. To further characterize its function in early pregnancy, we explored the effect of FK506 on the human-derived first trimester extravillous trophoblast cells (HTR8/SVneo cells) and found that FK506 promoted invasion, tube formation and proliferation, but inhibited apoptosis of HTR8/SVneo cells. Based on the integrated metabolomics and transcriptomics analyses, the present study provided the cellular and molecular cues evidently showing that FK506 had positive effects on the placentation and maternal-fetal tolerance through modulating FASN-CEACAM1 pathway. The spontaneous-abortion-prone model gave further evidence that FK506 exerted a protective effect on pregnancy by regulating the FASN-CEACAM1 axis. These findings might provide a new fundamental mechanism and promising potential of low-dose FK506 in preventing pregnancy loss.
Collapse
Affiliation(s)
- Xinhang Meng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Minfeng Shi
- Reproductive Medicine Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jinfeng Qian
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yujie Luo
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Liyuan Cui
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Songcun Wang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
16
|
Valencia J, Yáñez RM, Muntión S, Fernández-García M, Martín-Rufino JD, Zapata AG, Bueren JA, Vicente Á, Sánchez-Guijo F. Improving the therapeutic profile of MSCs: Cytokine priming reduces donor-dependent heterogeneity and enhances their immunomodulatory capacity. Front Immunol 2025; 16:1473788. [PMID: 40034706 PMCID: PMC11872697 DOI: 10.3389/fimmu.2025.1473788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction MSCs exhibit regenerative, anti-inflammatory and immunomodulatory properties due to the large amount of cytokines, chemokines and growth factors they secrete. MSCs have been extensively evaluated in clinical trials, however, in some cases their therapeutic effects are variable. Therefore, strategies to improve their therapeutic potential, such as preconditioning with proinflammatory factors, have been proposed. Several priming approaches have provided non-conclusive results, and the duration of priming effects on MSC properties or their response to a second inflammatory stimulus have not been fully addressed. Methods We have investigated the impact of triple cytokine priming in MSCs on their characterization and viability, their transcriptomic profile, the functionality of innate and acquired immune cells, as well as the maintenance of the response to priming over time, their subsequent responsiveness to a second inflammatory stimulus. Results Priming MSCs with proinflammatory cytokines (CK-MSCs) do not modify the differentiation capacity of MSCs, nor their immunophenotype and viability. Moreover, cytokine priming enhances the anti-inflammatory and immunomodulatory properties of MSCs against NK and dendritic cells, while maintaining the same T cell immunomodulatory capacity as unstimulated MSCs. Thus, they decrease T-lymphocytes and NK cell proliferation, inhibit the differentiation and allostimulatory capacity of dendritic cells and promote the differentiation of monocytes with an immunosuppressive profile. In addition, we have shown for the first time that proinflammatory priming reduces the variability between different donors and MSC origins. Finally, the effect on CK-MSC is maintained over time and even after a secondary inflammatory stimulus. Conclusions Cytokine-priming improves the therapeutic potential of MSCs and reduces inter-donor variability.
Collapse
Affiliation(s)
- Jaris Valencia
- Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Heath Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa M. Yáñez
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Sandra Muntión
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, University of Salamanca and Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, Salamanca, Spain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, Salamanca, Spain
| | - María Fernández-García
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Jorge Diego Martín-Rufino
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Agustín G. Zapata
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
- Heath Research Institute Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Juan A. Bueren
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Heath Research Institute-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Ángeles Vicente
- Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Heath Research Institute Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Fermín Sánchez-Guijo
- RICORS TERAV, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, University of Salamanca and Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, Salamanca, Spain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y León, Salamanca, Spain
| |
Collapse
|
17
|
Abu-El-Rub E, Khaswaneh RR, Almahasneh FA, Almazari R, Alzu'bi A. Adipose Tissue and Bone Marrow-Derived Mesenchymal Stem Cells are not Really the Same: Investigating the Differences in Their Immunomodulatory, Migratory, and Adhesive Profile. Biochem Genet 2025; 63:378-392. [PMID: 38441812 DOI: 10.1007/s10528-024-10724-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/28/2024] [Indexed: 09/03/2024]
Abstract
Mesenchymal stem cells (MSCs) are the most widely used stem cells in regenerative medicine. They can be isolated from multiple sources, most commonly bone marrow and adipose tissue. MSCs derived from different sources show similar molecular and biological characteristics, but there is ongoing debate regarding the best source of MSCs and the potential biological differences between MSCs from different origins. Bone marrow derived-MSCs (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) share many molecular and immunomodulatory properties. In this study, we compared the levels of major immunomodulatory, adhesive, and migratory factors in human BM-MSCs and AD-MSCs under normal conditions, which will help determine the suitability and specificity of each type for certain therapeutic applications. WST1 assay and fluorescent assay SUC-LLVY-AMC were used to measure MSC proliferation and 26S proteasome activity, respectively. Western blotting, ELISA Assays, and bright field live imaging were also used. AD-MSCs and BM-MSCs exhibited similar morphology and proliferation rate. A significantly higher 26S proteasome activity was detected in AD-MSCs than in BM-MSCs. Levels of ICAM-1, integrin α5 and integrin α6 were significantly higher in AD-MSCs compared to BM-MSCs, while no significant difference in CXCR4 levels was observed. Expression of IDO and factor H was significantly higher in AD-MSCs, while CTLA-4 and IL-10 levels were higher in BM-MSCs. This indicates that AD-MSCs and BM-MSCs have different immunomodulatory and adhesion profiles. MSCs isolated from different sources may show differences in their biological and immunomodulatory properties, suggesting a potential suitability of certain MSCs type for specific conditions. Also, combination of different MSCs types could help optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ramada R Khaswaneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan.
| | - Fatimah A Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Rawan Almazari
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ayman Alzu'bi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| |
Collapse
|
18
|
Baran Z, Çetinkaya M, Baran Y. Mesenchymal Stem Cells in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:149-177. [PMID: 39470980 DOI: 10.1007/5584_2024_824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
The mesenchymal stem/stromal cells (MSCs) are multipotent cells that were initially discovered in the bone marrow in the late 1960s but have so far been discovered in almost all tissues of the body. The multipotent property of MSCs enables them to differentiate into various cell types and lineages, such as adipocytes, chondrocytes, and osteocytes. The immunomodulation capacity and tumor-targeting features of MSCs made their use crucial for cell-based therapies in cancer treatment, yet limited advancement could be observed in translational medicine prospects due to the need for more information regarding the controversial roles of MSCs in crosstalk tumors. In this review, we discuss the therapeutic potential of MSCs, the controversial roles played by MSCs in cancer progression, and the anticancer therapeutic strategies that are in association with MSCs. Finally, the clinical trials designed for the direct use of MSCs for cancer therapy or for their use in decreasing the side effects of other cancer therapies are also mentioned in this review to evaluate the current status of MSC-based cancer therapies.
Collapse
Affiliation(s)
- Züleyha Baran
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Anadolu University, Eskişehir, Turkey
| | - Melisa Çetinkaya
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Yusuf Baran
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey.
| |
Collapse
|
19
|
Moratin H, Mache I, Goncalves M, Ehret Kasemo T, Stöth M, Meyer TJ, Hackenberg S, Scherzad A, Herrmann M. Preconditioning with Wound Fluid Enhances Immunosuppressive Properties of Mesenchymal Stromal Cells In Vitro. Int J Mol Sci 2024; 26:293. [PMID: 39796154 PMCID: PMC11719632 DOI: 10.3390/ijms26010293] [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: 11/27/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Immunosuppression is one key feature of mesenchymal stromal cells (MSCs) that has high expectations for therapeutic use. The influence of pro-inflammatory stimuli can modify the characteristics of MSCs and enhance immunosuppressive properties. The local postoperative environment contains cytokines, MSCs, and immune cells in high quantities, and their mutual influence is still unclear. Knowledge of in vivo processes is pivotal for potential therapeutic applications, and therefore, the aim of this study was to investigate the influence of wound fluid (WF) on the immunomodulatory potential of MSCs. CD4+ cells were co-cultured with native or WF-preconditioned MSCs for 5 days. CFSE staining revealed significant suppression of T cell proliferation after co-culture that was even more distinct in co-culture with WF-MSCs. The concentration of IDO-1, TGF-β1 and IFN-γ was higher while TNF-α was reduced in co-culture supernatants, indicating a transition to an anti-inflammatory milieu. In summary, the results provide evidence that the influence of WF alters the immunomodulatory potential of MSCs. These findings should serve as the basis for further investigations with a focus on T cell subpopulations.
Collapse
Affiliation(s)
- Helena Moratin
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Isabel Mache
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Miguel Goncalves
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Totta Ehret Kasemo
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Manuel Stöth
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Till Jasper Meyer
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Stephan Hackenberg
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Agmal Scherzad
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Würzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | - Marietta Herrmann
- IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, D-97070 Wuerzburg, Germany
- Department of Orthopedics, Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, D-97074 Wuerzburg, Germany
| |
Collapse
|
20
|
Chen B, Chen Z, He M, Zhang L, Yang L, Wei L. Recent advances in the role of mesenchymal stem cells as modulators in autoinflammatory diseases. Front Immunol 2024; 15:1525380. [PMID: 39759531 PMCID: PMC11695405 DOI: 10.3389/fimmu.2024.1525380] [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: 11/09/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Mesenchymal stem cells (MSCs), recognized for their self-renewal and multi-lineage differentiation capabilities, have garnered considerable wide attention since their discovery in bone marrow. Recent studies have underscored the potential of MSCs in immune regulation, particularly in the context of autoimmune diseases, which arise from immune system imbalances and necessitate long-term treatment. Traditional immunosuppressive drugs, while effective, can lead to drug tolerance and adverse effects, including a heightened risk of infections and malignancies. Consequently, adjuvant therapy incorporating MSCs has emerged as a promising new treatment strategy, leveraging their immunomodulatory properties. This paper reviews the immunomodulatory mechanisms of MSCs and their application in autoimmune diseases, highlighting their potential to regulate immune responses and reduce inflammation. The immunomodulatory mechanisms of MSCs are primarily mediated through direct cell contact and paracrine activity with immune cells. This review lays the groundwork for the broader clinical application of MSCs in the future and underscores their significant scientific value and application prospects. Further research is expected to enhance the efficacy and safety of MSCs-based treatments for autoimmune diseases.
Collapse
Affiliation(s)
- Baiyu Chen
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Zhilei Chen
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Mengfei He
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lingling Wei
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| |
Collapse
|
21
|
Liu Y, Zhou Y, Hao B, Wu Z, Gao M, Liu L, Xia Q, Zheng K, Yang S, Tang Y, Gong M, Feng C, Diao H, Tan Y, Zheng H. Inhibition of ferroptosis attenuate lipopolysaccharide-induced early pregnancy loss by protecting against decidual damage of stromal cells. Biochem Biophys Res Commun 2024; 736:150904. [PMID: 39476755 DOI: 10.1016/j.bbrc.2024.150904] [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: 08/26/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/10/2024]
Abstract
Endometrial decidualization is critical for successful embryo implantation. Dysregulation of the immune microenvironment can disrupt normal decidualization processes, potentially resulting in early pregnancy loss. Ferroptosis, a form of cell death dependent on iron and lipid hydroperoxides, is closely associated with inflammation. In this study, we developed an inflammatory early pregnancy loss model to elucidate the mechanisms of decidual damage induced by lipopolysaccharide (LPS) and to assess whether ferroptosis contributes to LPS-induced early pregnancy loss. Through in vivo experiments, we observed that embryo implantation was significantly inhibited and endometrial decidualization was impaired during LPS-induced early pregnancy loss. LPS exposure resulted in abnormal mitochondrial morphology, reduced antioxidant capacity, accumulation of reactive oxygen species (ROS) and disruptions in iron metabolism during decidualization in mouse endometrial stromal cells (mESCs). The administration of ferroptosis inhibitors, specifically ferrostatin-1 (Fer-1) and deferoxamine (DFO), effectively reversed embryo loss and mitigated the decidual damage associated with LPS-induced early pregnancy loss. Fer-1 and DFO exhibited resistance to ferroptosis during decidualization by modulating the antioxidant system and iron metabolism in mESCs, respectively. Our findings indicate that the inhibition of ferroptosis can confer protective effects against decidual damage during LPS-induced early pregnancy loss in mice.
Collapse
Affiliation(s)
- Yanmei Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Yaping Zhou
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Binhe Hao
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Zining Wu
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Min Gao
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Ling Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Qiang Xia
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Kainan Zheng
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Shuang Yang
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Yaoting Tang
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Ming Gong
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Cun Feng
- School of Public Health, Hubei University of Medicine, Shiyan, 442000, China
| | - Honglu Diao
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China
| | - Yan Tan
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China; Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China.
| | - Hongtao Zheng
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China; Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
22
|
Fu L, Zheng X, Luo J, Zhang Y, Gao X, Jin L, Liu W, Zhang C, Gao D, Xu B, Jiang Q, Chou S, Luo L. Machine learning accelerates the discovery of epitope-based dual-bioactive peptides against skin infections. Int J Antimicrob Agents 2024; 64:107371. [PMID: 39486466 DOI: 10.1016/j.ijantimicag.2024.107371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/03/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVES Skin injuries and infections are an inevitable part of daily human life, particularly with chronic wounds, becoming an increasing socioeconomic burden. In treating skin infections and promoting wound healing, bioactive peptides may hold significant potential, particularly those possessing antimicrobial and anti-inflammatory properties. However, obtaining these peptides solely through traditional wet laboratory experiments is costly and time-consuming, and peptides identified by current computer-assisted predictive models largely lack validation of their effects via wet laboratory experiments. Consequently, this study aimed to integrate computer-assisted methods and traditional wet laboratory experiments to identify anti-inflammatory and antimicrobial peptides. METHODS We developed a computer-assisted mining pipeline to screen potential peptides from the epitopes of the major histocompatibility complex class II. RESULTS The peptide AIMP1 was identified, with the ability to physically damage Escherichia coli by increasing bacterial cell membrane permeability, and with the ability to inhibit inflammation by binding to endotoxin-lipopolysaccharide. Additionally, in an LPS-induced inflammation animal model, AIMP1 slightly increased levels of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), and in a skin wound infection animal model, AIMP1 effectively accelerated healing, reduced levels of these pro-inflammatory cytokines, and showed no acute hepatotoxicity or nephrotoxicity. CONCLUSIONS In conclusion, this study not only developed a computer-assisted mining pipeline for identifying anti-inflammatory and antimicrobial peptides but also successfully pinpointed the peptide AIMP1, demonstrating its therapeutic potential for skin injury treatment.
Collapse
Affiliation(s)
- Le Fu
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Xu Zheng
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Jiawen Luo
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Yiyu Zhang
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Xue Gao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, PR China
| | - Li Jin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, PR China
| | - Wenting Liu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, PR China
| | - Chaoqun Zhang
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Dongyu Gao
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Bocheng Xu
- Hangzhou Shenji Technology Co. Ltd, Hangzhou, PR China
| | - Qingru Jiang
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China.
| | - Shuli Chou
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Liang Luo
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China.
| |
Collapse
|
23
|
Gao F, Li X, Wang H, Xu Z, Qian W, Bai G. Single-cell profiling of the peripheral blood immune landscape during mid- and late-stage pregnancy. Physiol Genomics 2024; 56:855-868. [PMID: 39555960 DOI: 10.1152/physiolgenomics.00041.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 11/19/2024] Open
Abstract
We aimed to determine the peripheral blood mononuclear cell (PBMC) immune profiles of mid- and late-stage pregnant women to establish a foundation for studying pregnancy-related diseases. Peripheral blood samples were collected from three women each during mid- and late-stage pregnancy, and PBMCs were extracted for single-cell RNA sequencing (scRNA-seq). Peripheral blood samples were also collected for flow cytometry analysis to validate the analytical results. HOPX+ CD4+ T cells, ZNF683+CD8+ T cells, and KLRB1+CD8+ T cells significantly differed in quantitative ratio and gene transcript level between women at mid- and late-stage pregnancy. In late pregnancy, cell-to-cell communication was enhanced and effector CD8+ T cells highly expressed infection-related pathways. A rare T cell subtype, "XIST+ T cells," exhibited high XIST expression, a gene that may be involved in the regulation of immune-related gene transcription and translation, and insulin signaling pathway, during pregnancy. Monocytes exhibited significant proinflammatory and metabolic properties in mid- and late-stage pregnancy, respectively. Natural killer (NK) cells were mainly involved in T- and B-cell-mediated signaling pathways, and in T cell differentiation, in mid-pregnancy. Enhanced innate immunity of NK cells was observed. Moreover, NK cells expressed genes associated with diabetes-related pathways in late-stage pregnancy. To conclude, we present detailed changes in the immune response occurring in pregnant women from mid- to late-stage gestation, revealing significant differences in PBMC subtypes and molecular properties. These findings provide insights into the physiopathological mechanisms of chronic hepatitis B infection, systemic lupus erythematosus, and gestational diabetes mellitus underlying systemic immune responses during mid- and late-stage pregnancy.NEW & NOTEWORTHY There are significant differences in three subtypes of memory/effector T cells (HOPX+ CD4+ T cells, ZNF683+CD8+ T cells, and KLRB1+CD8+ T cells) between mid- and late pregnancy. In late pregnancy, intercellular interaction was enhanced and effector CD8+ T cells highly expressed infection-related pathways. A rare T cell subtype, "XIST+ T cells," may be involved in the regulation of immune-related gene transcription and translation with a strong female bias.
Collapse
Affiliation(s)
- Fan Gao
- Gene Joint Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xia Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Hongyan Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zhen Xu
- Gene Joint Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Wenjun Qian
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Guiqin Bai
- Gene Joint Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
24
|
Li Y, Jin M, Guo D, Shen S, Lu K, Pan R, Sun L, Zhang H, Shao J, Pan G. Unveiling the immunogenicity of allogeneic mesenchymal stromal cells: Challenges and strategies for enhanced therapeutic efficacy. Biomed Pharmacother 2024; 180:117537. [PMID: 39405918 DOI: 10.1016/j.biopha.2024.117537] [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: 07/27/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) exhibit significant potential in the context of cell therapy because of their capacity to perform a range of interconnected functions in damaged tissues, including immune modulation, hematopoietic support, and tissue regeneration. MSCs are hypoimmunogenic because of their diminished expression of major histocompatibility molecules, absence of costimulatory molecules, and presence of coinhibitory molecules. While autologous MSCs reduce the risk of rejection and infection, variability in cell numbers and proliferation limits their potential applications. Conversely, allogeneic MSCs (allo-MSCs) possess broad clinical applications unconstrained by donor physiology. Nonetheless, preclinical and clinical investigations highlight that transplanted allo-MSCs are subject to immune attack from recipients. These cells exhibit anti-inflammatory and proinflammatory phenotypes contingent on the microenvironment. Notably, the proinflammatory phenotype features enhanced immunogenicity and diminished immunosuppression, potentially triggering allogeneic immune reactions that impede long-term clinical efficacy. Consequently, preserving the low immunogenicity of allo-MSCs in vivo and mitigating immune rejection in diverse microenvironments represent crucial challenges for the widespread clinical application of MSCs. In this review, we elucidate the immune regulation of allo-MSCs, specifically focusing on two distinct subgroups, MSC1 and MSC2, that exhibit varying polarization states and immunogenicity. We discuss the factors and underlying mechanisms that induce MSC immunogenicity and polarization, highlighting the crucial role of major histocompatibility complex class I/II molecules in rejection post-transplantation. Additionally, we summarize the immunogenic regulatory targets and applications of allo-MSCs and outline strategies to address challenges in this promising field, aiming to enhance allo-MSC therapeutic efficacy for patients.
Collapse
Affiliation(s)
- Yuanhui Li
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Mengting Jin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Dongyang Guo
- Hangzhou City University, School of Medicine, 50 Huzhou Street, Hangzhou, China
| | - Shuang Shen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Kaining Lu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruolang Pan
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Li Sun
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Hongchen Zhang
- Department of Gatroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, No. 261 HuanSha Road, Hangzhou, China.
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Gang Pan
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Xu G, Tian C, Li Y, Fang L, Wang J, Jing Z, Li S, Chen P. Inhibition of BCAT1 expression improves recurrent miscarriage by regulating cellular dysfunction and inflammation of trophoblasts. Cell Tissue Res 2024; 398:111-121. [PMID: 39356334 DOI: 10.1007/s00441-024-03921-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Sustained or chronic inflammation in the placenta can result in placental insufficiency, leading to adverse reproductive outcomes such as pregnancy loss. Branched-chain amino acid transaminase 1 (BCAT1) expresses in the placenta and is involved in the pathological inflammatory response, but its role in recurrent miscarriage (RM) has not been fully investigated. In the present study, we delved into the effects of BCAT1 on trophoblast inflammation induced by lipopolysaccharide (LPS) and a mouse model of pregnancy loss induced by LPS. In vitro, after the HTR-8/SVneo cells were treated with LPS and BCATc inhibitor 2 (a selective BCAT inhibitor), the cell apoptosis was verified by TUNEL assay, and the activity of caspase-3 and caspase-9 was detected. Real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence (IF) were used to determine the expression of inflammatory cytokines (TNF-α, IL-6, and IL-1β) and inflammasomes (NLRP3 and ASC) in LPS-treated trophoblast cells. Western blot analysis was performed to verify the expression of phospho-IκBα (p-IκBα) in cells and NF-κB p65 in the nuclei. IF staining was used to detect the nuclear translocation of NF-κB p65. The DNA binding activity of NF-κB was detected by an electrophoretic mobility shift assay (EMSA). The results demonstrated that inhibition of BCAT1 reduced trophoblast apoptosis, suppressed the release of proinflammatory cytokines, and prevented NLRP3 inflammasome activation in response to LPS. Additionally, BCAT1 inhibition blocked the activation of the NF-κB pathway in trophoblasts. This study highlights the potential therapeutic role of targeting BCAT1 in preventing adverse reproductive outcomes associated with chronic placental inflammation.
Collapse
Affiliation(s)
- Guangli Xu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| | - Chao Tian
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Yanru Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Lei Fang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Jing Wang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Zhuqing Jing
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Simeng Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Ping Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| |
Collapse
|
26
|
Li MY, Wu Y, Tang HL, Wang Y, Li B, He YY, Yan GJ, Yang ZM. Embryo-Derived Cathepsin B Promotes Implantation and Decidualization by Activating Pyroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402299. [PMID: 39316370 DOI: 10.1002/advs.202402299] [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: 03/03/2024] [Revised: 08/17/2024] [Indexed: 09/25/2024]
Abstract
Embryo implantation and decidualization are crucial for a successful pregnancy. How the inflammatory response is regulated during these processes is undefined. Pyroptosis is an inflammatory form of cell death mediated by gasdermin D (GSDMD). Through in vivo, cultured epithelial cells and organoids, it is shown that pyroptosis occurs in epithelial cells at the implantation site. Compared with those on day 4 of pseudopregnancy and delayed implantation, pyroptosis-related protein levels are significantly increased on day 4 of pregnancy and activated implantation, suggesting that blastocysts are involved in regulating pyroptosis. Blastocyst-derived cathepsin B (CTSB) is stimulated by preimplantation estradiol-17β and induces pyroptosis in epithelial cells. Pyroptosis-induced IL-18 secretion from epithelial cells activates a disintegrin and metalloprotease 12 (ADAM12) to process the epiregulin precursor into mature epiregulin. Epiregulin (EREG) enhances in vitro decidualization in mice. Pyroptosis-related proteins are detected in the mid-secretory human endometrium and are elevated in the recurrent implantation failure endometrium. Lipopolysaccharide treatment in pregnant mice causes implantation failure and increases pyroptosis-related protein levels. Therefore, the data suggest that modest pyroptosis is beneficial for embryo implantation and decidualization. Excessive pyroptosis can be harmful and lead to pregnancy failure.
Collapse
Affiliation(s)
- Meng-Yuan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hao-Lan Tang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ying Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bo Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu-Ying He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Gui-Jun Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| | - Zeng-Ming Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
| |
Collapse
|
27
|
Motlagh Asghari K, Novinbahador T, Mehdizadeh A, Zolfaghari M, Yousefi M. Revolutionized attitude toward recurrent pregnancy loss and recurrent implantation failure based on precision regenerative medicine. Heliyon 2024; 10:e39584. [PMID: 39498089 PMCID: PMC11532865 DOI: 10.1016/j.heliyon.2024.e39584] [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: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Traditional treatment strategies for recurrent pregnancy loss (RPL) and recurrent implantation failure (RIF) often result in limited success, placing significant emotional and financial burdens on couples. However, novel approaches such as diagnostic gene profiling, cell therapy, stem cell-derived exosome therapy, and pharmacogenomics offer promising, personalized treatments. Combining traditional treatments with precision and regenerative medicine may enhance the efficacy of these approaches and improve pregnancy outcomes. This review explores how integrating these strategies can potentially transform the lives of couples experiencing repeated pregnancy loss or implantation failure, providing hope for improved treatment success. Precision and regenerative medicine represent a new frontier for managing RPL and RIF, offering promising solutions.
Collapse
Affiliation(s)
| | - Tannaz Novinbahador
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
29
|
Liu L, Fandiño J, McCarthy SD, Masterson CH, Sallent I, Du S, Warren A, Laffey JG, O’Toole D. The Effects of the Pneumonia Lung Microenvironment on MSC Function. Cells 2024; 13:1581. [PMID: 39329762 PMCID: PMC11430541 DOI: 10.3390/cells13181581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Despite promise in preclinical models of acute respiratory distress syndrome (ARDS), mesenchymal stem cells (MSC) have failed to translate to therapeutic benefit in clinical trials. The MSC is a live cell medicine and interacts with the patient's disease state. Here, we explored this interaction, seeking to devise strategies to enhance MSC therapeutic function. METHODS Human bone-marrow-derived MSCs were exposed to lung homogenate from healthy and E. coli-induced ARDS rat models. Apoptosis and functional assays of the MSCs were performed. RESULTS The ARDS model showed reduced arterial oxygenation, decreased lung compliance and an inflammatory microenvironment compared to controls. MSCs underwent more apoptosis after stimulation by lung homogenate from controls compared to E. coli, which may explain why MSCs persist longer in ARDS subjects after administration. Changes in expression of cell surface markers and cytokines were associated with lung homogenate from different groups. The anti-microbial effects of MSCs did not change with the stimulation. Moreover, the conditioned media from lung-homogenate-stimulated MSCs inhibited T-cell proliferation. CONCLUSIONS These findings suggest that the ARDS microenvironment plays an important role in the MSC's therapeutic mechanism of action, and changes can inform strategies to modulate MSC-based cell therapy for ARDS.
Collapse
Affiliation(s)
- Lanzhi Liu
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Physiology, University of Galway, H91 W5P7 Galway, Ireland
| | - Juan Fandiño
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Sean D. McCarthy
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Claire H. Masterson
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Ignacio Sallent
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Shanshan Du
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- School of Medicine, University of Galway, H91 W5P7 Galway, Ireland
| | - Abigail Warren
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Anaesthesia, University of Galway, H91 V4AY Galway, Ireland
| | - John G. Laffey
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Anaesthesia, University of Galway, H91 V4AY Galway, Ireland
- Anaesthesia and Critical Care, Galway University Hospital, H91 V4AY Galway, Ireland
| | - Daniel O’Toole
- CÚRAM Institute for Medical Devices, University of Galway, H91 W2TY Galway, Ireland; (L.L.); (J.F.); (S.D.M.); (C.H.M.); (I.S.); (S.D.); (A.W.); (J.G.L.)
- Discipline of Physiology, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
30
|
Di Santo C, Siniscalchi A, La Russa D, Tonin P, Bagetta G, Amantea D. Brain Ischemic Tolerance Triggered by Preconditioning Involves Modulation of Tumor Necrosis Factor-α-Stimulated Gene 6 (TSG-6) in Mice Subjected to Transient Middle Cerebral Artery Occlusion. Curr Issues Mol Biol 2024; 46:9970-9983. [PMID: 39329947 PMCID: PMC11430743 DOI: 10.3390/cimb46090595] [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: 08/18/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Ischemic preconditioning (PC) induced by a sub-lethal cerebral insult triggers brain tolerance against a subsequent severe injury through diverse mechanisms, including the modulation of the immune system. Tumor necrosis factor (TNF)-α-stimulated gene 6 (TSG-6), a hyaluronate (HA)-binding protein, has recently been involved in the regulation of the neuroimmune response following ischemic stroke. Thus, we aimed at assessing whether the neuroprotective effects of ischemic PC involve the modulation of TSG-6 in a murine model of transient middle cerebral artery occlusion (MCAo). The expression of TSG-6 was significantly elevated in the ischemic cortex of mice subjected to 1 h MCAo followed by 24 h reperfusion, while this effect was further potentiated (p < 0.05 vs. MCAo) by pre-exposure to ischemic PC (i.e., 15 min MCAo) 72 h before. By immunofluorescence analysis, we detected TSG-6 expression mainly in astrocytes and myeloid cells populating the lesioned cerebral cortex, with a more intense signal in tissue from mice pre-exposed to ischemic PC. By contrast, levels of TSG-6 were reduced after 24 h of reperfusion in plasma (p < 0.05 vs. SHAM), but were dramatically elevated when severe ischemia (1 h MCAo) was preceded by ischemic PC (p < 0.001 vs. MCAo) that also resulted in significant neuroprotection. In conclusion, our data demonstrate that neuroprotection exerted by ischemic PC is associated with the elevation of TSG-6 protein levels both in the brain and in plasma, further underscoring the beneficial effects of this endogenous modulator of the immune system.
Collapse
Affiliation(s)
- Chiara Di Santo
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (C.D.S.)
| | - Antonio Siniscalchi
- Department of Neurology and Stroke Unit, Annunziata Hospital, 87100 Cosenza, Italy
| | - Daniele La Russa
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (C.D.S.)
| | - Paolo Tonin
- Regional Center for Serious Brain Injuries, S. Anna Institute, 88900 Crotone, Italy
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (C.D.S.)
| | - Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (C.D.S.)
| |
Collapse
|
31
|
Xiao Y, Zeng F, Sun J. The improvement of inflammatory infiltration and pregnancy outcome in mice with recurrent spontaneous abortion by human amniotic mesenchymal stem cells. Biol Reprod 2024; 111:351-360. [PMID: 38718142 PMCID: PMC11327314 DOI: 10.1093/biolre/ioae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 08/17/2024] Open
Abstract
Recurrent spontaneous abortion is thought to be mostly triggered by immune-related causes. Mesenchymal stem cells, which exhibit the traits of multi-directional differentiation capacity and low immunogenicity, have recently been recommended as a viable treatment for spontaneous abortion-prone mice to increase the success of pregnancy. Amniotic membrane tissue is a byproduct of pregnancy and delivery that has a wide range of potential uses due to its easy access to raw materials and little ethical constraints. To construct an abortion-prone mouse model for this investigation, CBA/J female mice were coupled with male DBA/2 mice, while CBA/J female mice were paired with male BALB/c mice as a control. The identical volume of human amniotic mesenchymal stem cells or phosphate buffer was injected intraperitoneally on the 4.5th day of pregnancy. CBA/J female mice were sacrificed by cervical dislocation on the 13.5th day of pregnancy, the embryo absorption rate was calculated, and the uterus, decidua tissues and placenta were gathered for examination. Through detection, it was discovered that human amniotic mesenchymal stem cells significantly increased the expression of interleukin 10 and transforming growth factor beta, while they significantly decreased the expression of interleukin 1 beta and interleukin 6, improved vascular formation and angiogenesis, and minimized the embryo absorption rate and inflammatory cell infiltration in the recurrent spontaneous abortion + human amniotic mesenchymal stem cells group. In any case, human amniotic mesenchymal stem cells regulate inflammatory factors and cell balance at the maternal-fetal interface, which result in a reduction in the rate of embryo absorption and inflammatory infiltration and provide an innovative perspective to the clinical therapy of recurrent spontaneous abortion.
Collapse
Affiliation(s)
- Yi Xiao
- Graduate School of China Medical University, Shenyang 110000, China
| | - Fanyu Zeng
- School of Public Health, Fudan University, Shanghai 200000, China
| | - Jingli Sun
- Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang 110000, China
| |
Collapse
|
32
|
Yu X, Li L, Ning A, Wang H, Guan C, Ma X, Xia H. Primary cilia abnormalities participate in the occurrence of spontaneous abortion through TGF-β/SMAD2/3 signaling pathway. J Cell Physiol 2024; 239:e31292. [PMID: 38704705 DOI: 10.1002/jcp.31292] [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: 01/08/2024] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Spontaneous abortion is the most common complication in early pregnancy, the exact etiology of most cases cannot be determined. Emerging studies suggest that mutations in ciliary genes may be associated with progression of pregnancy loss. However, the involvement of primary cilia on spontaneous abortion and the underlying molecular mechanisms remains poorly understood. We observed the number and length of primary cilia were significantly decreased in decidua of spontaneous abortion in human and lipopolysaccharide (LPS)-induced abortion mice model, accompanied with increased expression of proinflammatory cytokines interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. The length of primary cilia in human endometrial stromal cell (hESC) was significantly shortened after TNF-α treatment. Knocking down intraflagellar transport 88 (IFT88), involved in cilia formation and maintenance, promoted the expression of TNF-α. There was a reverse regulatory relationship between cilia shortening and TNF-α expression. Further research found that shortened cilia impair decidualization in hESC through transforming growth factor (TGF)-β/SMAD2/3 signaling. Primary cilia were impaired in decidua tissue of spontaneous abortion, which might be mainly caused by inflammatory injury. Primary cilia abnormalities resulted in dysregulation of TGF-β/SMAD2/3 signaling transduction and decidualization impairment, which led to spontaneous abortion.
Collapse
Affiliation(s)
- Xiaoqin Yu
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Linyuan Li
- University of Michigan College of Literature, Science, and the Arts, Ann Arbor, Michigan, USA
| | - Anfeng Ning
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Hu Wang
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Chunyi Guan
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Xu Ma
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Hongfei Xia
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Zhang J, Wu P, Wen Q. Optimization strategies for mesenchymal stem cell-based analgesia therapy: a promising therapy for pain management. Stem Cell Res Ther 2024; 15:211. [PMID: 39020426 PMCID: PMC11256674 DOI: 10.1186/s13287-024-03828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
Pain is a very common and complex medical problem that has a serious impact on individuals' physical and mental health as well as society. Non-steroidal anti-inflammatory drugs and opioids are currently the main drugs used for pain management, but they are not effective in controlling all types of pain, and their long-term use can cause adverse effects that significantly impair patients' quality of life. Mesenchymal stem cells (MSCs) have shown great potential in pain treatment. However, limitations such as the low proliferation rate of MSCs in vitro and low survival rate in vivo restrict their analgesic efficacy and clinical translation. In recent years, researchers have explored various innovative approaches to improve the therapeutic effectiveness of MSCs in pain treatment. This article reviews the latest research progress of MSCs in pain treatment, with a focus on methods to enhance the analgesic efficacy of MSCs, including engineering strategies to optimize the in vitro culture environment of MSCs and to improve the in vivo delivery efficiency of MSCs. We also discuss the unresolved issues to be explored in future MSCs and pain research and the challenges faced by the clinical translation of MSC therapy, aiming to promote the optimization and clinical translation of MSC-based analgesia therapy.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Ping Wu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
34
|
Li K, Nie H, Jin R, Wu X. Mesenchymal stem cells-macrophages crosstalk and myeloid malignancy. Front Immunol 2024; 15:1397005. [PMID: 38779660 PMCID: PMC11109455 DOI: 10.3389/fimmu.2024.1397005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
As major components of the tumor microenvironment, both mesenchymal stem cells (MSCs) and macrophages can be remodelled and exhibit different phenotypes and functions during tumor initiation and progression. In recent years, increasing evidence has shown that tumor-associated macrophages (TAMs) play a crucial role in the growth, metastasis, and chemotherapy resistance of hematological malignancies, and are associated with poor prognosis. Consequently, TAMs have emerged as promising therapeutic targets. Notably, MSCs exert a profound influence on modulating immune cell functions such as macrophages and granulocytes, thereby playing a crucial role in shaping the immunosuppressive microenvironment surrounding tumors. However, in hematological malignancies, the cellular and molecular mechanisms underlying the interaction between MSCs and macrophages have not been clearly elucidated. In this review, we provide an overview of the role of TAMs in various common hematological malignancies, and discuss the latest advances in understanding the interaction between MSCs and macrophages in disease progression. Additionally, potential therapeutic approaches targeting this relationship are outlined.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Nie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Jiang X, Li L. Decidual macrophage: a reversible role in immunotolerance between mother and fetus during pregnancy. Arch Gynecol Obstet 2024; 309:1735-1744. [PMID: 38329548 DOI: 10.1007/s00404-023-07364-3] [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: 10/11/2023] [Accepted: 12/17/2023] [Indexed: 02/09/2024]
Abstract
The tolerance of the semi-allogeneic fetus by the maternal immune system is an eternal topic of reproductive immunology for ensuring a satisfactory outcome. The maternal-fetal interface serves as a direct portal for communication between the fetus and the mother. It is composed of placental villi trophoblast cells, decidual immune cells, and stromal cells. Decidual immune cells engage in maintaining the homeostasis of the maternal-fetal interface microenvironment. Furthermore, growing evidence has shown that decidual macrophages play a crucial role in maternal-fetal tolerance during pregnancy. As the second largest cell population among decidual immune cells, decidual macrophages are divided into two subtypes: classically activated macrophages (M1) and alternatively activated macrophages (M2). M2 polarization is critical for placentation and embryonic development. Cytokines, exosomes, and metabolites regulate the polarization of decidual macrophages, and thereby modulate maternal-fetal immunotolerance. Explore the initial relationship between decidual macrophages polarization and maternal-fetal immunotolerance will help diagnose and treat the relevant pregnancy diseases, reverse the undesirable outcomes of mothers and infants.
Collapse
Affiliation(s)
- Xiaotong Jiang
- Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Lei Li
- Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China, No. 324, Jingwu Weiqi Road, Huaiyin District, 250021.
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, China, No. 6699, Qingdao Road, Huaiyin District, 250117.
| |
Collapse
|
36
|
Sun H, Dong J, Fu Z, Lu X, Chen X, Lei H, Xiao X, Chen S, Lu J, Su D, Xiong Y, Fang Z, Mao J, Chen L, Wang X. TSG6-Exo@CS/GP Attenuates Endometrium Fibrosis by Inhibiting Macrophage Activation in a Murine IUA Model. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308921. [PMID: 38588501 DOI: 10.1002/adma.202308921] [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: 09/01/2023] [Revised: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Intrauterine adhesion (IUA) is characterized by the formation of fibrous scar tissue within the uterine cavity, which significantly impacts female reproductive health and even leads to infertility. Unfortunately, severe cases of IUA currently lack effective treatments. This study presents a novel approach that utilizes tumor necrosis factor-(TNF) stimulated gene 6 (TSG6)-modified exosomes (Exos) in conjunction with an injectable thermosensitive hydrogel (CS/GP) to mitigate the occurrence of IUA by reducing endometrium fibrosis in a mouse IUA model. This study demonstrate that TSG6-modified Exos effectively inhibits the activation of inflammatory M1-like macrophages during the initial stages of inflammation and maintains the balance of macrophage phenotypes (M1/M2) during the repair phase. Moreover, TSG6 inhibits the interaction between macrophages and endometrial stromal fibroblasts, thereby preventing the activation of stromal fibroblasts into myofibroblasts. Furthermore, this research indicates that CS/GP facilitates the sustained release of TSG6-modified Exos, leading to a significant reduction in both the manifestations of IUA and the extent of endometrium fibrosis. Collectively, through the successful construction of CS/GP loaded with TSG6-modified Exos, a reduction in the occurrence and progression of IUA is achieved by mitigating endometrium fibrosis. Consequently, this approach holds promise for the treatment of IUA.
Collapse
Affiliation(s)
- Huijun Sun
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jie Dong
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Zhaoyue Fu
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Xueyan Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Xutao Chen
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Hui Lei
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Xifeng Xiao
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Shuqiang Chen
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jie Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Danjie Su
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Yujing Xiong
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Zheng Fang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jiaqin Mao
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Xiaohong Wang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| |
Collapse
|
37
|
Zhang FF, Hao Y, Zhang KX, Yang JJ, Zhao ZQ, Liu HJ, Li JT. Interplay between mesenchymal stem cells and macrophages: Promoting bone tissue repair. World J Stem Cells 2024; 16:375-388. [PMID: 38690513 PMCID: PMC11056637 DOI: 10.4252/wjsc.v16.i4.375] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/14/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
The repair of bone tissue damage is a complex process that is well-orchestrated in time and space, a focus and difficulty in orthopedic treatment. In recent years, the success of mesenchymal stem cells (MSCs)-mediated bone repair in clinical trials of large-area bone defects and bone necrosis has made it a candidate in bone tissue repair engineering and regenerative medicine. MSCs are closely related to macrophages. On one hand, MSCs regulate the immune regulatory function by influencing macrophages proliferation, infiltration, and phenotype polarization, while also affecting the osteoclasts differentiation of macrophages. On the other hand, macrophages activate MSCs and mediate the multilineage differentiation of MSCs by regulating the immune microenvironment. The cross-talk between MSCs and macrophages plays a crucial role in regulating the immune system and in promoting tissue regeneration. Making full use of the relationship between MSCs and macrophages will enhance the efficacy of MSCs therapy in bone tissue repair, and will also provide a reference for further application of MSCs in other diseases.
Collapse
Affiliation(s)
- Fei-Fan Zhang
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
- Graduate School, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Yang Hao
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
- Graduate School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Kuai-Xiang Zhang
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
- Graduate School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jiang-Jia Yang
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
- Graduate School, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Zhi-Qiang Zhao
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
| | - Hong-Jian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Ji-Tian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou 450000, Henan Province, China
- Graduate School, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
- Graduate School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
38
|
Sun J, Zhong H, Kang B, Lum T, Liu D, Liang S, Hao J, Guo R. Roles of PD-L1 in human adipose-derived mesenchymal stem cells under inflammatory microenvironment. J Cell Biochem 2024; 125:e30544. [PMID: 38450777 DOI: 10.1002/jcb.30544] [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: 11/30/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
Mesenchymal stem cells (MSCs) display unique homing and immunosuppression features which make them promising candidates for cell therapy in inflammatory disorders. It is known that C-X-C chemokine receptor type 4 (CXCR4, also known as CD184) is a critical receptor implicated in MSCs migration, and the protein programmed death ligand-1 (PD-L1) is involved in MSC's immunosuppression. However, it remains unclear how the molecular mechanisms regulate PD-L1 expression for migration and immunosuppression of MSCs under the inflammatory microenvironment. In this article, we used the human adipose-derived mesenchymal stem cells (hADMSCs) treated with lipopolysaccharide (LPS) as an in vitro inflammatory model to explore the roles of PD-L1 on the migration and immunosuppression of MSC. Our results demonstrate that in hADMSCs, LPS significantly increased PD-L1 expression, which mediated the migration of the LPS-treated hADMSCs via CXCR4. In addition, we found that the increased PD-L1 expression in the LPS-treated hADMSCs inhibited B cell proliferation and immunoglobulin G secretion through nuclear factor-κB. Our study suggests that the PD-L1 plays critical roles in the homing and immunosuppression of MSCs which are a promising cell therapy to treat inflammatory diseases.
Collapse
Affiliation(s)
- Jinqiu Sun
- Institute of Life Science and Green Development, College of Life Sciences, Hebei University, Baoding, China
| | - Hannah Zhong
- College of Letters and Science, University of California, Los Angeles, California, USA
| | - Bo Kang
- Department of Health Policy and Management, Jonathan and Karin Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Trenton Lum
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Dongxue Liu
- Institute of Life Science and Green Development, College of Life Sciences, Hebei University, Baoding, China
| | - Shengxian Liang
- Institute of Life Science and Green Development, College of Life Sciences, Hebei University, Baoding, China
| | - Jijun Hao
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA
| | - Rui Guo
- Institute of Life Science and Green Development, College of Life Sciences, Hebei University, Baoding, China
| |
Collapse
|
39
|
Yue W, Huiling Z, Yuxin L, Ling W, Feng G, Qicai L. Neu5Gc regulates decidual macrophages leading to abnormal embryo implantation. Genes Immun 2024; 25:149-157. [PMID: 38499667 DOI: 10.1038/s41435-024-00268-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024]
Abstract
Repeated implantation failure (RIF) is one of the most prominent problems in the field of assisted reproduction. Neu5Gc on the surface of decidual macrophages (dMΦ) leads to different activation patterns of dMΦ, which affects embryo implantation and development. Cmah-/- (Neu5Gc-deficient) mice induced to produce anti-Neu5Gc antibodies in vivo were given a special diet rich in Neu5Gc and their fertility was monitored. The long-term diet rich in Neu5Gc induced the decrease of endometrial receptivity of female mice. The pregnancy rate of female mice fed the normal diet was 63.6% (n = 11) and the average number of embryos was 9.571 ± 1.272, while the pregnancy rate of female mice fed the diet rich in Neu5Gc was 36.4% (n = 11) and the average number of embryos in pregnant mice was 5.750 ± 3.304. The intake of Neu5Gc and the production of anti-Neu5Gc antibody led to M1 polarization of endometrial dMΦ and abnormal embryo implantation.
Collapse
Affiliation(s)
- Wu Yue
- Center of Reproductive Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Zhou Huiling
- Center of Reproductive Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Liu Yuxin
- Center of Reproductive Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Wang Ling
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Gao Feng
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
| | - Liu Qicai
- Fujian Provincial Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Medical University, China, Fuzhou.
| |
Collapse
|
40
|
Büyükbayrak EE, Gündoğdu NEÖ, Gürkan N, Kahraman FR, Akalın M, Akkoç T. Immunological effects of human decidual mesenchymal stem cells in spontaneous and recurrent abortions. J Reprod Immunol 2024; 162:104193. [PMID: 38281405 DOI: 10.1016/j.jri.2024.104193] [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: 07/07/2023] [Revised: 09/02/2023] [Accepted: 01/08/2024] [Indexed: 01/30/2024]
Abstract
The aim of this study was to evaluate the immunological activities of human decidual mesenchymal stem cells (MSCs) on proliferation, apoptosis and percentage of regulatory T cells (Treg) in abortions and to investigate whether these activities differ in spontaneous abortions (SA) and recurrent abortions (RA). This prospective cohort study included women who had a first-trimester abortion between 2019 and 2022. Women with uterine anomaly, endocrinological disease, known autoimmune or thrombophilic disease, and fetal chromosomal abnormality in abortion material were excluded. Decidual MSCs isolated from abortion materials were classified as spontaneous abortion-MSCs (SA-MSCs) and recurrent abortion-MSCs (RA-MSCs). Peripheral blood mononuclear cells were isolated from venous blood and co-cultured with SA-MSCs and RA-MSCs. The effects of MSCs on proliferation and apoptosis of lymphocytes, and Tregs levels were compared between SA-MSCs and RA-MSCs groups. Thirty cases (15 SA-MSCs and 15 RA-MSCs) were included in the study. The presence of MSC in co-cultures increased percentage of Treg cells while reducing proliferation and apoptosis compared to those without MSCs (p < 0.0001, p < 0.0001 and p < 0.0001). The increase in percentage of Treg cells and the reduction in apoptosis were significantly lower in the RA-MSCs group compared to the SA-MSCs group (p < 0.0001 and p < 0.001, respectively). Although the proliferation reducing effect of the presence of MSCs was lower in the RA-MSCs group compared to the SA-MSCs group, the difference was not significant (p = 0.07). MSCs contribute to maternal immunotolerance to semi-allogeneic fetus by suppressing proliferation and apoptosis, and increasing percentage of Treg cells. However, the immunoregulatory effects of MSCs are lower in RA compared to SA.
Collapse
Affiliation(s)
- Esra Esim Büyükbayrak
- Department of Perinatology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | | | - Nihan Gürkan
- Department of Obstetrics and Gynaecology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Fatma Rabia Kahraman
- Department of Immunology, Marmara University Pendik Research Hospital, Istanbul, Turkey
| | - Münip Akalın
- Department of Perinatology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey.
| | - Tunç Akkoç
- Department of Immunology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
41
|
Kolahi Azar H, Imanpour A, Rezaee H, Ezzatifar F, Zarei-Behjani Z, Rostami M, Azami M, Behestizadeh N, Rezaei N. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: The homing procedure and their effective parameters. Eur J Haematol 2024; 112:153-173. [PMID: 37254607 DOI: 10.1111/ejh.14014] [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: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Mesenchymal stromal cells (MSCs) and chimeric antigen receptor (CAR)-T cells are two core elements in cell therapy procedures. MSCs have significant immunomodulatory effects that alleviate inflammation in the tissue regeneration process, while administration of specific chemokines and adhesive molecules would primarily facilitate CAR-T cell trafficking into solid tumors. Multiple parameters affect cell homing, including the recipient's age, the number of cell passages, proper cell culture, and the delivery method. In addition, several chemokines are involved in the tumor microenvironment, affecting the homing procedure. This review discusses parameters that improve the efficiency of cell homing and significant cell therapy challenges. Emerging comprehensive mechanistic strategies such as non-systemic and systemic homing that revealed a significant role in cell therapy remodeling were also reviewed. Finally, the primary implications for the development of combination therapies that incorporate both MSCs and CAR-T cells for cancer treatment were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aylar Imanpour
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Rezaee
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ezzatifar
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeinab Zarei-Behjani
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, Advanced School of Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Behestizadeh
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
42
|
Tao X, Wang J, Liu B, Cheng P, Mu D, Du H, Niu B. Plasticity and crosstalk of mesenchymal stem cells and macrophages in immunomodulation in sepsis. Front Immunol 2024; 15:1338744. [PMID: 38352879 PMCID: PMC10861706 DOI: 10.3389/fimmu.2024.1338744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Sepsis is a multisystem disease characterized by dysregulation of the host immune response to infection. Immune response kinetics play a crucial role in the pathogenesis and progression of sepsis. Macrophages, which are known for their heterogeneity and plasticity, actively participate in the immune response during sepsis. These cells are influenced by the ever-changing immune microenvironment and exhibit two-sided immune regulation. Recently, the immunomodulatory function of mesenchymal stem cells (MSCs) in sepsis has garnered significant attention. The immune microenvironment can profoundly impact MSCs, prompting them to exhibit dual immunomodulatory functions akin to a double-edged sword. This discovery holds great importance for understanding sepsis progression and devising effective treatment strategies. Importantly, there is a close interrelationship between macrophages and MSCs, characterized by the fact that during sepsis, these two cell types interact and cooperate to regulate inflammatory processes. This review summarizes the plasticity of macrophages and MSCs within the immune microenvironment during sepsis, as well as the intricate crosstalk between them. This remains an important concern for the future use of these cells for immunomodulatory treatments in the clinic.
Collapse
Affiliation(s)
- Xingyu Tao
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Jialian Wang
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Bin Liu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Peifeng Cheng
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Dan Mu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Niu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Mao XF, Zhang XQ, Yao ZY, Mao HJ. Advances in mesenchymal stem cells therapy for tendinopathies. Chin J Traumatol 2024; 27:11-17. [PMID: 38052701 PMCID: PMC10859297 DOI: 10.1016/j.cjtee.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023] Open
Abstract
Tendinopathies are chronic diseases of an unknown etiology and associated with inflammation. Mesenchymal stem cells (MSCs) have emerged as a viable therapeutic option to combat the pathological progression of tendinopathies, not only because of their potential for multidirectional differentiation and self-renewal, but also their excellent immunomodulatory properties. The immunomodulatory effects of MSCs are increasingly being recognized as playing a crucial role in the treatment of tendinopathies, with MSCs being pivotal in regulating the inflammatory microenvironment by modulating the immune response, ultimately contributing to improved tissue repair. This review will discuss the current knowledge regarding the application of MSCs in tendinopathy treatments through the modulation of the immune response.
Collapse
Affiliation(s)
- Xu-Feng Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Xi-Qian Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Zhe-Yu Yao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Hai-Jiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China.
| |
Collapse
|
44
|
Ti D, Yi J, Chen H, Hao H, Shi C. The Role of Mesenchymal Stem/Stromal Cells Secretome in Macrophage Polarization: Perspectives on Treating Inflammatory Diseases. Curr Stem Cell Res Ther 2024; 19:894-905. [PMID: 37723965 DOI: 10.2174/1574888x18666230811093101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 09/20/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) have exhibited potential for treating multiple inflammation- related diseases (IRDs) due to their easy acquisition, unique immunomodulatory and tissue repair properties, and immune-privileged characteristics. It is worth mentioning that MSCs release a wide array of soluble bioactive components in the secretome that modulate host innate and adaptive immune responses and promote the resolution of inflammation. As the first line of defense, macrophages exist throughout the entire inflammation process. They continuously switch their molecular phenotypes accompanied by complementary functional regulation ranging from classically activated pro-inflammatory M1-type (M1) to alternatively activated anti-inflammatory M2-type macrophages (M2). Recent studies have shown that the active intercommunication between MSCs and macrophages is indispensable for the immunomodulatory and regenerative behavior of MSCs in pharmacological cell therapy products. In this review, we systematically summarized the emerging capacities and detailed the molecular mechanisms of the MSC-derived secretome (MSC-SE) in immunomodulating macrophage polarization and preventing excessive inflammation, providing novel insights into the clinical applications of MSC-based therapy in IRD management.
Collapse
Affiliation(s)
| | - Jun Yi
- Newlife R&D Center, Beijing, China
| | | | | | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
45
|
Zhou YK, Han CS, Zhu ZL, Chen P, Wang YM, Lin S, Chen LJ, Zhuang ZM, Zhou YH, Yang RL. M2 exosomes modified by hydrogen sulfide promoted bone regeneration by moesin mediated endocytosis. Bioact Mater 2024; 31:192-205. [PMID: 37593496 PMCID: PMC10429289 DOI: 10.1016/j.bioactmat.2023.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023] Open
Abstract
Bone defects caused by trauma or tumor led to high medical costs and poor life quality for patients. The exosomes, micro vesicles of 30-150 nm in diameter, derived from macrophages manipulated bone regeneration. However, the role of hydrogen sulfide (H2S) in the biogenesis and function of exosomes and its effects on bone regeneration remains elusive. In this study, we used H2S slow releasing donor GYY4137 to stimulate macrophages and found that H2S promoted the polarization of M2 macrophages to increase bone regeneration of MSCs in vitro and in vivo. Moreover, we developed the H2S pre-treated M2 macrophage exosomes and found these exosomes displayed significantly higher capacity to promote bone regeneration in calvarial bone defects by re-establishing the local immune microenvironment. Mechanically, H2S treatment altered the protein profile of exosomes derived from M2 macrophages. One of the significantly enriched exosomal proteins stimulated by H2S, moesin protein, facilitated the exosomes endocytosis into MSCs, leading to activated the β-catenin signaling pathway to promote osteogenic differentiation of MSCs. In summary, H2S pretreated M2 exosomes promoted the bone regeneration of MSCs via facilitating exosomes uptake by MSCs and activate β-catenin signaling pathway. This study not only provides new strategies for promoting bone regeneration, but also provides new insights for the effect and mechanism of exosomes internalization.
Collapse
Affiliation(s)
- Yi-kun Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Chun-shan Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-lu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Peng Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yi-ming Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Liu-jing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-meng Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yan-heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Rui-li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| |
Collapse
|
46
|
Xue Z, Liao Y, Li Y. Effects of microenvironment and biological behavior on the paracrine function of stem cells. Genes Dis 2024; 11:135-147. [PMID: 37588208 PMCID: PMC10425798 DOI: 10.1016/j.gendis.2023.03.013] [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: 01/08/2023] [Revised: 02/14/2023] [Accepted: 03/05/2023] [Indexed: 08/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs), the most well-studied cell type in the field of stem cell therapy, have multi-lineage differentiation and self-renewal potential. MSC-based therapies have been used to treat diverse diseases because of their ability to potently repair tissue and locally restore function. An increasing body of evidence demonstrates that paracrine function is central to the effects of MSC-based therapy. Growth factors, cytokines, chemokines, extracellular matrix components, and extracellular vehicles all contribute to the beneficial effects of MSCs on tissue regeneration and repair. The paracrine substances secreted by MSCs change depending on the tissue microenvironment and biological behavior. In this review, we discuss the bioactive substances secreted by MSCs depending on the microenvironment and biological behavior and their regulatory mechanisms, which explain their potential to treat human diseases, to provide new ideas for further research and clinical cell-free therapy.
Collapse
Affiliation(s)
- Zhixin Xue
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yunjun Liao
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ye Li
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
47
|
Bai X, Chen T, Li Y, Ge X, Qiu C, Gou H, Wei S, Liu T, Yang W, Yang L, Liang Y, Jia Z, Lv L, Li T. PD-L1 expression levels in mesenchymal stromal cells predict their therapeutic values for autoimmune hepatitis. Stem Cell Res Ther 2023; 14:370. [PMID: 38111045 PMCID: PMC10729378 DOI: 10.1186/s13287-023-03594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Autoimmune hepatitis is a chronic inflammatory hepatic disorder with no effective treatment. Mesenchymal stromal cells (MSCs) have emerged as a promising treatment owing to their unique advantages. However, their heterogeneity is hampering use in clinical applications. METHODS Wharton's jelly derived MSCs (WJ-MSCs) were isolated from 58 human donors using current good manufacturing practice conditions. Gene expression profiles of the WJ-MSCs were analyzed by transcriptome and single-cell RNA-sequencing (scRNA-seq), and subsequent functional differences were assessed. Expression levels of programmed death-ligand 1 (PD-L1) were used as an indicator to screen WJ-MSCs with varied immunomodulation activities and assessed their corresponding therapeutic effects in a mouse model of concanavalin A-induced autoimmune hepatitis. RESULTS The 58 different donor-derived WJ-MSCs were grouped into six gene expression profile clusters. The gene in different clusters displayed obvious variations in cell proliferation, differentiation bias, trophic factor secretion, and immunoregulation. Data of scRNA-seq revealed four distinct WJ-MSCs subpopulations. Notably, the different immunosuppression capacities of WJ-MSCs were positively correlated with PD-L1 expression. WJ-MSCs with high expression of PD-L1 were therapeutically superior to WJ-MSCs with low PD-L1 expression in treating autoimmune hepatitis. CONCLUSION PD-L1 expression levels of WJ-MSCs could be regarded as an indicator to choose optimal MSCs for treating autoimmune disease. These findings provided novel insights into the quality control of MSCs and will inform improvements in the therapeutic benefits of MSCs.
Collapse
Affiliation(s)
- Xilong Bai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Yuqi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Xiaofan Ge
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Caie Qiu
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Huili Gou
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Sili Wei
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Tingting Liu
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Wei Yang
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Liting Yang
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Yingmin Liang
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Zhansheng Jia
- Department of Infection and Liver Disease, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Liangshan Lv
- Department of Minimally Invasive Interventional Radiology, Xi'an Gaoxin Hospital, Xi'an, , 710075, Shaanxi, China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China.
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China.
| |
Collapse
|
48
|
Yu S, Lu J. The potential of mesenchymal stem cells to induce immune tolerance to allogeneic transplants. Transpl Immunol 2023; 81:101939. [PMID: 37866668 DOI: 10.1016/j.trim.2023.101939] [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: 05/03/2023] [Revised: 10/04/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Organ allograft transplantation is an effective treatment plan for patients with organ failure. Although the application of continuous immunosuppressants makes successful allograft survival possible, the patients' long-term survival rate and quality of life are not ideal. Therefore, it is necessary to find a new strategy to alleviate transplant rejection by developing therapies for permanent allograft acceptance. One promising approach is the application of tolerogenic mesenchymal stem cells (MSCs). Extensive research on MSCs has revealed that MSCs have potent differentiation potential and immunomodulatory properties. This review describes the molecular markers and functional properties of MSCs as well as the immunomodulatory mechanisms of MSCs in transplantation, focuses on the research progress in clinical trials of MSCs, and expounds on the future development prospects and possible limitations.
Collapse
Affiliation(s)
- Shaochen Yu
- Department of Emergency and Critical Care Medicine, Guangdong Second Provincial General Hospital, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, Guangdong 510317, China.
| | - Jian Lu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, China.
| |
Collapse
|
49
|
Liu J, Deng Y, Wang A, Liu B, Zhou X, Yin T, Wang Y, Tang T, Qiu Y, Chen J, Yang J. Investigation into the role of the MITA-TRIM38 interaction in regulating pyroptosis and maintaining immune tolerance at the maternal-fetal interface. Cell Death Dis 2023; 14:780. [PMID: 38012139 PMCID: PMC10682411 DOI: 10.1038/s41419-023-06314-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: 07/13/2023] [Revised: 10/29/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The maternal-fetal interface shares similarities with tumor tissues in terms of the immune microenvironment. Normal pregnancy is maintained due to the immunosuppressed state, but pyroptosis induced by MITA can trigger the body's immune response and disrupt the immunosuppressed state of the maternal-fetal interface, leading to abortion. In this study, we explored the role of MITA and TRIM38 in regulating pyroptosis and maintaining the immune tolerance of the maternal-fetal interface during pregnancy. Our findings show that the interaction between MITA and TRIM38 plays a crucial role in maintaining the immunosuppressed state of the maternal-fetal interface. Specifically, we observed that TRIM38-mediated K48 ubiquitination of MITA was higher in M2 macrophages, leading to low expression levels of MITA and thus inhibiting pyroptosis. Conversely, in M1 macrophages, the ubiquitination of K48 was lower, resulting in higher expression levels of MITA and promoting pyroptosis. Our results also indicated that pyroptosis played an important role in hindering the transformation of M1 to M2 and maintaining the immunosuppressed state of the maternal-fetal interface. These discoveries help elucidate the mechanisms that support the preservation of the immune tolerance microenvironment at the maternal-fetal interface, playing a vital role in ensuring successful pregnancy.
Collapse
Affiliation(s)
- Jun Liu
- Reproductive Medical Center, Renmin Hospital, Wuhan University, Wuhan, China
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yan Deng
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - An Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences (CAS), Wuhan, China
| | - Bowen Liu
- Reproductive Medical Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xi Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences (CAS), Wuhan, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yan Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Tao Tang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Yang Qiu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences (CAS), Wuhan, China.
| | - Jiao Chen
- Reproductive Medical Center, Renmin Hospital, Wuhan University, Wuhan, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital, Wuhan University, Wuhan, China.
| |
Collapse
|
50
|
Chen X, Song QL, Wang JY, Ji R, Cao ML, Guo DY, Zhang Y, Yang J. FKBP5 regulates trophoblast-macrophage crosstalk in recurrent spontaneous abortion through PI3K/AKT and NF-κB signaling pathways. Free Radic Biol Med 2023; 209:55-69. [PMID: 37827456 DOI: 10.1016/j.freeradbiomed.2023.10.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/23/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
FK506-binding protein 5 (FKBP5) contributes to many diseases; However, it remains unclear whether FKBP5 is relevant to recurrent spontaneous abortion (RSA) and the mechanisms by which it is involved in maternal-fetal immunological tolerance. Placental tissue was collected in women with normal pregnancy and RSA and examined for FKBP5 expression. Human trophoblast cell lines and THP-1-derived M0 macrophages were used to explore the role of FKBP5 in RSA and its mechanism. The role of FKBP5 on pregnancy outcomes was assessed using a mouse model of miscarriage. This study found that upregulation of FKBP5 at the placental interface is involved in the pathogenesis of RSA by depressing trophoblast function and promoting M1-type macrophage polarization. First, FKBP5 expression was upregulated in the villi of RSA, and FKBP5 regulated trophoblast function by inhibiting HAPLN1 expression through suppression of PI3K/AKT signaling. In addition, FKBP5 inhibited trophoblast IL-6 secretion by suppressing PI3K/AKT signaling, thereby promoting macrophage polarization toward the M1 phenotype. Meanwhile, FKBP5 was significantly elevated in decidual macrophages from patients with RSA and promoted M1 macrophage polarization via ROS/NF-κB signaling and further inhibited trophoblast function. Finally, FKBP5 inhibitors improved embryo resorption rate in miscarried mice. In conclusion, FKBP5 is essential in maintaining pregnancy and trophoblast-macrophage crosstalk in the maternal-fetal interface, which may be a potential target for diagnosing and treating RSA.
Collapse
Affiliation(s)
- Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Qian Lin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jia Yu Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Rui Ji
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Ming Liang Cao
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Duan Ying Guo
- Longgang District People's Hospital of Shenzhen, Shenzhen, China.
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China.
| |
Collapse
|