1
|
Zhang X, Shao W, Gao Y, Wang X. Macrophage polarization-mediated PKM2/mTORC1/YME1L signaling pathway activation in fibrosis associated with Cardiorenal syndrome. Cell Signal 2025; 131:111664. [PMID: 39961408 DOI: 10.1016/j.cellsig.2025.111664] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay between cardiac and renal dysfunction, often culminating in renal fibrosis. The role of macrophage polarization and its downstream effects in CRS-induced renal fibrosis remains an area of active investigation. METHODS Single-cell RNA sequencing (scRNA-seq) and immune infiltration analyses were employed to identify key immune cells and genes involved in renal fibrosis in CRS. Meta-analysis and pseudo-time analysis were conducted to validate the functional relevance of these genes. Functional studies utilizing CRISPR/Cas9 gene editing and lentiviral vectors assessed macrophage polarization and epithelial-to-mesenchymal transition (EMT). In vivo, a CRS mouse model was established, and fibrosis progression was tracked using histological and imaging methods. RESULTS The PKM2/mTORC1/YME1L signaling axis was identified as a critical pathway driving renal fibrosis, mediated by HIF-1α-induced M1 macrophage polarization. Inhibition of HIF-1α significantly alleviated renal fibrosis by restricting M1 polarization and suppressing the PKM2/mTORC1/YME1L axis. Co-culture models further demonstrated the involvement of EMT and metabolic reprogramming in affected cells. CONCLUSION Targeting the HIF-1α signaling pathway offers a promising therapeutic strategy for renal fibrosis by modulating macrophage polarization and the PKM2/mTORC1/YME1L axis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| | - Wen Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yun Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaojun Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
2
|
Zou T, Tang X, Wang H, Shang X, Liang X, Ma X. Nanocrystalline cellulose-geniposide complex enhances gut-brain axis modulation for depression treatment. Commun Biol 2025; 8:667. [PMID: 40287572 PMCID: PMC12033350 DOI: 10.1038/s42003-025-07934-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite's effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China.
| | - Xiang Tang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Haiping Wang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaolong Shang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaoyu Liang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xuemiao Ma
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| |
Collapse
|
3
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [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: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
4
|
Lu L, Wang L, Yang M, Wang H. Role of METTL16 in PPARγ methylation and osteogenic differentiation. Cell Death Dis 2025; 16:271. [PMID: 40210616 PMCID: PMC11986173 DOI: 10.1038/s41419-025-07527-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: 06/23/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/12/2025]
Abstract
Osteoporosis, a prevalent bone disease, is characterized by the deterioration of bone tissue microstructure and imbalanced osteogenesis. The regulatory role of PPARγ m6A methylation mediated by METTL16 remains poorly elucidated. This study utilized advanced single-cell RNA sequencing (scRNA-seq) and Bulk RNA-seq techniques to explore how METTL16 influences the osteogenic differentiation of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) and its implication in osteoporosis. The research revealed that METTL16 enhances the suppression of osteogenic differentiation in BMSCs, while PPARγ is associated with BMSC ferroptosis. Mechanistically, METTL16 facilitates the m6A modification of PPARγ transcription, thereby promoting ferroptosis in BMSCs and impeding their osteogenic differentiation. The in vivo animal experiments confirmed the pivotal role of the METTL16-PPARγ axis in osteoporosis development in mice. These findings suggest that the regulation of PPARγ m6A methylation by METTL16, leading to ferroptosis, is a critical mechanism impacting BMSC osteogenic differentiation and the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, Ningbo, China.
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Minjie Yang
- Department of Orthopaedics, Jiu jiang NO.1 People's Hospital, Jiu jiang, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Guo J, Wang K, Sun Q, Liu J, Zheng J. Targeting B4GALT3 in BMSCs-EVs for Therapeutic Control of HCC via NF-κB pathway inhibition. Cell Biol Toxicol 2025; 41:67. [PMID: 40186771 PMCID: PMC11972216 DOI: 10.1007/s10565-025-10013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Examining the communications in the tumor microenvironment (TME) specific to hepatocellular carcinoma (HCC), this exploration looks into the role played by beta-1,4-Galactosyltransferase III (B4GALT3) in bone marrow mesenchymal stromal cell-derived extracellular vesicles (BMSCs-EVs) regarding the NF-κB pathway and the triggering of cancer-associated fibroblasts (CAF). Through a multidisciplinary approach combining transcriptome sequencing, bioinformatic analysis, and various experimental models, the involvement of B4GALT3 in regulating CAF activity by modulating NF-κB signaling was brought to light in our study. The outcomes suggest that targeting B4GALT3 could impede HCC cell migration and invasion, promote apoptosis, and dampen tumor progression and metastasis, offering novel insights into potential therapeutic strategies for combating HCC.
Collapse
Affiliation(s)
- Juncheng Guo
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Kaiqiong Wang
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Qigang Sun
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jun Liu
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jinfang Zheng
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
6
|
Zhang P, Hu J, Liu X, Li Y, Pang S, Liu S. Antiadhesion Biomaterials in Tendon Repair: Application Status and Future Prospect. TISSUE ENGINEERING. PART B, REVIEWS 2025; 31:20-30. [PMID: 38534262 DOI: 10.1089/ten.teb.2023.0313] [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: 03/28/2024]
Abstract
The healing process after tendon injury is often accompanied by the formation of peritendinous adhesion, contributing to limb dysfunction and exerting detrimental effects on the individuals, as well as the development of society and economy. With the continuous development of material science, as well as the augmented understanding of tendon healing and the mechanism of peritendinous adhesion formation, materials used for the fabrication of barrier membranes against peritendinous adhesion emerge endlessly. In this article, based on the analysis of the mechanism of adhesion formation, we first review the commonly used natural and synthetic materials, along with their corresponding fabrication strategies, in order to furnish valuable insights for the future optimization and development of antiperitendinous adhesion barrier membranes. This article also discusses the interaction between antiadhesion materials and cells for ameliorating peritendinous adhesion.
Collapse
Affiliation(s)
- Peilin Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiacheng Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Xiaonan Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhao Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sa Pang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shen Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Zou P, Tao Z, Yang Z, Xiong T, Deng Z, Chen Q. The contribution of the novel CLTC-VMP1 fusion gene to autophagy regulation and energy metabolism in cisplatin-resistant osteosarcoma. Am J Physiol Cell Physiol 2025; 328:C148-C167. [PMID: 39466176 DOI: 10.1152/ajpcell.00302.2024] [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: 05/06/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024]
Abstract
Osteosarcoma (OS) is a highly malignant tumor, and chemotherapy resistance is a major challenge in the treatment of this disease. This study aims to explore the role of the CLTC-VMP1 gene fusion in the mechanism of chemotherapy resistance in OS and investigate its molecular mechanisms in mediating energy metabolism reprogramming by regulating autophagy and apoptosis balance. Using single-cell transcriptome analysis, the heterogeneity of OS cells and their correlation with resistance to platinum drugs were revealed. Cisplatin-resistant cell lines were established in human OS cell lines for subsequent experiments. Based on transcriptomic analysis, the importance of VMP1 in chemotherapy resistance was confirmed. Lentiviral vectors overexpressing or interfering with VMP1 were used, and it was observed that inhibiting VMP1 could reverse cisplatin resistance, promote cell apoptosis, and inhibit autophagy, and mitochondrial respiration and glycolysis. Furthermore, the presence of CLTC-VMP1 gene fusion was validated, and its ability to regulate autophagy and apoptosis balance, promote mitochondrial respiration, and glycolysis was demonstrated. Mouse model experiments further confirmed the promoting effect of CLTC-VMP1 on tumor growth and chemotherapy resistance. In summary, the CLTC-VMP1 gene fusion mediates energy metabolism reprogramming by regulating autophagy and apoptosis balance, which promotes chemotherapy resistance in OS.NEW & NOTEWORTHY This study identifies the CLTC-VMP1 gene fusion as a key driver of chemotherapy resistance in osteosarcoma by regulating autophagy and reprogramming cellular energy metabolism. Through single-cell transcriptomics, the research reveals the heterogeneity of tumor cells and the role of VMP1 in promoting resistance to cisplatin. The findings suggest that targeting the CLTC-VMP1 fusion gene may offer new therapeutic strategies to overcome chemotherapy resistance in osteosarcoma.
Collapse
Affiliation(s)
- Pingan Zou
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhiwei Tao
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhengxu Yang
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Tao Xiong
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhi Deng
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Qincan Chen
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| |
Collapse
|
8
|
Sun Q, Li Y, Yang W, Feng W, Zhou J, Cao Y, Zhang B, Zhu Z, Han C. CircMYH9/miR-133a-3p/CXCR4 axis: a novel regulatory network in sperm fertilization and embryo development. MOLECULAR BIOMEDICINE 2024; 5:69. [PMID: 39671083 PMCID: PMC11645365 DOI: 10.1186/s43556-024-00236-5] [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: 05/16/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
This study aimed to investigate the influence of sperm miRNAs on fertilization rates (FR) in in vitro fertilization (IVF) and to explore potential regulatory mechanisms in sperm-mediated fertilization and embryo development. Through high-throughput sequencing, we identified differentially expressed miRNAs in sperm, with miR-133a-3p significantly upregulated in samples associated with low FR and available embryo rate (AER). Key regulatory circRNAs and mRNAs were further identified via the Starbase database, intersected with differentially expressed RNA, and analyzed through GO, KEGG, and PPI analyses. The circMYH9/miR-133a-3p/CXCR4 axis emerged as a critical regulatory network. In vitro assays using the GC-2 spd mouse spermatogenic cell line revealed that miR-133a-3p inhibited cell growth and proliferation while promoting apoptosis. circMYH9, acting as a competing endogenous RNA (ceRNA) for miR-133a-3p, modulated CXCR4 expression, enhancing GC-2 spd cell growth and inhibiting apoptosis through the miR-133a-3p/CXCR4 axis. In vivo experiments using a mouse model confirmed that circMYH9 overexpression increased IVF success rates and promoted embryo development via this axis. Mechanistically, miR-133a-3p suppresses sperm fertilization and embryo development by targeting the circMYH9/miR-133a-3p/CXCR4 axis. These findings suggest that this regulatory network could serve as a novel biomarker for assessing fertilization potential and embryo quality in clinical settings and as a potential therapeutic target to improve IVF outcomes and address infertility. This study provides valuable insights into the molecular mechanisms governing sperm function and early embryonic development.
Collapse
Affiliation(s)
- Qian Sun
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Yanyu Li
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China
| | - Wen Yang
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Wen Feng
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Jiayun Zhou
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China
| | - Yijuan Cao
- Department of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Bei Zhang
- Suzhou Medical College, Soochow University, Suzhou, 215123, China.
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China.
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China.
| | - Zuobin Zhu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Conghui Han
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China.
- Department of Urology, Xuzhou Central Hospital, Xuzhou, 221009, China.
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, 221009, China.
- School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, China.
- Department of Urology, Heilongjiang Provincial Hospital, Harbin, 150006, China.
| |
Collapse
|
9
|
Abozaid M, Adam E, Sarcon A, An KN, Zhao C. Biomechanical properties of various rat rotator cuff repair techniques. J Biomech 2024; 177:112399. [PMID: 39514986 DOI: 10.1016/j.jbiomech.2024.112399] [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: 05/17/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
While rat models are frequently used to study tendon healing, there is a lack of research comparing various rotator cuff repair methods in this animal model. Determining the most effective method to begin with is pivotal for biological studies focused on healing augmentation. No study to date has shown the superiority of one repair over the other for rotator cuff repair in a rat model. We performed a biomechanic study using a rat model to study the strength of four common grasping techniques. We assessed if the bone tunnel trajectory influenced the early biomechanics of the repair at postoperative day 0 (POD0). Sixty cadaveric rat shoulders were divided equally into 6 groups; 4 groups were allocated for the biomechanical strength testing based on either a (1) modified Mason Allen (MM), (2) modified Kessler loop (MK), (3) horizontal mattress (HM), or a (4) simple interrupted stitch (SS) technique. The remaining 2 groups were used to evaluate two tunneling angles: a transverse tunnel (TT) that was perpendicular to the long humeral axis, or a longitudinal tunnel (LT) that was 30◦ angle to the humerus. MM had the highest mean failure load, followed by MK, HM, and SS. Pairwise comparison revealed that MM was stronger than SS and HM (P = 0.025 and P = 0.026, respectively), although similar to the MK (P = 0.881). MM was stiffer than MK (P < 0.001), HM (P = 0.008), and SS (P < 0.001). The TT and LT had similar loads to failure and stiffness. Our study suggests that the MM technique provides a stronger and stiffer rotator cuff repair than the others.
Collapse
Affiliation(s)
- Mohamed Abozaid
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Elameen Adam
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Aida Sarcon
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Kai-Nan An
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
10
|
Yan F, Yuan WQ, Wu SM, Yang YH, Cui DJ. Novel mechanisms of intestinal flora regulation in high-altitude hypoxia. Heliyon 2024; 10:e38220. [PMID: 39498080 PMCID: PMC11534185 DOI: 10.1016/j.heliyon.2024.e38220] [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: 03/05/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
Background This study investigates the molecular mechanisms behind firmicutes-mediated macrophage (Mψ) polarization and glycolytic metabolic reprogramming through HIF-1α in response to intrinsic mucosal barrier injury induced by high-altitude hypoxia. Methods Establishing a hypoxia mouse model of high altitude, we utilized single-cell transcriptome sequencing to identify key cell types involved in regulating intestinal mucosal barrier damage caused by high-altitude hypoxia. Through proteomic analysis of colonic tissue Mψ and metabolomic analysis of Mψ metabolites, we determined crucial proteins and metabolic pathways influencing intestinal mucosal barrier damage induced by high-altitude hypoxia. Mechanistic validation was conducted using RAW264.7 Mψ in vitro by assessing cell viability with CCK-8 assay following treatment with different metabolites. The hypoxia mouse model was further validated in vivo by transplanting gut microbiota of Firmicutes. Histological examinations through H&E staining assessed colonic cell morphology and structure, while the FITC-dextran assay evaluated intestinal tissue permeability. Hypoxia probe signal intensity in mouse colonic tissue was assessed via metronidazole staining. Various experimental techniques, including flow cytometry, immunofluorescence, ELISA, Western blot, and RT-qPCR, were employed to study the impact of HIF-1α/glycolysis pathway and different gut microbiota metabolites on Mψ polarization. Results Bioinformatics analysis revealed that single-cell transcriptomics identified Mψ as a key cell type, with their polarization pattern playing a crucial role in the intestinal mucosal barrier damage induced by high-altitude hypoxia. Proteomics combined with metabolomics analysis indicated that HIF-1α and the glycolytic pathway are pivotal proteins and signaling pathways in the intestinal mucosal barrier damage caused by high-altitude hypoxia. In vitro cell experiments demonstrated that activation of the glycolytic pathway by HIF-1α led to a significant upregulation of mRNA levels of IL-1β, IL-6, and TNFα while downregulating mRNA levels of IL-10 and TGFβ, thereby promoting M1 Mψ activation and inhibiting M2 Mψ polarization. Further mechanistic validation experiments revealed that the metabolite butyric acid from Firmicutes bacteria significantly downregulated the protein expression of HIF-1α, GCK, PFK, PKM, and LDH, thus inhibiting the HIF-1α/glycolytic pathway that suppresses M1 Mψ and activates M2 Mψ, consequently alleviating the hypoxic symptoms in RAW264.7 cells. Subsequent animal experiments confirmed that Firmicutes bacteria inhibited the HIF-1α/glycolytic pathway to modulate Mψ polarization, thereby mitigating intestinal mucosal barrier damage in high-altitude hypoxic mice. Conclusion The study reveals that firmicutes, through the inhibition of the HIF-1α/glycolysis pathway, mitigate Mψ polarization, thereby alleviating intrinsic mucosal barrier injury in high-altitude hypoxia.
Collapse
Affiliation(s)
- Fang Yan
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Wen-qiang Yuan
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Shi-min Wu
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Zunyi Medical University, Zunyi, 563006, China
| | - Yun-han Yang
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - De-jun Cui
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| |
Collapse
|
11
|
Liu W, Xiong Z, Fu T, Yang J, Zou J, Wu Y, Kuang L, Wang Q, Li S, Le A. Regulation of renal ischemia-reperfusion injury and tubular epithelial cell ferroptosis by pparγ m6a methylation: mechanisms and therapeutic implications. Biol Direct 2024; 19:99. [PMID: 39444036 PMCID: PMC11515743 DOI: 10.1186/s13062-024-00515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/08/2024] [Indexed: 10/25/2024] Open
Abstract
This study aimed to elucidate the role and underlying mechanisms of Peroxisome proliferator-activated receptor gamma (PPARγ) and its m6A methylation in renal ischemia-reperfusion (I/R) injury and ferroptosis of tubular epithelial cells (TECs). High-throughput transcriptome sequencing was performed on renal tissue samples from I/R injury models and sham-operated mice, complemented by in vivo and in vitro experiments focusing on the PPARγ activator Rosiglitazone and the manipulation of METTL14 and IGF2BP2 expression. Key evaluations included renal injury assessment, ferroptosis indicator measurement, and m6A methylation analysis of PPARγ. Our findings highlight the critical role of the PPARγ pathway and ferroptosis in renal I/R injury, with Rosiglitazone ameliorating renal damage and TEC ferroptosis. METTL14-mediated m6A methylation of PPARγ, dependent on IGF2BP2, emerged as a pivotal regulator of PPARγ expression, renal injury, and ferroptosis. This study reveals that PPARγ m6A methylation, orchestrated by METTL14 through an IGF2BP2-dependent mechanism, plays a crucial role in mitigating renal I/R injury and TEC ferroptosis. These insights offer promising avenues for therapeutic strategies targeting acute kidney injury.
Collapse
Affiliation(s)
- Wei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Ziqing Xiong
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Tianmei Fu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Juan Yang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Juan Zou
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Yize Wu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Linju Kuang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Qian Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Song Li
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
12
|
Zhan T, Zou Y, Han Z, Tian X, Chen M, Liu J, Yang X, Zhu Q, Liu M, Chen W, Chen M, Huang X, Tan J, Liu W, Tian X. Single-cell sequencing combined with spatial transcriptomics reveals that the IRF7 gene in M1 macrophages inhibits the occurrence of pancreatic cancer by regulating lipid metabolism-related mechanisms. Clin Transl Med 2024; 14:e1799. [PMID: 39118300 PMCID: PMC11310283 DOI: 10.1002/ctm2.1799] [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: 07/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
AIM The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression. METHODS By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry. RESULTS Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis. CONCLUSION Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
Collapse
Affiliation(s)
- Ting Zhan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Yanli Zou
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Zheng Han
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - XiaoRong Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Mengge Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiaxi Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiulin Yang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qingxi Zhu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Meng Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Wei Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Mingtao Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xiaodong Huang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jie Tan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Weijie Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xia Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| |
Collapse
|
13
|
Wan M, Yu Q, Xu F, You LX, Liang X, Kang Ren K, Zhou J. Novel hypoxia-induced HIF-1αactivation in asthma pathogenesis. Respir Res 2024; 25:287. [PMID: 39061007 PMCID: PMC11282634 DOI: 10.1186/s12931-024-02869-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Asthma's complexity, marked by airway inflammation and remodeling, is influenced by hypoxic conditions. This study focuses on the role of Hypoxia-Inducible Factor-1 Alpha (HIF-1α) and P53 ubiquitination in asthma exacerbation. METHODS High-throughput sequencing and bioinformatics were used to identify genes associated with asthma progression, with an emphasis on GO and KEGG pathway analyses. An asthma mouse model was developed, and airway smooth muscle cells (ASMCs) were isolated to create an in vitro hypoxia model. Cell viability, proliferation, migration, and apoptosis were assessed, along with ELISA and Hematoxylin and Eosin (H&E) staining. RESULTS A notable increase in HIF-1α was observed in both in vivo and in vitro asthma models. HIF-1α upregulation enhanced ASMCs' viability, proliferation, and migration, while reducing apoptosis, primarily via the promotion of P53 ubiquitination through MDM2. In vivo studies showed increased inflammatory cell infiltration and airway structural changes, which were mitigated by the inhibitor IDF-11,774. CONCLUSION The study highlights the critical role of the HIF-1α-MDM2-P53 axis in asthma, suggesting its potential as a target for therapeutic interventions. The findings indicate that modulating this pathway could offer new avenues for treating the complex respiratory disorder of asthma.
Collapse
Affiliation(s)
- Mengzhi Wan
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Qi Yu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Fei Xu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Lu Xia You
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiao Liang
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Kang Kang Ren
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Jing Zhou
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
14
|
Shao G, Cui X, Wang Y, Luo S, Li C, Jiang Y, Cai D, Li N, Li X. Targeting MS4A4A: A novel pathway to improve immunotherapy responses in glioblastoma. CNS Neurosci Ther 2024; 30:e14791. [PMID: 38997808 PMCID: PMC11245405 DOI: 10.1111/cns.14791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 07/14/2024] Open
Abstract
INTRODUCTION Glioblastoma (GBM) remains a challenging brain tumor to treat, with limited response to PD-1 immunotherapy due to tumor-associated macrophages (TAMs), specifically the M2 phenotype. This study explores the potential of MS4A4A (membrane spanning four domains, subfamily A, member 4A) inhibition in driving M2 macrophage polarization toward the M1 phenotype via the ferroptosis pathway to enhance the effectiveness of immunotherapy in GBM. METHODS Single-cell RNA sequencing and spatial transcriptomic analyses were employed to characterize M2 macrophages and MS4A4A expression in GBM. In vitro studies utilizing TAM cultures, flow cytometry, and western blot validations were conducted to assess the impact of MS4A4A on the tumor immune microenvironment and M2 macrophage polarization. In vivo models, including subcutaneous and orthotopic transplantation in mice, were utilized to evaluate the effects of MS4A4A knockout and combined immune checkpoint blockade (ICB) therapy on tumor growth and response to PD-1 immunotherapy. RESULTS Distinct subsets of GBM-associated macrophages were identified, with spatial distribution in tumor tissue elucidated. In vivo experiments demonstrated that inhibiting MS4A4A and combining ICB therapy effectively inhibited tumor growth, reshaped the tumor immune microenvironment by reducing M2 TAM infiltration and enhancing CD8+ T-cell infiltration, ultimately leading to complete tumor eradication. CONCLUSION MS4A4A inhibition shows promise in converting M2 macrophages to M1 phenotype via ferroptosis, decreasing M2-TAM infiltration, and enhancing GBM response to PD-1 immunotherapy. These findings offer a novel approach to developing more effective immunotherapeutic strategies for GBM.
Collapse
Affiliation(s)
- Guangcai Shao
- Department of Neurosurgery, Shengjing HospitalChina Medical UniversityShenyangChina
- Department of NeurosurgeryAnshan Central HospitalAnshanChina
| | - Xiangguo Cui
- Department of Otolaryngology Head and Neck Surgery, Shengjing HospitalChina Medical UniversityShenyangChina
| | - Yiliang Wang
- Department of AnesthesiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Shuyan Luo
- Department of NeurosurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Chuanyu Li
- Department of NeurosurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Yu Jiang
- Department of NeurosurgeryAnshan Central HospitalAnshanChina
| | - Dasheng Cai
- Department of AnesthesiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Nu Li
- Department of Breast SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Xiang Li
- Department of NeurosurgeryThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
15
|
Dec P, Żyłka M, Burszewski P, Modrzejewski A, Pawlik A. Recent Advances in the Use of Stem Cells in Tissue Engineering and Adjunct Therapies for Tendon Reconstruction and Future Perspectives. Int J Mol Sci 2024; 25:4498. [PMID: 38674084 PMCID: PMC11050411 DOI: 10.3390/ijms25084498] [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: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Due to their function, tendons are exposed to acute injuries. This type of damage to the musculoskeletal system represents a challenge for clinicians when natural regeneration and treatment methods do not produce the expected results. Currently, treatment is long and associated with long-term complications. In this review, we discuss the use of stem cells in the treatment of tendons, including how to induce appropriate cell differentiation based on gene therapy, growth factors, tissue engineering, proteins involved in regenerative process, drugs and three-dimensional (3D) structures. A multidirectional approach as well as the incorporation of novel components of the therapy will improve the techniques used and benefit patients with tendon injuries in the future.
Collapse
Affiliation(s)
- Paweł Dec
- Plastic and Reconstructive Surgery Department, 109 Military Hospital, 71-422 Szczecin, Poland; (P.D.); (M.Ż.); (P.B.)
| | - Małgorzata Żyłka
- Plastic and Reconstructive Surgery Department, 109 Military Hospital, 71-422 Szczecin, Poland; (P.D.); (M.Ż.); (P.B.)
| | - Piotr Burszewski
- Plastic and Reconstructive Surgery Department, 109 Military Hospital, 71-422 Szczecin, Poland; (P.D.); (M.Ż.); (P.B.)
| | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
16
|
He W, Xu C, Huang Y, Zhang Q, Chen W, Zhao C, Chen Y, Zheng D, XinyueLin, Luo Q, Chen X, Zhang Z, Wu X, Huang J, Lin C, Huang Y, Zhang S. Therapeutic potential of ADSC-EV-derived lncRNA DLEU2: A novel molecular pathway in alleviating sepsis-induced lung injury via the miR-106a-5p/LXN axis. Int Immunopharmacol 2024; 130:111519. [PMID: 38442573 DOI: 10.1016/j.intimp.2024.111519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 03/07/2024]
Abstract
This study investigates the molecular mechanisms by which extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ADSCs) promote M2 polarization of macrophages and thus reduce lung injury caused by sepsis. High-throughput sequencing was used to identify differentially expressed genes related to long non-coding RNA (lncRNA) in ADSC-derived EVs (ADSC-EVs) in sepsis lung tissue. Weighted gene co-expression network analysis (WGCNA) was employed to predict the downstream target genes of the lncRNA DLEU2. The RNAInter database predicted miRNAs that interact with DLEU2 and LXN. Functional and pathway enrichment analyses were performed using GO and KEGG analysis. A mouse model of sepsis was established, and treatment with a placebo or ADSC-EVs was administered, followed by RT-qPCR analysis. ADSC-EVs were isolated and identified. In vitro cell experiments were conducted using the mouse lung epithelial cell line MLE-12, mouse macrophage cell line RAW264.7, and mouse lung epithelial cell line (LEPC). ADSC-EVs were co-cultured with RAW264.7 and MLE-12/LEPC cells to study the regulatory mechanism of the lncRNA DLEU2. Cell viability, proliferation, and apoptosis of lung injury cells were assessed using CCK-8, EdU, and flow cytometry. ELISA was used to measure the levels of inflammatory cytokines in the sepsis mouse model, flow cytometry was performed to determine the number of M1 and M2 macrophages, lung tissue pathology was evaluated by H&E staining, and immunohistochemistry was conducted to examine the expression of proliferation- and apoptosis-related proteins. High-throughput sequencing and bioinformatics analysis revealed enrichment of the lncRNA DLEU2 in ADSC-EVs in sepsis lung tissue. Animal and in vitro cell experiments showed increased expression of the lncRNA DLEU2 in sepsis lung tissue after treatment with ADSC-EVs. Furthermore, ADSC-EVs were found to transfer the lncRNA DLEU2 to macrophages, promoting M2 polarization, reducing inflammation response in lung injury cells, and enhancing their viability, proliferation, and apoptosis inhibition. Further functional experiments indicated that lncRNA DLEU2 promotes M2 polarization of macrophages by regulating miR-106a-5p/LXN, thereby enhancing the viability and proliferation of lung injury cells and inhibiting apoptosis. Overexpression of miR-106a-5p could reverse the biological effects of ADSC-EVs-DLEU2 on MLE-12 and LEPC in vitro cell models. Lastly, in vivo animal experiments confirmed that ADSC-EVs-DLEU2 promotes high expression of LXN by inhibiting the expression of miR-106a-5p, further facilitating M2 macrophage polarization and reducing lung edema, thus alleviating sepsis-induced lung injury. lncRNA DLEU2 in ADSC-EVs may promote M2 polarization of macrophages and enhance the viability and proliferation of lung injury cells while inhibiting inflammation and apoptosis reactions, thus ameliorating sepsis-induced lung injury in a mechanism involving the regulation of the miR-106a-5p/LXN axis.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Yuying Huang
- School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, PR China
| | - Qiuzhen Zhang
- Department of Pharmacy, Jiangmen central Hospital, Jiangmen 529030, PR China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Danling Zheng
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China; Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - XinyueLin
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - Qianhua Luo
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, PR China
| | - Xiaoshan Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China
| | - Zhihan Zhang
- School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, PR China
| | - Xiaolong Wu
- College of Pharmacy, Jinan University, Guangzhou 510220, PR China
| | - Jianxiang Huang
- College of Pharmacy, Jinan University, Guangzhou 510220, PR China
| | - Chaoxian Lin
- Shantou Chaonan Minsheng Hospital, Shantou 515041, PR China.
| | - Yihui Huang
- Department of Pediatrics, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China.
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, (Guangzhou Red Cross Hospital of Jinan University), Guangzhou 510220, PR China.
| |
Collapse
|
17
|
Biological and Mechanical Factors and Epigenetic Regulation Involved in Tendon Healing. Stem Cells Int 2023; 2023:4387630. [PMID: 36655033 PMCID: PMC9842431 DOI: 10.1155/2023/4387630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Tendons are an important part of the musculoskeletal system. Connecting muscles to bones, tendons convert force into movement. Tendon injury can be acute or chronic. Noticeably, tendon healing requires a long time span and includes inflammation, proliferation, and remodeling processes. The mismatch between endogenous and exogenous healing may lead to adhesion causing further negative effects. Management of tendon injuries and complications such as subsequent adhesion formation are still challenges for clinicians. Due to numerous factors, tendon healing is a complex process. This review introduces the role of various biological and mechanical factors and epigenetic regulation processes involved in tendon healing.
Collapse
|
18
|
Jin J, Yang QQ, Zhou YL. Non-Viral Delivery of Gene Therapy to the Tendon. Polymers (Basel) 2022; 14:3338. [PMID: 36015594 PMCID: PMC9415435 DOI: 10.3390/polym14163338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/18/2022] [Indexed: 01/19/2023] Open
Abstract
The tendon, as a compact connective tissue, is difficult to treat after an acute laceration or chronic degeneration. Gene-based therapy is a highly efficient strategy for diverse diseases which has been increasingly applied in tendons in recent years. As technology improves by leaps and bounds, a wide variety of non-viral vectors have been manufactured that attempt to have high biosecurity and transfection efficiency, considered to be a promising treatment modality. In this review, we examine the unwanted biological barriers, the categories of applicable genes, and the introduction and comparison of non-viral vectors. We focus on lipid-based nanoparticles and polymer-based nanoparticles, differentiating between them based on their combination with diverse chemical modifications and scaffolds.
Collapse
Affiliation(s)
| | | | - You Lang Zhou
- Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
19
|
Titan AL, Davitt M, Foster D, Salhotra A, Menon S, Chen K, Fahy E, Lopez M, Jones RE, Baiu I, Burcham A, Januszyk M, Gurtner G, Fox P, Chan C, Quarto N, Longaker M. Partial Tendon Injury at the Tendon-to-Bone Enthesis Activates Skeletal Stem Cells. Stem Cells Transl Med 2022; 11:715-726. [PMID: 35640155 PMCID: PMC9299518 DOI: 10.1093/stcltm/szac027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/09/2022] [Indexed: 11/24/2022] Open
Abstract
The tendon enthesis plays a critical role in facilitating movement and reducing stress within joints. Partial enthesis injuries heal in a mechanically inferior manner and never achieve healthy tissue function. The cells responsible for tendon-to-bone healing remain incompletely characterized and their origin is unknown. Here, we evaluated the putative role of mouse skeletal stem cells (mSSCs) in the enthesis after partial-injury. We found that mSSCs were present at elevated levels within the enthesis following injury and that these cells downregulated TGFβ signaling pathway elements at both the RNA and protein levels. Exogenous application of TGFβ post-injury led to a reduced mSSC response and impaired healing, whereas treatment with a TGFβ inhibitor (SB43154) resulted in a more robust mSSC response. Collectively, these data suggest that mSSCs may augment tendon-to-bone healing by dampening the effects of TGFβ signaling within the mSSC niche.
Collapse
Affiliation(s)
- Ashley L Titan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Davitt
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Deshka Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ankit Salhotra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kellen Chen
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Evan Fahy
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Lopez
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - R Ellen Jones
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ioana Baiu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Austin Burcham
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Paige Fox
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles Chan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| |
Collapse
|
20
|
He P, Ruan D, Huang Z, Wang C, Xu Y, Cai H, Liu H, Fei Y, Heng BC, Chen W, Shen W. Comparison of Tendon Development Versus Tendon Healing and Regeneration. Front Cell Dev Biol 2022; 10:821667. [PMID: 35141224 PMCID: PMC8819183 DOI: 10.3389/fcell.2022.821667] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Tendon is a vital connective tissue in human skeletal muscle system, and tendon injury is very common and intractable in clinic. Tendon development and repair are two closely related but still not fully understood processes. Tendon development involves multiple germ layer, as well as the regulation of diversity transcription factors (Scx et al.), proteins (Tnmd et al.) and signaling pathways (TGFβ et al.). The nature process of tendon repair is roughly divided in three stages, which are dominated by various cells and cell factors. This review will describe the whole process of tendon development and compare it with the process of tendon repair, focusing on the understanding and recent advances in the regulation of tendon development and repair. The study and comparison of tendon development and repair process can thus provide references and guidelines for treatment of tendon injuries.
Collapse
Affiliation(s)
- Peiwen He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Honglu Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School of Stomatology, Bejing, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| |
Collapse
|
21
|
Zhao J, Wang X, Han J, Yu Y, Chen F, Yao J. Boost Tendon/Ligament Repair With Biomimetic and Smart Cellular Constructs. Front Bioeng Biotechnol 2021; 9:726041. [PMID: 34532315 PMCID: PMC8438196 DOI: 10.3389/fbioe.2021.726041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Tendon and ligament are soft connective tissues that play essential roles in transmitting forces from muscle to bone or bone to bone. Despite significant progress made in the field of ligament and tendon regeneration over the past decades, many strategies struggle to recapitulate basic structure-function criteria of native ligament/tendon. The goal here is to provide a fundamental understanding of the structure and composition of ligament/tendon and highlight few key challenges in functional regeneration of these connective tissues. The remainder of the review will examine several biomaterials strategies including biomimetic scaffold with non-linear mechanical behavior, hydrogel patch with anisotropic adhesion and gene-activated scaffold for interactive healing of tendon/ligament. Finally, emerging technologies and research avenues are suggested that have the potential to enhance treatment outcomes of tendon/ligament injuries.
Collapse
Affiliation(s)
- Jianping Zhao
- Department of Orthopedics Trauma and Hand Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Center for Materials Synthetic Biology, Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiang Wang
- Center for Materials Synthetic Biology, Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jinyu Han
- Center for Materials Synthetic Biology, Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yin Yu
- Center for Materials Synthetic Biology, Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Chen
- Center for Materials Synthetic Biology, Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jun Yao
- Department of Orthopedics Trauma and Hand Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
22
|
Gargano G, Oliviero A, Oliva F, Maffulli N. Small interfering RNAs in tendon homeostasis. Br Med Bull 2021; 138:58-67. [PMID: 33454750 DOI: 10.1093/bmb/ldaa040] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Tenogenesis and tendon homeostasis are guided by genes encoding for the structural molecules of tendon fibres. Small interfering RNAs (siRNAs), acting on gene regulation, can therefore participate in the process of tendon healing. SOURCES OF DATA A systematic search of different databases to October 2020 identified 17 suitable studies. AREAS OF AGREEMENT SiRNAs can be useful to study reparative processes of tendons and identify possible therapeutic targets in tendon healing. AREAS OF CONTROVERSY Many genes and growth factors involved in the processes of tendinopathy and tendon healing can be regulated by siRNAs. It is however unclear which gene silencing determines the expected effect. GROWING POINTS Gene dysregulation of growth factors and tendon structural proteins can be influenced by siRNA. AREAS TIMELY FOR DEVELOPING RESEARCH It is not clear whether there is a direct action of the siRNAs that can be used to facilitate the repair processes of tendons.
Collapse
Affiliation(s)
- Giuseppe Gargano
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, 84131 Salerno, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Antonio Oliviero
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, 84131 Salerno, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Francesco Oliva
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, 84131 Salerno, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, 84131 Salerno, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy.,Centre for Sports and Exercise Medicine, Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Mile End Hospital, 275 Bancroft Road, London E1 4DG, UK.,School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent, UK
| |
Collapse
|