1
|
Lv K, Gao J, Yang L, Yuan X. The role of mesenchymal stem cell‑derived exosomes in asthma (Review). Mol Med Rep 2025; 31:166. [PMID: 40242981 PMCID: PMC12012432 DOI: 10.3892/mmr.2025.13531] [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: 11/04/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Asthma is a chronic respiratory disorder characterized by persistent inflammation, airway hyper‑responsiveness and remodeling, leading to notable morbidity and decreased quality of life for patients. Mesenchymal stem cells (MSCs) have potential in regenerative medicine due to their potent immunomodulatory properties and anti‑inflammatory effects. The therapeutic benefits of MSCs are largely mediated by secreted exosomes that facilitate intercellular communication by transferring bioactive molecules, including proteins, lipids and microRNAs. The present review explores the therapeutic potential of MSC‑derived exosomes in asthma, highlighting their ability to modulate key pathological mechanisms underlying the disease.
Collapse
Affiliation(s)
- Kaiying Lv
- Department of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Jiawei Gao
- Department of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Liuxin Yang
- Department of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Xingxing Yuan
- Department of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, Heilongjiang 150006, P.R. China
| |
Collapse
|
2
|
Li M, Tang Y, Zhou C, Geng Y, Zhang C, Hsu Y, Ma L, Guo W, Li M, Wang Y. The Application of Stem Cells and Exosomes in Promoting Nerve Conduits for Peripheral Nerve Repair. Biomater Res 2025; 29:0160. [PMID: 40231207 PMCID: PMC11994886 DOI: 10.34133/bmr.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/16/2025] Open
Abstract
The repair of peripheral nerve injury (PNI) presents a multifaceted and protracted challenge, with current therapeutic approaches failing to achieve optimal repair outcomes, thereby not satisfying the considerable clinical demand. The advent of tissue engineering has led to a growing body of experimental evidence indicating that the synergistic application of nerve conduits, which provide structural guidance, alongside the biological signals derived from exosomes and stem cells, yields superior therapeutic results for PNI compared to isolated interventions. This combined approach holds great promise for clinical application. In this review, we present the latest advancements in the treatment of PNI through the integration of stem cells or exosomes with nerve conduits. We have addressed the inadequate efficiency of exosomes or stem cells in conjunction with nerve conduits from 3 perspectives: enhancing stem cells or exosomes, improving nerve conduits, and incorporating physical stimulation.
Collapse
Affiliation(s)
- Mengen Li
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| | - Ye Tang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| | - Chengkai Zhou
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yan Geng
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Chenxi Zhang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yuwei Hsu
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Emergency Department,
Peking University People’s Hospital, Beijing 100044, China
| | - Le Ma
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Wei Guo
- Emergency Department,
Peking University People’s Hospital, Beijing 100044, China
| | - Ming Li
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yanhua Wang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
3
|
Liu J, Li Y, Zhang Y, Zhao Z, Liu B. Engineered stromal vascular fraction for tissue regeneration. Front Pharmacol 2025; 16:1510508. [PMID: 40183080 PMCID: PMC11966044 DOI: 10.3389/fphar.2025.1510508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
The treatment of various tissue injuries presents significant challenges, particularly in the reconstruction of large and severe tissue defects, with conventional clinical methods often yielding suboptimal results. However, advances in engineering materials have introduced new possibilities for tissue repair. Bioactive components are commonly integrated with synthetic materials to enhance tissue reconstruction. Stromal vascular fraction (SVF), an adipose-derived cell cluster, has shown considerable potential in tissue regeneration due to its simple and efficient way of obtaining and its richness in growth factors. Therefore, this review illustrated the preparation, characterization, mechanism of action, and applications of engineered SVF in various tissue repair processes, to provide some references for the option of better methods for tissue defect reconstruction.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Yiwei Li
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Yanan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Zhiwei Zhao
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| | - Bin Liu
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, China
- Engineering Laboratory of Tissue Engineering Biomaterials of Jilin Province, Changchun, China
| |
Collapse
|
4
|
Chang MM, Chu DT, Lin SC, Lee JS, Vu TD, Vu HT, Ramasamy TS, Lin SP, Wu CC. Enhanced mitochondrial function and delivery from adipose-derived stem cell spheres via the EZH2-H3K27me3-PPARγ pathway for advanced therapy. Stem Cell Res Ther 2025; 16:129. [PMID: 40069892 PMCID: PMC11899936 DOI: 10.1186/s13287-025-04164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Microenvironmental alterations induce significant genetic and epigenetic changes in stem cells. Mitochondria, essential for regenerative capabilities, provide the necessary energy for stem cell function. However, the specific roles of histone modifications and mitochondrial dynamics in human adipose-derived stem cells (ASCs) during morphological transformations remain poorly understood. In this study, we aim to elucidate the mechanisms by which ASC sphere formation enhances mitochondrial function, delivery, and rescue efficiency. METHODS ASCs were cultured on chitosan nano-deposited surfaces to form 3D spheres. Mitochondrial activity and ATP production were assessed using MitoTracker staining, Seahorse XF analysis, and ATP luminescence assays. Single-cell RNA sequencing, followed by Ingenuity Pathway Analysis (IPA), was conducted to uncover key regulatory pathways, which were validated through molecular techniques. Pathway involvement was confirmed using epigenetic inhibitors or PPARγ-modulating drugs. Mitochondrial structural integrity and delivery efficiency were evaluated after isolation. RESULTS Chitosan-induced ASC spheres exhibited unique compact mitochondrial morphology, characterized by condensed cristae, enhanced mitochondrial activity, and increased ATP production through oxidative phosphorylation. High expressions of mitochondrial complex I genes and elevated levels of mitochondrial complex proteins were observed without an increase in reactive oxygen species (ROS). Epigenetic modification of H3K27me3 and PPARγ involvement were discovered and confirmed by inhibiting H3K27me3 with the specific EZH2 inhibitor GSK126 and by adding the PPARγ agonist Rosiglitazone (RSG). Isolated mitochondria from ASC spheres showed improved structural stability and delivery efficiency, suppressed the of inflammatory cytokines in LPS- and TNFα-induced inflamed cells, and rescued cells from damage, thereby enhancing function and promoting recovery. CONCLUSION Enhancing mitochondrial ATP production via the EZH2-H3K27me3-PPARγ pathway offers an alternative strategy to conventional cell-based therapies. High-functional mitochondria and delivery efficiency show significant potential for regenerative medicine applications.
Collapse
Affiliation(s)
- Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Dinh Toi Chu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Sheng-Che Lin
- Division of Plastic and Reconstructive Surgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, 70965, Taiwan
| | - Jung-Shun Lee
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, 701401, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thuy Duong Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Hue Thi Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, 10672, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan.
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101, Taiwan.
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
5
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2025; 27:36-50. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [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: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
6
|
Edirisinghe O, Ternier G, Alraawi Z, Suresh Kumar TK. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024; 14:1622. [PMID: 39766329 PMCID: PMC11726770 DOI: 10.3390/biom14121622] [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: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fibroblast Growth Factors (FGFs) and their cognate receptors, FGFRs, play pivotal roles in a plethora of biological processes, including cell proliferation, differentiation, tissue repair, and metabolic homeostasis. This review provides a comprehensive overview of FGF-FGFR signaling pathways while highlighting their complex regulatory mechanisms and interconnections with other signaling networks. Further, we briefly discuss the FGFs involvement in developmental, metabolic, and housekeeping functions. By complementing current knowledge and emerging research, this review aims to enhance the understanding of FGF-FGFR-mediated signaling and its implications for health and disease, which will be crucial for therapeutic development against FGF-related pathological conditions.
Collapse
Affiliation(s)
- Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Gaëtane Ternier
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Zeina Alraawi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Thallapuranam Krishnaswamy Suresh Kumar
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| |
Collapse
|
7
|
Wang Y, Zhang H, Zhan Y, Li Z, Li S, Guo S. Comprehensive in silico analysis of prognostic and immune infiltrates for FGFs in human ovarian cancer. J Ovarian Res 2024; 17:197. [PMID: 39385288 PMCID: PMC11465590 DOI: 10.1186/s13048-024-01496-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/14/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Fibroblast growth factors (FGFs) are cell signaling proteins that perform multiple biological processes in many biological processes (cell development, repair, and metabolism). The dynamics of tumor cells, such as angiogenesis, transformation, and proliferation, have a significant impact on neoplasia and are modulated by FGFs. FGFs' expression and prognostic significance in ovarian cancer (OC), however, remain unclear. METHODS Through a series of in silico analysis, we investigated the transcriptional, survival data, genetic variation, gene-gene interaction network, ferroptosis-related genes, and DNA methylation of FGFs in OC patients. RESULTS We discovered that while FGF18 expression levels were higher in OC tissues than in normal OC tissues, FGF2/7/10/17/22 expression levels were lower in the former, and that FGF1/19 expression was related to the tumor stage in OC patients. According to the survival analysis, the clinical prognosis of individuals with OC was associated with the aberrant expression of FGFs. The function of FGFs and their neighboring genes was mainly connected to the cellular response to FGF stimulus. There was a negative correlation between FGF expression and various immune cell infiltration. CONCLUSIONS This study clarifies the relationship between FGFs and OC, which might provide new insights into the choice of prognostic biomarkers of OC patients.
Collapse
Affiliation(s)
- Yu Wang
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China
| | - Haiyue Zhang
- Thrombosis research center, Beijing Jishuitan hospital, Capital Medical University, Beijing, China, Xicheng District, Beijing 100035, China
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Xicheng District, Beijing, China
| | - Yuanyuan Zhan
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, P.R. China
| | - Zhuoran Li
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China
| | - Sujing Li
- Department of Plastic Surgery, Zhengzhou First People's Hospital, Zhengzhou, China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, 100020, P.R. China.
| |
Collapse
|
8
|
Li T, Zhang Y, Zhou Z, Guan L, Zhang Y, Zhou Z, Wang W, Zhou X, Cui D, Jiang C, Ruan Y. Phosphodiesterase type 5 inhibitor tadalafil reduces prostatic fibrosis via MiR-3126-3p/FGF9 axis in benign prostatic hyperplasia. Biol Direct 2024; 19:61. [PMID: 39095835 PMCID: PMC11295313 DOI: 10.1186/s13062-024-00504-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: 03/14/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Myofibroblast buildup and prostatic fibrosis play a crucial role in the development of benign prostatic hyperplasia (BPH). Treatments specifically targeting myofibroblasts could be a promising approach for treating BPH. Tadalafil, a phosphodiesterase type 5 (PDE5) inhibitor, holds the potential to intervene in this biological process. This study employs prostatic stromal fibroblasts to induce myofibroblast differentiation through TGFβ1 stimulation. As a result, tadalafil significantly inhibited prostatic stromal fibroblast proliferation and fibrosis process, compared to the control group. Furthermore, our transcriptome sequencing results revealed that tadalafil inhibited FGF9 secretion and simultaneously improved miR-3126-3p expression via TGFβ1 suppression. Overall, TGFβ1 can trigger pro-fibrotic signaling through miR-3126-3p in the prostatic stroma, and the use of tadalafil can inhibit this process.
Collapse
Affiliation(s)
- Tiewen Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Yu Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Zeng Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Lvxin Guan
- Shenzhen Institute of Translational Medicine, the First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Zhiyuan Zhou
- Department of Urology, Shanghai Pudong New Area GongLi Hospital, 219 Miaopu Road, Shanghai, 200135, China
| | - Wenhao Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Xuehao Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China.
| | - Chenyi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China.
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Wujin Road 85, Shanghai, 200080, China.
| |
Collapse
|
9
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
10
|
Lai H, Yip HC, Gong Y, Chan KF, Leung KKC, Chan MS, Xia X, Chiu PWY. MFGE8 in exosomes derived from mesenchymal stem cells prevents esophageal stricture after endoscopic submucosal dissection in pigs. J Nanobiotechnology 2024; 22:143. [PMID: 38561800 PMCID: PMC10986023 DOI: 10.1186/s12951-024-02429-0] [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: 12/24/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Endoscopic submucosal dissection (ESD) is the current standard treatment for early-stage esophageal neoplasms. However, the postoperative esophageal stricture after extensive mucosal dissection remains a severe challenge with limited effective treatments available. In this study, we introduced a chitosan/gelatin (ChGel) sponge encapsulating the adipose mesenchymal stem cells (ADMSCs)-derived exosomes (ChGelMSC-Exo) for the prevention of esophageal stenosis after ESD in a porcine model. RESULTS Pigs were randomly assigned into (1) ChGelMSC-Exo treatment group, (2) ChGelPBS group, and (3) the controls. Exosome treatments were applied immediately on the day after ESD as well as on day 7. Exosome components crucial for wound healing were investigated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and small RNA sequencing. ChGelMSC-Exo treatment significantly reduced mucosal contraction on day 21, with less fiber accumulation and inflammatory infiltration, and enhanced angiogenesis when compared with the control and ChGelPBS groups. The anti-fibrotic effects following MSC-Exo treatment were further found to be associated with the anti-inflammatory M2 polarization of the resident macrophages, especially within the M2b subset characterized by the reduced TGFβ1 secretion, which sufficiently inhibited inflammation and prevented the activation of myofibroblast with less collagen production at the early stage after ESD. Moreover, the abundant expression of exosomal MFGE8 was identified to be involved in the transition of the M2b-macrophage subset through the activation of MFGE8/STAT3/Arg1 axis. CONCLUSIONS Our study demonstrates that exosomal MFGE8 significantly promotes the polarization of the M2b-macrophage subset, consequently reducing collagen deposition. These findings suggest a promising potential for MSC-Exo therapy in preventing the development of esophageal stricture after near-circumferential ESD.
Collapse
Affiliation(s)
- Huasheng Lai
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, People's Republic of China
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
| | - Hon-Chi Yip
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
| | - Yu Gong
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Kai-Fung Chan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
| | - Kevin Kai-Chung Leung
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
| | - Melissa Shannon Chan
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China
| | - Xianfeng Xia
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China.
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China.
| | - Philip Wai-Yan Chiu
- Department of Surgery and State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China.
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, People's Republic of China.
| |
Collapse
|
11
|
Cai J, Lian C, Lu Z, Shang Q, Wang L, Han Z, Gu Y. FGF19-Based Mini Probe Targeting FGFR4 for Diagnosis and Surgical Navigation of Hepatocellular Carcinoma. J Med Chem 2024; 67:3764-3777. [PMID: 38385325 DOI: 10.1021/acs.jmedchem.3c02198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Hepatocellular carcinoma (HCC) is a frequent malignancy that has a high death rate and a high rate of recurrence following surgery, owing to insufficient surgical resection. Furthermore, HCC is prone to peritoneal metastasis (HCC-PM), resulting in a significant number of tiny cancer lesions, making surgical removal more challenging. As a potential imaging target, FGFR4 is highly expressed in tumors, especially in HCC, but is less expressed in the normal liver. In this study, we used computational simulation approaches to develop peptide I0 derived from FGF19, a particular ligand of FGFR4, and labeled it with the NIRF dye, MPA, for HCC detection. In surgical navigation, the TBR was 9.31 ± 1.36 and 8.57 ± 1.15 in HepG2 in situ tumor and HCC-PM models, respectively, indicating considerable tumor uptake. As a result, peptide I0 is an excellent clinical diagnostic reagent for HCC, as well as a tool for surgically resecting HCC peritoneal metastases.
Collapse
Affiliation(s)
- Jiaxian Cai
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Lian
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Zeyu Lu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Qian Shang
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Li Wang
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering and Diagnostic Pharmacy, School of engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
12
|
Qin Y, Ge G, Yang P, Wang L, Qiao Y, Pan G, Yang H, Bai J, Cui W, Geng D. An Update on Adipose-Derived Stem Cells for Regenerative Medicine: Where Challenge Meets Opportunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207334. [PMID: 37162248 PMCID: PMC10369252 DOI: 10.1002/advs.202207334] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/24/2023] [Indexed: 05/11/2023]
Abstract
Over the last decade, adipose-derived stem cells (ADSCs) have attracted increasing attention in the field of regenerative medicine. ADSCs appear to be the most advantageous cell type for regenerative therapies owing to their easy accessibility, multipotency, and active paracrine activity. This review highlights current challenges in translating ADSC-based therapies into clinical settings and discusses novel strategies to overcome the limitations of ADSCs. To further establish ADSC-based therapies as an emerging platform for regenerative medicine, this review also provides an update on the advancements in this field, including fat grafting, wound healing, bone regeneration, skeletal muscle repair, tendon reconstruction, cartilage regeneration, cardiac repair, and nerve regeneration. ADSC-based therapies are expected to be more tissue-specific and increasingly important in regenerative medicine.
Collapse
Affiliation(s)
- Yi Qin
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Gaoran Ge
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Peng Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Liangliang Wang
- Department of OrthopaedicsThe Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical UniversityChangzhouJiangsu213000China
| | - Yusen Qiao
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Guoqing Pan
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangJiangsu212013China
| | - Huilin Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Jiaxiang Bai
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Dechun Geng
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversityOrthopaedic Institute, Medical CollegeSoochow UniversitySuzhouJiangsu215006China
| |
Collapse
|
13
|
Wang Y, Zhu L, Wei L, Zhou Y, Yang Y, Zhang L. A bio-orthogonally functionalized chitosan scaffold with esterase-activatable release for nerve regeneration. Int J Biol Macromol 2023; 229:146-157. [PMID: 36528149 DOI: 10.1016/j.ijbiomac.2022.12.113] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/04/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Developing nerve conduits with biological cues is a promising approach for repairing peripheral nerve injuries. Although most biological cues incorporated into conduits generally exert their biological functions at the surface, they could not be released into the on-demand regeneration sites under physiological conditions. Herein, we firstly report a bio-orthogonally functionalized chitosan scaffold with esterase-activatable release for peripheral nerve regeneration. In this study, biological cues are not only selectively conjugated into nerve conduits by bio-orthogonal reaction, but also precisely released in on-demand regeneration sites via esterase-activatable cleavage for peripheral nerve repair. Moreover, this nerve scaffold with esterase-activatable release could promote Schwann cells proliferation. In a rat sciatic nerve defect model, the bio-orthogonally functionalized scaffold with esterase-activatable release significantly increased sciatic nerve function recovery and improved target muscles weight. This strategy of incorporating esterase-activatable bioactive cues into peripheral nerve conduits offers great potential in preclinical studies.
Collapse
Affiliation(s)
- Yuqing Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Linglin Zhu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Le Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Youlang Zhou
- Hand Surgery Research Center, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, Jiangsu, PR China.
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, Jiangsu, PR China.
| |
Collapse
|
14
|
Yadav A, Ramasamy TS, Lin SC, Chen SH, Lu J, Liu YH, Lu FI, Hsueh YY, Lin SP, Wu CC. Autologous Platelet-Rich Growth Factor Reduces M1 Macrophages and Modulates Inflammatory Microenvironments to Promote Sciatic Nerve Regeneration. Biomedicines 2022; 10:biomedicines10081991. [PMID: 36009539 PMCID: PMC9406033 DOI: 10.3390/biomedicines10081991] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
The failure of peripheral nerve regeneration is often associated with the inability to generate a permissive molecular and cellular microenvironment for nerve repair. Autologous therapies, such as platelet-rich plasma (PRP) or its derivative platelet-rich growth factors (PRGF), may improve peripheral nerve regeneration via unknown mechanistic roles and actions in macrophage polarization. In the current study, we hypothesize that excessive and prolonged inflammation might result in the failure of pro-inflammatory M1 macrophage transit to anti-inflammatory M2 macrophages in large nerve defects. PRGF was used in vitro at the time the unpolarized macrophages (M0) macrophages were induced to M1 macrophages to observe if PRGF altered the secretion of cytokines and resulted in a phenotypic change. PRGF was also employed in the nerve conduit of a rat sciatic nerve transection model to identify alterations in macrophages that might influence excessive inflammation and nerve regeneration. PRGF administration reduced the mRNA expression of tumor necrosis factor-α (TNFα), interleukin-1β (IL-1β), and IL-6 in M0 macrophages. Increased CD206 substantiated the shift of pro-inflammatory cytokines to the M2 regenerative macrophage. Administration of PRGF in the nerve conduit after rat sciatic nerve transection promoted nerve regeneration by improving nerve gross morphology and its targeted gastrocnemius muscle mass. The regenerative markers were increased for regrown axons (protein gene product, PGP9.5), Schwann cells (S100β), and myelin basic protein (MBP) after 6 weeks of injury. The decreased expression of TNFα, IL-1β, IL-6, and CD68+ M1 macrophages indicated that the inflammatory microenvironments were reduced in the PRGF-treated nerve tissue. The increase in RECA-positive cells suggested the PRGF also promoted angiogenesis during nerve regeneration. Taken together, these results indicate the potential role and clinical implication of autologous PRGF in regulating inflammatory microenvironments via macrophage polarization after nerve transection.
Collapse
Affiliation(s)
- Anjali Yadav
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 701, Taiwan
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sheng-Che Lin
- Division of Plastic and Reconstructive Surgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan 709, Taiwan
| | - Szu-Han Chen
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 701, Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan 701, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Jean Lu
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Ya-Hsin Liu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Fu-I Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Yuan-Yu Hsueh
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 701, Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan 701, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei 106, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 701, Taiwan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: ; Tel.: +886-6-235-3535 (ext. 5327); Fax: +886-6-209-3007
| |
Collapse
|
15
|
Zhang Q, Burrell JC, Zeng J, Motiwala FI, Shi S, Cullen DK, Le AD. Implantation of a nerve protector embedded with human GMSC-derived Schwann-like cells accelerates regeneration of crush-injured rat sciatic nerves. Stem Cell Res Ther 2022; 13:263. [PMID: 35725660 PMCID: PMC9208168 DOI: 10.1186/s13287-022-02947-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/08/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Peripheral nerve injuries (PNIs) remain one of the great clinical challenges because of their considerable long-term disability potential. Postnatal neural crest-derived multipotent stem cells, including gingiva-derived mesenchymal stem cells (GMSCs), represent a promising source of seed cells for tissue engineering and regenerative therapy of various disorders, including PNIs. Here, we generated GMSC-repopulated nerve protectors and evaluated their therapeutic effects in a crush injury model of rat sciatic nerves. METHODS GMSCs were mixed in methacrylated collagen and cultured for 48 h, allowing the conversion of GMSCs into Schwann-like cells (GiSCs). The phenotype of GiSCs was verified by fluorescence studies on the expression of Schwann cell markers. GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were co-cultured with THP-1-derived macrophages, and the secretion of anti-inflammatory cytokine IL-10 or inflammatory cytokines TNF-α and IL-1β in the supernatant was determined by ELISA. In addition, GMSCs mixed in the methacrylated collagen were filled into a nerve protector made from the decellularized small intestine submucosal extracellular matrix (SIS-ECM) and cultured for 24 h, allowing the generation of functionalized nerve protectors repopulated with GiSCs. We implanted the nerve protector to wrap the injury site of rat sciatic nerves and performed functional and histological assessments 4 weeks post-surgery. RESULTS GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were directly converted into Schwann-like cells (GiSCs) characterized by the expression of S-100β, p75NTR, BDNF, and GDNF. In vitro, co-culture of GMSCs encapsulated in the 3D-collagen hydrogel with macrophages remarkably increased the secretion of IL-10, an anti-inflammatory cytokine characteristic of pro-regenerative (M2) macrophages, but robustly reduced LPS-stimulated secretion of TNF-1α and IL-1β, two cytokines characteristic of pro-inflammatory (M1) macrophages. In addition, our results indicate that implantation of functionalized nerve protectors repopulated with GiSCs significantly accelerated functional recovery and axonal regeneration of crush-injured rat sciatic nerves accompanied by increased infiltration of pro-regenerative (M2) macrophages while a decreased infiltration of pro-inflammatory (M1) macrophages. CONCLUSIONS Collectively, these findings suggest that Schwann-like cells converted from GMSCs represent a promising source of supportive cells for regenerative therapy of PNI through their dual functions, neurotrophic effects, and immunomodulation of pro-inflammatory (M1)/pro-regenerative (M2) macrophages.
Collapse
Affiliation(s)
- Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA.
| | - Justin C. Burrell
- grid.25879.310000 0004 1936 8972Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA ,grid.410355.60000 0004 0420 350XCenter for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Jincheng Zeng
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA ,grid.410560.60000 0004 1760 3078Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808 China
| | - Faizan I. Motiwala
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA
| | - Shihong Shi
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA
| | - D. Kacy Cullen
- grid.25879.310000 0004 1936 8972Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA ,grid.410355.60000 0004 0420 350XCenter for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Anh D. Le
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA ,grid.411115.10000 0004 0435 0884Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| |
Collapse
|
16
|
Su Q, Nasser MI, He J, Deng G, Ouyang Q, Zhuang D, Deng Y, Hu H, Liu N, Li Z, Zhu P, Li G. Engineered Schwann Cell-Based Therapies for Injury Peripheral Nerve Reconstruction. Front Cell Neurosci 2022; 16:865266. [PMID: 35602558 PMCID: PMC9120533 DOI: 10.3389/fncel.2022.865266] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Compared with the central nervous system, the adult peripheral nervous system possesses a remarkable regenerative capacity, which is due to the strong plasticity of Schwann cells (SCs) in peripheral nerves. After peripheral nervous injury, SCs de-differentiate and transform into repair phenotypes, and play a critical role in axonal regeneration, myelin formation, and clearance of axonal and myelin debris. In view of the limited self-repair capability of SCs for long segment defects of peripheral nerve defects, it is of great clinical value to supplement SCs in necrotic areas through gene modification or stem cell transplantation or to construct tissue-engineered nerve combined with bioactive scaffolds to repair such tissue defects. Based on the developmental lineage of SCs and the gene regulation network after peripheral nerve injury (PNI), this review summarizes the possibility of using SCs constructed by the latest gene modification technology to repair PNI. The therapeutic effects of tissue-engineered nerve constructed by materials combined with Schwann cells resembles autologous transplantation, which is the gold standard for PNI repair. Therefore, this review generalizes the research progress of biomaterials combined with Schwann cells for PNI repair. Based on the difficulty of donor sources, this review also discusses the potential of “unlimited” provision of pluripotent stem cells capable of directing differentiation or transforming existing somatic cells into induced SCs. The summary of these concepts and therapeutic strategies makes it possible for SCs to be used more effectively in the repair of PNI.
Collapse
Affiliation(s)
- Qisong Su
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Moussa Ide Nasser
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Jiaming He
- School of Basic Medical Science, Shandong University, Jinan, China
| | - Gang Deng
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Qing Ouyang
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Donglin Zhuang
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuzhi Deng
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Haoyun Hu
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Nanbo Liu
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zhetao Li
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ping Zhu
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Structural Heart Disease, Guangzhou, China
- *Correspondence: Ping Zhu,
| | - Ge Li
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Structural Heart Disease, Guangzhou, China
- Ge Li,
| |
Collapse
|
17
|
Liu T, Wang Y, Lu L, Liu Y. SPIONs mediated magnetic actuation promotes nerve regeneration by inducing and maintaining repair-supportive phenotypes in Schwann cells. J Nanobiotechnology 2022; 20:159. [PMID: 35351151 PMCID: PMC8966266 DOI: 10.1186/s12951-022-01337-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/26/2022] [Indexed: 12/18/2022] Open
Abstract
Background Schwann cells, the glial cells in the peripheral nervous system, are highly plastic. In response to nerve injury, Schwann cells are reprogrammed to a series of specialized repair-promoting phenotypes, known as repair Schwann cells, which play a pivotal role in nerve regeneration. However, repair Schwann cells represent a transient and unstable cell state, and these cells progressively lose their repair phenotypes and repair‐supportive capacity; the transience of this state is one of the key reasons for regeneration failure in humans. Therefore, the ability to control the phenotypic stability of repair Schwann cells is of great practical importance as well as biological interest. Results We designed and prepared a type of fluorescent–magnetic bifunctional superparamagnetic iron oxide nanoparticles (SPIONs). In the present study, we established rat sciatic nerve injury models, then applied SPIONs to Schwann cells and established an effective SPION-mediated magnetic actuation system targeting the sciatic nerves. Our results demonstrate that magnetic actuation mediated by SPIONs can induce and maintain repair-supportive phenotypes of Schwann cells, thereby promoting regeneration and functional recovery of the sciatic nerve after crush injury. Conclusions Our research indicate that Schwann cells can sense these external, magnetically driven mechanical forces and transduce them to intracellular biochemical signals that promote nerve regeneration by inducing and maintaining the repair phenotypes of Schwann cells. We hope that this study will provide a new therapeutic strategy to promote the regeneration and repair of injured peripheral nerves. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01337-5.
Collapse
Affiliation(s)
- Ting Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yang Wang
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, People's Republic of China.
| |
Collapse
|
18
|
Xu Z, Cai Y, Liu W, Kang F, He Q, Hong Q, Zhang W, Li J, Yan Y, Peng J. Downregulated exosome-associated gene FGF9 as a novel diagnostic and prognostic target for ovarian cancer and its underlying roles in immune regulation. Aging (Albany NY) 2022; 14:1822-1835. [PMID: 35190498 PMCID: PMC8908935 DOI: 10.18632/aging.203905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023]
Abstract
Exosome has been demonstrated to be secreted from cells and seized by targeted cells. Exosome could transmit signals and exert biological functions in cancer progression. Nevertheless, the underlying mechanisms of exosome in ovarian cancer (OC) have not been fully explored. In this study, we wanted to explore whether Fibroblast growth factor 9 (FGF9), as an exosome-associated gene, was importantly essential in OC progression and prognosis. Firstly, comprehensive bioinformatics platforms were applied to find that FGF9 expression was lower in OC tissues compared to normal ovarian tissues. Meanwhile, downregulated FGF9 displayed favorable prognostic values in OC patients. The gene enrichment of biological functions indicated that abnormally expressed FGF9 could be involved in the OC-related immune signatures, such as immunoinhibitors and chemokine receptors. Taken together, these findings could provide a novel insight into the significance of FGF9 in OC progress and supply a new destination of FGF9-related immunotherapy in clinical treatment.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuan Cai
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wei Liu
- Department of Orthopedic Surgery, The Second Hospital University of South China, Hengyang 421001, Hunan, China
| | - Fanhua Kang
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
| | - Qingchun He
- Department of Emergency, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Emergency, Xiangya Changde Hospital, Changde 415000, Hunan, China
| | - Qianhui Hong
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
| | - Wenqin Zhang
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
| | - Jianbo Li
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, Hunan, China
| |
Collapse
|
19
|
Prospects of Cordycepin and Polysaccharides Produced by Cordyceps. Fungal Biol 2022. [DOI: 10.1007/978-981-16-8877-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Yuan X, Li L, Liu H, Luo J, Zhao Y, Pan C, Zhang X, Chen Y, Gou M. Strategies for improving adipose-derived stem cells for tissue regeneration. BURNS & TRAUMA 2022; 10:tkac028. [PMID: 35992369 PMCID: PMC9382096 DOI: 10.1093/burnst/tkac028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/27/2022] [Indexed: 11/13/2022]
Abstract
Abstract
Adipose-derived stem cells (ADSCs) have promising applications in tissue regeneration. Currently, there are only a few ADSC products that have been approved for clinical use. The clinical application of ADSCs still faces many challenges. Here, we review emerging strategies to improve the therapeutic efficacy of ADSCs in tissue regeneration. First, a great quantity of cells is often needed for the stem cell therapies, which requires the advanced cell expansion technologies. In addition cell-derived products are also required for the development of ‘cell-free’ therapies to overcome the drawbacks of cell-based therapies. Second, it is necessary to strengthen the regenerative functions of ADSCs, including viability, differentiation and paracrine ability, for the tissue repair and regeneration required for different physiological and pathophysiological conditions. Third, poor delivery efficiency also restricts the therapeutic effect of ADSCs. Effective methods to improve cell delivery include alleviating harsh microenvironments, enhancing targeting ability and prolonging cell retention. Moreover, we also point out some critical issues about the sources, effectiveness and safety of ADSCs. With these advanced strategies to improve the therapeutic efficacy of ADSCs, ADSC-based treatment holds great promise for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Xin Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Li Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Haofan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Jing Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yongchao Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Cheng Pan
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Xue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yuwen Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| |
Collapse
|
21
|
Yadav A, Huang TC, Chen SH, Ramasamy TS, Hsueh YY, Lin SP, Lu FI, Liu YH, Wu CC. Sodium phenylbutyrate inhibits Schwann cell inflammation via HDAC and NFκB to promote axonal regeneration and remyelination. J Neuroinflammation 2021; 18:238. [PMID: 34656124 PMCID: PMC8520633 DOI: 10.1186/s12974-021-02273-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/08/2021] [Indexed: 11/24/2022] Open
Abstract
Background Epigenetic regulation by histone deacetylases (HDACs) in Schwann cells (SCs) after injury facilitates them to undergo de- and redifferentiation processes necessary to support various stages of nerve repair. Although de-differentiation activates the synthesis and secretion of inflammatory cytokines by SCs to initiate an immune response during nerve repair, changes in either the timing or duration of prolonged inflammation mediated by SCs can affect later processes associated with repair and regeneration. Limited studies have investigated the regulatory processes through which HDACs in SCs control inflammatory cytokines to provide a favorable environment for peripheral nerve regeneration. Methods We employed the HDAC inhibitor (HDACi) sodium phenylbutyrate (PBA) to address this question in an in vitro RT4 SC inflammation model and an in vivo sciatic nerve transection injury model to examine the effects of HDAC inhibition on the expression of pro-inflammatory cytokines. Furthermore, we assessed the outcomes of suppression of extended inflammation on the regenerative potential of nerves by assessing axonal regeneration, remyelination, and reinnervation. Results Significant reductions in lipopolysaccharide (LPS)-induced pro-inflammatory cytokine (tumor necrosis factor-α [TNFα]) expression and secretion were observed in vitro following PBA treatment. PBA treatment also affected the transient changes in nuclear factor κB (NFκB)-p65 phosphorylation and translocation in response to LPS induction in RT4 SCs. Similarly, PBA mediated long-term suppressive effects on HDAC3 expression and activity. PBA administration resulted in marked inhibition of pro-inflammatory cytokine secretion at the site of transection injury when compared with that in the hydrogel control group at 6-week post-injury. A conducive microenvironment for axonal regrowth and remyelination was generated by increasing expression levels of protein gene product 9.5 (PGP9.5) and myelin basic protein (MBP) in regenerating nerve tissues. PBA administration increased the relative gastrocnemius muscle weight percentage and maintained the intactness of muscle bundles when compared with those in the hydrogel control group. Conclusions Suppressing the lengthened state of inflammation using PBA treatment favors axonal regrowth and remyelination following nerve transection injury. PBA treatment also regulates pro-inflammatory cytokine expression by inhibiting the transcriptional activation of NFκB-p65 and HDAC3 in SCs in vitro. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02273-1.
Collapse
Affiliation(s)
- Anjali Yadav
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chieh Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Szu-Han Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Division of Plastic and Reconstructive Surgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yuan-Yu Hsueh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Division of Plastic and Reconstructive Surgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Fu-I Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Hsin Liu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan. .,Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Chen Z, Shen G, Tan X, Qu L, Zhang C, Ma L, Luo P, Cao X, Yang F, Liu Y, Wang Y, Shi C. ID1/ID3 mediate the contribution of skin fibroblasts to local nerve regeneration through Itga6 in wound repair. Stem Cells Transl Med 2021; 10:1637-1649. [PMID: 34520124 PMCID: PMC8641086 DOI: 10.1002/sctm.21-0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/28/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022] Open
Abstract
Cutaneous wound healing requires intricate synchronization of several key processes. Among them, local nerve regeneration is known to be vitally important for proper repair. However, the underlying mechanisms of local nerve regeneration are still unclear. Fibroblasts are one of the key cell types within the skin whose role in local nerve regeneration has not been extensively studied. In our study, we found skin fibroblasts were in tight contact with regenerated nerves during wound healing, while rare interactions were shown under normal circumstances. Moreover, skin fibroblasts surrounding the nerves were shown to be activated and reprogrammed to exhibit neural cell‐like properties by upregulated expressing inhibitor of DNA binding 1 (ID1) and ID3. Furthermore, we identified the regulation of integrin α6 (Itga6) by ID1/ID3 in fibroblasts as the mechanism for axon guidance. Accordingly, transplantation of the ID1/ID3‐overexpressing fibroblasts or topical injection of ID1/ID3 lentivirus significantly promoted local nerve regeneration and wound healing following skin excision or sciatic nerve injury. Therefore, we demonstrated a new role for skin fibroblasts in nerve regeneration following local injury by directly contacting and guiding axon regrowth, which might hold therapeutic potential in peripheral nerve disorders and peripheral neuropathies in relatively chronic refractory wounds.
Collapse
Affiliation(s)
- Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Gufang Shen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Langfan Qu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Can Zhang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Le Ma
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Xiaohui Cao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Fan Yang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Yunsheng Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
23
|
Balakrishnan A, Belfiore L, Chu TH, Fleming T, Midha R, Biernaskie J, Schuurmans C. Insights Into the Role and Potential of Schwann Cells for Peripheral Nerve Repair From Studies of Development and Injury. Front Mol Neurosci 2021; 13:608442. [PMID: 33568974 PMCID: PMC7868393 DOI: 10.3389/fnmol.2020.608442] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries arising from trauma or disease can lead to sensory and motor deficits and neuropathic pain. Despite the purported ability of the peripheral nerve to self-repair, lifelong disability is common. New molecular and cellular insights have begun to reveal why the peripheral nerve has limited repair capacity. The peripheral nerve is primarily comprised of axons and Schwann cells, the supporting glial cells that produce myelin to facilitate the rapid conduction of electrical impulses. Schwann cells are required for successful nerve regeneration; they partially “de-differentiate” in response to injury, re-initiating the expression of developmental genes that support nerve repair. However, Schwann cell dysfunction, which occurs in chronic nerve injury, disease, and aging, limits their capacity to support endogenous repair, worsening patient outcomes. Cell replacement-based therapeutic approaches using exogenous Schwann cells could be curative, but not all Schwann cells have a “repair” phenotype, defined as the ability to promote axonal growth, maintain a proliferative phenotype, and remyelinate axons. Two cell replacement strategies are being championed for peripheral nerve repair: prospective isolation of “repair” Schwann cells for autologous cell transplants, which is hampered by supply challenges, and directed differentiation of pluripotent stem cells or lineage conversion of accessible somatic cells to induced Schwann cells, with the potential of “unlimited” supply. All approaches require a solid understanding of the molecular mechanisms guiding Schwann cell development and the repair phenotype, which we review herein. Together these studies provide essential context for current efforts to design glial cell-based therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren Belfiore
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak-Ho Chu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Taylor Fleming
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Anti-Cancer Effect of Cordycepin on FGF9-Induced Testicular Tumorigenesis. Int J Mol Sci 2020; 21:ijms21218336. [PMID: 33172093 PMCID: PMC7672634 DOI: 10.3390/ijms21218336] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cordycepin, a bioactive constituent from the fungus Cordyceps sinensis, could inhibit cancer cell proliferation and promote cell death via induction of cell cycle arrest, apoptosis and autophagy. Our novel finding from microarray analysis of cordycepin-treated MA-10 mouse Leydig tumor cells is that cordycepin down-regulated the mRNA levels of FGF9, FGF18, FGFR2 and FGFR3 genes in MA-10 cells. Meanwhile, the IPA-MAP pathway prediction result showed that cordycepin inhibited MA-10 cell proliferation by suppressing FGFs/FGFRs pathways. The in vitro study further revealed that cordycepin decreased FGF9-induced MA-10 cell proliferation by inhibiting the expressions of p-ERK1/2, p-Rb and E2F1, and subsequently reducing the expressions of cyclins and CDKs. In addition, a mouse allograft model was performed by intratumoral injection of FGF9 and/or intraperitoneal injection of cordycepin to MA-10-tumor bearing C57BL/6J mice. Results showed that FGF9-induced tumor growth in cordycepin-treated mice was significantly smaller than that in a PBS-treated control group. Furthermore, cordycepin decreased FGF9-induced FGFR1-4 protein expressions in vitro and in vivo. In summary, cordycepin inhibited FGF9-induced testicular tumor growth by suppressing the ERK1/2, Rb/E2F1, cell cycle pathways, and the expressions of FGFR1-4 proteins, suggesting that cordycepin can be used as a novel anticancer drug for testicular cancers.
Collapse
|
25
|
Liang Z, Huang D, Zhang M, Yi X, Wu F, Zhu D, Ning Y, Gan H, Li H. [ In vitro study on promoting migration ability of rat adipose derived stem cells modified by stromal cell-derived factor 1α]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1305-1312. [PMID: 33063498 DOI: 10.7507/1002-1892.202004134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explored the effect of stromal cell-derived factor 1α (SDF-1α) on promoting the migration ability of rat adipose derived stem cells (rADSCs) by constructed the rADSCs overexpression SDF-1α via adenovirus transfection. Methods rADSCs were isolated from adipose tissue of 6-week-old SPF Sprague Dawley rats. Morphological observation, multi-directional differentiations (osteogenic, adipogenic, and chondrogenic inductions), and flow cytometry identification were performed. Transwell cell migration experiment was used to observe and screen the optimal concentration of exogenous SDF-1α to optimize the migration ability of rADSCs; the optimal multiplicity of infection (MOI) of rADSCs was screened by observing the cell status and fluorescence expression after transfection. Then the third generation of rADSCs were divided into 4 groups: group A was pure rADSCs; group B was rADSCs co-cultured with SDF-1α at the best concentration; group C was rADSCs infected with recombinant adenovirus-mediated green fluorescent protein (Adv-GFP) with the best MOI; group D was rADSCs infected with Adv-GFP-SDF-1α overexpression adenovirus with the best MOI. Cell counting kit 8 (CCK-8) and Transwell cell migration experiment were preformed to detect and compare the effect of exogenous SDF-1α and SDF-1α overexpression on the proliferation and migration ability of rADSCs. Results The cell morphology, multi-directional differentiations, and flow cytometry identification showed that the cultured cells were rADSCs. After screening, the optimal stimulating concentration of exogenous SDF-1α was 12.5 nmol/L; the optimal MOI of Adv-GFP adenovirus was 200; the optimal MOI of Adv-GFP-SDF-1α overexpression adenovirus was 400. CCK-8 method and Transwell cell migration experiment showed that compared with groups A and C, groups B and D could significantly improve the proliferation and migration of rADSCs ( P<0.05); the effect of group D on enhancing the migration of rADSCs was weaker than that of group B, but the effect of promoting the proliferation of rADSCs was stronger than that of group D ( P<0.05). Conclusion SDF-1α overexpression modification on rADSCs can significantly promote the proliferation and migration ability, which may be a potential method to optimize the application of ADSCs in tissue regeneration and wound repair.
Collapse
Affiliation(s)
- Zhijie Liang
- Department of Wound Repair, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | | | - Muzi Zhang
- Department of Plastic Surgery, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Xiaolin Yi
- Department of Plastic Surgery, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Fangxiao Wu
- Department of Plastic Surgery, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Dandan Zhu
- Department of Wound Repair, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Yan Ning
- Department of Plastic Surgery, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Huimin Gan
- Department of Radiotherapy, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| | - Hongmian Li
- Department of Plastic Surgery, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530022, P.R.China
| |
Collapse
|
26
|
Xia B, Gao J, Li S, Huang L, Zhu L, Ma T, Zhao L, Yang Y, Luo K, Shi X, Mei L, Zhang H, Zheng Y, Lu L, Luo Z, Huang J. Mechanical stimulation of Schwann cells promote peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b-3p. Theranostics 2020; 10:8974-8995. [PMID: 32802175 PMCID: PMC7415818 DOI: 10.7150/thno.44912] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Peripheral nerves are unique in their remarkable elasticity. Schwann cells (SCs), important components of the peripheral nervous system (PNS), are constantly subjected to physiological and mechanical stresses from dynamic stretching and compression forces during movement. So far, it is not clear if SCs sense and respond to mechanical signals. It is also unknown whether mechanical stimuli can interfere with the intercellular communications between neurons and SCs, and what role extracellular vesicles (EVs) play in this process. The present study aimed to examine the effect of mechanical stimuli on the EV-mediated intercellular communication between neurons and SCs, explore their effect on axonal regeneration, and investigate the underlying mechanism. Methods: Purified SCs were stimulated using a magnetic force-based mechanical stimulation (MS) system and EVs were purified from mechanically stimulated SCs (MS-SCs-EVs) and non-stimulated SCs (SCs-EVs). The effect of MS-SCs-EVs on axonal elongation was examined in vitro and in vivo. High throughput miRNA sequencing was performed to compare the differential miRNA profiles between MS-SCs-EVs and SCs-EVs. The functional role of differentially expressed miRNAs on neurite extension in MS-SCs-EVs was examined. Also, the putative target genes of differentially expressed miRNAs in MS-SCs-EVs were predicted by bioinformatics tools, and the regulatory effect of those miRNAs on putative target genes was validated both in vitro and in vivo. Results: The MS-SCs-EVs showed an average size of 137.52±1.77 nm, and could be internalized by dorsal root ganglion (DRG) neurons. Compared to SCs-EVs, MS-SCs-EVs showed a stronger ability to enhance neurite outgrowth in vitro and nerve regeneration in vivo. High throughput miRNA sequencing identified a number of differentially expressed miRNAs in MS-SCs-EVs. Further analysis of those EV-miRNAs demonstrated that miR-23b-3p played a predominant role in MS-SCs-EVs since its deprivation abolished their enhanced axonal elongation. Furthermore, we identified neuropilin 1 (Nrp1) in neurons as the target gene of miR-23b-3p in MS-SCs-EVs. This observation was supported by the evidence that miR-23b-3p could decrease Nrp1-3'-UTR-WT luciferase activity in vitro and down-regulate Nrp1 expression in neurons. Conclusion: Our findings suggested that mechanical stimuli are capable of modulating the intercellular communication between neurons and SCs by altering miRNA composition in MS-SCs-EVs. Transfer of miR-23b-3p by MS-SCs-EVs from mechanically stimulated SCs to neurons decreased neuronal Nrp1 expression, which was responsible, at least in part, for the beneficial effect of MS-SCs-EVs on axonal regeneration. Our results highlighted the potential therapeutic value of MS-SCs-EVs and miR-23b-3p-enriched EVs in peripheral nerve injury repair.
Collapse
Affiliation(s)
- Bing Xia
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianbo Gao
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Shengyou Li
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liangliang Huang
- Department of Orthopedics, the General Hospital of Central Theater Command of People's Liberation Army, Wuhan, 430070, People's Republic of China
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital Affiliated to Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710054, People's Republic of China
| | - Teng Ma
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Laihe Zhao
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yujie Yang
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Kai Luo
- Department of Orthopedics, the 985th Hospital People's Liberation Army Joint Logistics Support Force, Taiyuan, 030000, People's Republic of China
| | - Xiaowei Shi
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liangwei Mei
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Hao Zhang
- Department of Spinal Surgery, the People's Hospital of Longhua District, Shenzhen, 518109, People's Republic of China
| | - Yi Zheng
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Lei Lu
- Department of Oral Anatomy and Physiology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Zhuojing Luo
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jinghui Huang
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| |
Collapse
|