1
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Aisaiti A, Aierxiding S, Shoukeer K, Muheremu A. Mesenchymal stem cells for peripheral nerve injury and regeneration: a bibliometric and visualization study. Front Neurol 2024; 15:1420402. [PMID: 39161869 PMCID: PMC11330774 DOI: 10.3389/fneur.2024.1420402] [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: 04/20/2024] [Accepted: 06/10/2024] [Indexed: 08/21/2024] Open
Abstract
Objective To use bibliometric methods to analyze the research hotspots and future development trends regarding the application of mesenchymal stem cells in peripheral nerve injury and regeneration. Methods Articles published from January 1, 2013, to December 31, 2023, were meticulously screened using the MeSH terms: TS = ("Mesenchymal stem cells" AND "Peripheral nerve injury") OR TS = ("Mesenchymal stem cells" AND "Peripheral nerve regeneration") within the Web of Science database. The compiled data was then subjected to in-depth analysis with the aid of VOSviewer and Cite Space software, which facilitated the identification of the most productive countries, organizations, authors, and the predominant keywords prevalent within this research domain. Results An extensive search of the Web of Science database yielded 350 relevant publications. These scholarly works were authored by 2,049 collaborative researchers representing 41 countries and affiliated with 585 diverse academic and research institutions. The findings from this research were disseminated across 167 various journals, and the publications collectively cited 21,064 references from 3,339 distinct journals. Conclusion Over the past decade, there has been a consistent upward trajectory in the number of publications and citations pertaining to the use of mesenchymal stem cells in the realm of peripheral nerve injury and regeneration. The domain of stem cell therapy for nerve injury has emerged as a prime focus of research, with mesenchymal stem cell therapy taking center stage due to its considerable promise in the treatment of nerve injuries. This therapeutic approach holds the potential to significantly enhance treatment options and rehabilitation prospects for patients suffering from such injuries.
Collapse
Affiliation(s)
- Aikebaierjiang Aisaiti
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Shalayiding Aierxiding
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Kutiluke Shoukeer
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Aikeremujiang Muheremu
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
- Beijing Darwin Cell Biotechnology Co., Ltd., Beijing, China
| |
Collapse
|
3
|
Li X, Xu H, Li C, Guan Y, Liu Y, Zhang T, Meng F, Cheng H, Song X, Jia Z, He R, Zhao J, Chen S, Guan C, Yan S, Wang J, Wei Y, Zhang J, Tang J, Peng J, Wang Y. Biological characteristics of tissue engineered-nerve grafts enhancing peripheral nerve regeneration. Stem Cell Res Ther 2024; 15:215. [PMID: 39020413 PMCID: PMC11256578 DOI: 10.1186/s13287-024-03827-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND A favorable regenerative microenvironment is essential for peripheral nerve regeneration. Neural tissue-specific extracellular matrix (ECM) is a natural material that helps direct cell behavior and promote axon regeneration. Both bone marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived mesenchymal stem cells (ADSCs) transplantation are effective in repairing peripheral nerve injury (PNI). However, there is no study that characterizes the in vivo microenvironmental characteristics of these two MSCs for the early repair of PNI when combined with neural tissue-derived ECM materials, i.e., acellular nerve allograft (ANA). METHODS In order to investigate biological characteristics, molecular mechanisms of early stage, and effectiveness of ADSCs- or BMSCs-injected into ANA for repairing PNI in vivo, a rat 10 mm long sciatic nerve defect model was used. We isolated primary BMSCs and ADSCs from bone marrow and adipose tissue, respectively. First, to investigate the in vivo response characteristics and underlying molecular mechanisms of ANA combined with BMSCs or ADSCs, eighty-four rats were randomly divided into three groups: ANA group, ANA+BMSC group, and ANA+ADSC group. We performed flow cytometry, RT-PCR, and immunofluorescence staining up to 4 weeks postoperatively. To further elucidate the underlying molecular mechanisms, changes in long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs) were systematically investigated using whole transcriptome sequencing. We then constructed protein-protein interaction networks to find 10 top ranked hub genes among differentially expressed mRNAs. Second, in order to explore the effectiveness of BMSCs and ADSCs on neural tissue-derived ECM materials for repairing PNI, sixty-eight rats were randomized into four groups: ANA group, ANA+BMSC group, ANA+ADSC group, and AUTO group. In the ANA+BMSC and ANA+ADSC groups, ADSCs/BMSCs were equally injected along the long axis of the 10-mm ANA. Then, we performed histological and functional assessments up to 12 weeks postoperatively. RESULTS The results of flow cytometry and RT-PCR showed that ANA combined with BMSCs exhibited more significant immunomodulatory effects, as evidenced by the up-regulation of interleukin (IL)-10, down-regulation of IL-1β and tumor necrosis factor-alpha (TNF-α) expression, promotion of M1-type macrophage polarization to M2-type, and a significant increase in the number of regulatory T cells (Tregs). ANA combined with ADSCs exhibited more pronounced features of pro-myelination and angiogenesis, as evidenced by the up-regulation of myelin-associated protein gene (MBP and MPZ) and angiogenesis-related factors (TGF-β, VEGF). Moreover, differentially expressed genes from whole transcriptome sequencing results further indicated that ANA loaded with BMSCs exhibited notable immunomodulatory effects and ANA loaded with ADSCs was more associated with angiogenesis, axonal growth, and myelin formation. Notably, ANA infused with BMSCs or ADSCs enhanced peripheral nerve regeneration and motor function recovery with no statistically significant differences. CONCLUSIONS This study revealed that both ANA combined with BMSCs and ADSCs enhance peripheral nerve regeneration and motor function recovery, but their biological characteristics (mainly including immunomodulatory effects, pro-vascular regenerative effects, and pro-myelin regenerative effects) and underlying molecular mechanisms in the process of repairing PNI in vivo are different, providing new insights into MSC therapy for peripheral nerve injury and its clinical translation.
Collapse
Affiliation(s)
- Xiangling Li
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Hang Xu
- Department of General Surgery, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yuli Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Haofeng Cheng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiangyu Song
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Zhibo Jia
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Ruichao He
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinjuan Zhao
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Congcong Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shi Yan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinpeng Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yu Wei
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jian Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jinshu Tang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
| | - Jiang Peng
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
| | - Yu Wang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
4
|
Li Q, Zhang F, Fu X, Han N. Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes as Nanomedicine for Peripheral Nerve Injury. Int J Mol Sci 2024; 25:7882. [PMID: 39063125 PMCID: PMC11277195 DOI: 10.3390/ijms25147882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Peripheral nerve injury (PNI) is a complex and protracted process, and existing therapeutic approaches struggle to achieve effective nerve regeneration. Recent studies have shown that mesenchymal stem cells (MSCs) may be a pivotal choice for treating peripheral nerve injury. MSCs possess robust paracrine capabilities, and exosomes, as the primary secretome of MSCs, are considered crucial regulatory mediators involved in peripheral nerve regeneration. Exosomes, as nanocarriers, can transport various endogenous or exogenous bioactive substances to recipient cells, thereby promoting vascular and axonal regeneration while suppressing inflammation and pain. In this review, we summarize the mechanistic roles of exosomes derived from MSCs in peripheral nerve regeneration, discuss the engineering strategies for MSC-derived exosomes to improve therapeutic potential, and explore the combined effects of MSC-derived exosomes with biomaterials (nerve conduits, hydrogels) in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Qicheng Li
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Fengshi Zhang
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Xiaoyang Fu
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Na Han
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| |
Collapse
|
5
|
Zhu Y, Yi D, Wang J, Zhang Y, Li M, Ma J, Ji Y, Peng J, Wang Y, Luo Y. Harnessing three-dimensional porous chitosan microsphere embedded with adipose-derived stem cells to promote nerve regeneration. Stem Cell Res Ther 2024; 15:158. [PMID: 38824568 PMCID: PMC11144330 DOI: 10.1186/s13287-024-03753-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/05/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Nerve guide conduits are a promising strategy for reconstructing peripheral nerve defects. Improving the survival rate of seed cells in nerve conduits is still a challenge and microcarriers are an excellent three-dimensional (3D) culture scaffold. Here, we investigate the effect of the 3D culture of microcarriers on the biological characteristics of adipose mesenchymal stem cells (ADSCs) and to evaluate the efficacy of chitosan nerve conduits filled with microcarriers loaded with ADSCs in repairing nerve defects. METHODS In vitro, we prepared porous chitosan microspheres by a modified emulsion cross-linking method for loading ADSCs and evaluated the growth status and function of ADSCs. In vivo, ADSCs-loaded microcarriers were injected into chitosan nerve conduits to repair a 12 mm sciatic nerve defect in rats. RESULTS Compared to the conventional two-dimensional (2D) culture, the prepared microcarriers were more conducive to the proliferation, migration, and secretion of trophic factors of ADSCs. In addition, gait analysis, neuro-electrophysiology, and histological evaluation of nerves and muscles showed that the ADSC microcarrier-loaded nerve conduits were more effective in improving nerve regeneration. CONCLUSIONS The ADSCs-loaded chitosan porous microcarrier prepared in this study has a high cell engraftment rate and good potential for peripheral nerve repair.
Collapse
Affiliation(s)
- Yaqiong Zhu
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Chinese PLA General Hospital, Beijing, China
- Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
| | - Dan Yi
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jing Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui Province, China
| | - Yongyi Zhang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Rehabilitation Medicine, the Second Medical Centre, Chinese PLA General Hospital, Beijing, China
- No.962 Hospital of the PLA Joint Logistic Support Force, Harbin, China
| | - Molin Li
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jun Ma
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yongjiao Ji
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Chinese PLA General Hospital, Beijing, China.
- Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China.
- Department of Orthopaedics, The Fourth Centre of Chinese PLA General Hospital, Beijing, China.
| | - Yuexiang Wang
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yukun Luo
- Department of Ultrasound, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
6
|
Tang H, Li J, Wang H, Ren J, Ding H, Shang J, Wang M, Wei Z, Feng S. Human umbilical cord mesenchymal stem cell-derived exosomes loaded into a composite conduit promote functional recovery after peripheral nerve injury in rats. Neural Regen Res 2024; 19:900-907. [PMID: 37843227 PMCID: PMC10664107 DOI: 10.4103/1673-5374.380911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 10/17/2023] Open
Abstract
Complete transverse injury of peripheral nerves is challenging to treat. Exosomes secreted by human umbilical cord mesenchymal stem cells are considered to play an important role in intercellular communication and regulate tissue regeneration. In previous studies, a collagen/hyaluronic acid sponge was shown to provide a suitable regeneration environment for Schwann cell proliferation and to promote axonal regeneration. This three-dimensional (3D) composite conduit contains a collagen/hyaluronic acid inner sponge enclosed in an electrospun hollow poly (lactic-co-glycolic acid) tube. However, whether there is a synergy between the 3D composite conduit and exosomes in the repair of peripheral nerve injury remains unknown. In this study, we tested a comprehensive strategy for repairing long-gap (10 mm) peripheral nerve injury that combined the 3D composite conduit with human umbilical cord mesenchymal stem cell-derived exosomes. Repair effectiveness was evaluated by sciatic functional index, sciatic nerve compound muscle action potential recording, recovery of muscle mass, measuring the cross-sectional area of the muscle fiber, Masson trichrome staining, and transmission electron microscopy of the regenerated nerve in rats. The results showed that transplantation of the 3D composite conduit loaded with human umbilical cord mesenchymal stem cell-derived exosomes promoted peripheral nerve regeneration and restoration of motor function, similar to autograft transplantation. More CD31-positive endothelial cells were observed in the regenerated nerve after transplantation of the loaded conduit than after transplantation of the conduit without exosomes, which may have contributed to the observed increase in axon regeneration and distal nerve reconnection. Therefore, the use of a 3D composite conduit loaded with human umbilical cord mesenchymal stem cell-derived exosomes represents a promising cell-free therapeutic option for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Haoshuai Tang
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Junjin Li
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongda Wang
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Ren
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Han Ding
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Shang
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhijian Wei
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Othopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Othopedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong Province, China
- Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Shiqing Feng
- Department of Othopaedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Othopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Othopedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong Province, China
- Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
7
|
Li Y, Li P, Tao Q, Abuqeis IJA, Xiyang Y. Role and limitation of cell therapy in treating neurological diseases. IBRAIN 2024; 10:93-105. [PMID: 38682022 PMCID: PMC11045202 DOI: 10.1002/ibra.12152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 05/01/2024]
Abstract
The central role of the brain in governing systemic functions within human physiology underscores its paramount significance as the focal point of physiological regulation. The brain, a highly sophisticated organ, orchestrates a diverse array of physiological processes encompassing motor control, sensory perception, cognition, emotion, and the regulation of vital functions, such as heartbeat, respiration, and hormonal equilibrium. A notable attribute of neurological diseases manifests as the depletion of neurons and the occurrence of tissue necrosis subsequent to injury. The transplantation of neural stem cells (NSCs) into the brain exhibits the potential for the replacement of lost neurons and the reconstruction of neural circuits. Furthermore, the transplantation of other types of cells in alternative locations can secrete nutritional factors that indirectly contribute to the restoration of nervous system equilibrium and the mitigation of neural inflammation. This review summarized a comprehensive investigation into the role of NSCs, hematopoietic stem cells, mesenchymal stem cells, and support cells like astrocytes and microglia in alleviating neurological deficits after cell infusion. Moreover, a thorough assessment was undertaken to discuss extant constraints in cellular transplantation therapies, concurrently delineating indispensable model-based methodologies, specifically on organoids, which were essential for guiding prospective research initiatives in this specialized field.
Collapse
Affiliation(s)
- Yu‐Qi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Peng‐Fei Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Qian Tao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | | | - Yan‐Bin Xiyang
- School of Basic MedicineKunming Medical UniversityKunmingChina
- Department of Pharmacology and Toxicology, College of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
8
|
Abi Chahine NH, Mansour VJ, Nemer LI, Najjoum CF, El Asmar EA, Boulos RT. The Regentime stem cell procedure, successful treatment for a Charcot-Marie-Tooth disease case. Clin Case Rep 2024; 12:e8358. [PMID: 38161636 PMCID: PMC10753632 DOI: 10.1002/ccr3.8358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
This report highlights the successful treatment of a Charcot-Marie-Tooth disease case using the Regentime stem cell procedure, suggesting its potential as a promising therapeutic approach for patients suffering from this challenging condition.
Collapse
|
9
|
Ranjbar N, Bakhshandeh B, Pennisi CP. Electroconductive Nanofibrous Scaffolds Enable Neuronal Differentiation in Response to Electrical Stimulation without Exogenous Inducing Factors. Bioengineering (Basel) 2023; 10:1438. [PMID: 38136029 PMCID: PMC10740536 DOI: 10.3390/bioengineering10121438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Among the various biochemical and biophysical inducers for neural regeneration, electrical stimulation (ES) has recently attracted considerable attention as an efficient means to induce neuronal differentiation in tissue engineering approaches. The aim of this in vitro study was to develop a nanofibrous scaffold that enables ES-mediated neuronal differentiation in the absence of exogenous soluble inducers. A nanofibrous scaffold composed of polycaprolactone (PCL), poly-L-lactic acid (PLLA), and single-walled nanotubes (SWNTs) was fabricated via electrospinning and its physicochemical properties were investigated. The cytocompatibility of the electrospun composite with the PC12 cell line and bone marrow-derived mesenchymal stem cells (BMSCs) was investigated. The results showed that the PCL/PLLA/SWNT nanofibrous scaffold did not exhibit cytotoxicity and supported cell attachment, spreading, and proliferation. ES was applied to cells cultured on the nanofibrous scaffolds at different intensities and the expression of the three neural markers (Nestin, Microtubule-associated protein 2, and β tubulin-3) was evaluated using RT-qPCR analysis. The results showed that the highest expression of neural markers could be achieved at an electric field intensity of 200 mV/cm, suggesting that the scaffold in combination with ES can be an efficient tool to accelerate neural differentiation in the absence of exogenous soluble inducers. This has important implications for the regeneration of nerve injuries and may provide insights for further investigations of the mechanisms underlying ES-mediated neuronal commitment.
Collapse
Affiliation(s)
- Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| |
Collapse
|
10
|
Meng Q, Burrell JC, Zhang Q, Le AD. Potential Application of Orofacial MSCs in Tissue Engineering Nerve Guidance for Peripheral Nerve Injury Repair. Stem Cell Rev Rep 2023; 19:2612-2631. [PMID: 37642899 DOI: 10.1007/s12015-023-10609-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Injury to the peripheral nerve causes potential loss of sensory and motor functions, and peripheral nerve repair (PNR) remains a challenging endeavor. The current clinical methods of nerve repair, such as direct suture, autografts, and acellular nerve grafts (ANGs), exhibit their respective disadvantages like nerve tension, donor site morbidity, size mismatch, and immunogenicity. Even though commercially available nerve guidance conduits (NGCs) have demonstrated some clinical successes, the overall clinical outcome is still suboptimal, especially for nerve injuries with a large gap (≥ 3 cm) due to the lack of biologics. In the last two decades, the combination of advanced tissue engineering technologies, stem cell biology, and biomaterial science has significantly advanced the generation of a new generation of NGCs incorporated with biological factors or supportive cells, including mesenchymal stem cells (MSCs), which hold great promise to enhance peripheral nerve repair/regeneration (PNR). Orofacial MSCs are emerging as a unique source of MSCs for PNR due to their neural crest-origin and easy accessibility. In this narrative review, we have provided an update on the pathophysiology of peripheral nerve injury and the properties and biological functions of orofacial MSCs. Then we have highlighted the application of orofacial MSCs in tissue engineering nerve guidance for PNR in various preclinical models and the potential challenges and future directions in this field.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Justin C Burrell
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Qunzhou Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
| | - Anh D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Lahoda Brodska H, Klempir J, Zavora J, Kohout P. The Role of Micronutrients in Neurological Disorders. Nutrients 2023; 15:4129. [PMID: 37836413 PMCID: PMC10574090 DOI: 10.3390/nu15194129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023] Open
Abstract
Trace elements and vitamins, collectively known as micronutrients, are essential for basic metabolic reactions in the human body. Their deficiency or, on the contrary, an increased amount can lead to serious disorders. Research in recent years has shown that long-term abnormal levels of micronutrients may be involved in the etiopathogenesis of some neurological diseases. Acute and chronic alterations in micronutrient levels may cause other serious complications in neurological diseases. Our aim was to summarize the knowledge about micronutrients in relation to selected neurological diseases and comment on their importance and the possibilities of therapeutic intervention in clinical practice.
Collapse
Affiliation(s)
- Helena Lahoda Brodska
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 499/2, 128 08 Prague, Czech Republic; (H.L.B.); (J.Z.)
| | - Jiri Klempir
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Katerinska 30, 120 00 Prague, Czech Republic
| | - Jan Zavora
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 499/2, 128 08 Prague, Czech Republic; (H.L.B.); (J.Z.)
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Pavel Kohout
- Clinic of Internal Medicine, 3rd Faculty Medicine, Charles University and Thomayer University Hospital, Videnska 800, 140 59 Prague, Czech Republic;
| |
Collapse
|
12
|
Babu S, Krishnan M, Panneerselvam A, Chinnaiyan M. A comprehensive review on therapeutic application of mesenchymal stem cells in neuroregeneration. Life Sci 2023:121785. [PMID: 37196856 DOI: 10.1016/j.lfs.2023.121785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Each year, thousands of people suffer from traumatic peripheral nerve lesions, which impair mobility and sensibility and frequently have fatal outcomes. The recovery of peripheral nerves on its own is frequently insufficient. In this regard, cell therapy is currently one of the most cutting-edge techniques for nerve healing. The purpose of this review is to highlight the properties of various types of mesenchymal stem cells (MSCs) that are critical for peripheral nerve regeneration after nerve injury. The Preferred Reporting term used to review the available literature are "nerve regeneration," "stem cells," "peripheral nerve damage," "rat," and "human" were combined. In addition, using the phrases "stem cells" and "nerve regeneration" in PubMed, a "MeSH" search was conducted. This study describes the features of the most often utilized MSCs, as well as its paracrine potential, targeted stimulation, and propensity for differentiation into Schwann-like and neuronal-like cells. For the repair of peripheral nerve lesions, ADSCs appear to be the most relevant and promising MSCs, because of their ability to sustain and increase axonal growth, as well as their outstanding paracrine activity, putative differentiation potential, low immunogenicity, and excellent post-transplant survival rate.
Collapse
Affiliation(s)
- Shyamaladevi Babu
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India.
| | - Madhan Krishnan
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| | | | - Mayilvanan Chinnaiyan
- Department of Otolaryngology, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| |
Collapse
|
13
|
Dysregulated balance in Th17/Treg axis of Pristane-induced lupus mouse model, are mesenchymal stem cells therapeutic? Int Immunopharmacol 2023; 117:109699. [PMID: 36867923 DOI: 10.1016/j.intimp.2023.109699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Despite advances in general and targeted immunosuppressive therapies, limiting all mainstay treatment options in refractory systemic lupus erythematosus (SLE) cases has necessitated the development of new therapeutic strategies. Mesenchymal stem cells (MSCs) have recently emerged with unique properties, including a solid propensity to reduce inflammation, exert immunomodulatory effects, and repair injured tissues. METHODS An animal model of acquired SLE mice was induced via intraperitoneal immunization with Pristane and affirmed by measuring specific biomarkers. Bone marrow (BM) MSCs were isolated from healthy BALB/c mice and cultured in vitro, then were identified and confirmed by flow cytometry and cytodifferentiation. Systemic MSCs transplantation was performed and then several parameters were analyzed and compared, including specific cytokines (IL-17, IL-4, IFN-ɣ, TGF-β) at the serum level, the percentage of Th cell subsets (Treg/Th17, Th1/Th2) in splenocytes, and also the relief of lupus nephritis, respectively by enzyme-linked immunosorbent assay (ELISA), flow cytometry analysis and by hematoxylin & eosin staining and also immunofluorescence assessment. Experiments were carried out with different initiation treatment time points (early and late stages of disease). Analysis of variance (ANOVA) followed by post hoc Tukey's test was used for multiple comparisons. RESULTS The rate of proteinuria, anti-double-stranded deoxyribonucleic acid (anti-dsDNA) antibodies, and serum creatinine levels decreased with BM-MSCs transplantation. These results were associated with attenuated lupus renal pathology in terms of reducing IgG and C3 deposition and lymphocyte infiltration. Our findings suggested that TGF-β (associated with lupus microenvironment) can contribute to MSC-based immunotherapy by modulating the population of TCD4+ cell subsets. Obtained results indicated that MSCs-based cytotherapy could negatively affect the progression of induced SLE by recovering the function of Treg cells, suppressing Th1, Th2, and Th17 lymphocyte function, and downregulating their pro-inflammatory cytokines. CONCLUSION MSC-based immunotherapy showed a delayed effect on the progression of acquired SLE in a lupus microenvironment-dependent manner. Allogenic MSCs transplantation revealed the ability to re-establish the balance of Th17/Treg, Th1/Th2 and restore the plasma cytokines network in a pattern dependent on disease conditions. The conflicting results of early versus advanced therapy suggest that MSCs may produce different effects depending on when they are administered and their activation status.
Collapse
|
14
|
Hoseinzadeh A, Rezaieyazdi Z, Afshari JT, Mahmoudi A, Heydari S, Moradi R, Esmaeili SA, Mahmoudi M. Modulation of Mesenchymal Stem Cells-Mediated Adaptive Immune Effectors' Repertoire in the Recovery of Systemic Lupus Erythematosus. Stem Cell Rev Rep 2023; 19:322-344. [PMID: 36272020 DOI: 10.1007/s12015-022-10452-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2022] [Indexed: 02/07/2023]
Abstract
The breakdown of self-tolerance of the immune response can lead to autoimmune conditions in which chronic inflammation induces tissue damage. Systemic lupus erythematosus (SLE) is a debilitating multisystemic autoimmune disorder with a high prevalence in women of childbearing age; however, SLE incidence, prevalence, and severity are strongly influenced by ethnicity. Although the mystery of autoimmune diseases remains unsolved, disturbance in the proportion and function of B cell subsets has a major role in SLE's pathogenesis. Additionally, colocalizing hyperactive T helper cell subgroups within inflammatory niches are indispensable. Despite significant advances in standard treatments, nonspecific immunosuppression, the risk of serious infections, and resistance to conventional therapies in some cases have raised the urgent need for new treatment strategies. Without the need to suppress the immune system, mesenchymal stem cells (MSCs), as ''smart" immune modulators, are able to control cellular and humoral auto-aggression responses by participating in precursor cell development. In lupus, due to autologous MSCs disorder, the ability of allogenic engrafted MSCs in tissue regeneration and resetting immune homeostasis with the provision of a new immunocyte repertoire has been considered simultaneously. In Brief The bone marrow mesenchymal stem cells (BM-MSCs) lineage plays a critical role in maintaining the hematopoietic stem-cell microstructure and modulating immunocytes. The impairment of BM-MSCs and their niche partially contribute to the pathogenesis of SLE-like diseases. Allogenic MSC transplantation can reconstruct BM microstructure, possibly contributing to the recovery of immunocyte phenotype restoration of immune homeostasis. In terms of future prospects of MSCs, artificially gained by ex vivo isolation and culture adaptation, the wide variety of potential mediators and mechanisms might be linked to the promotion of the immunomodulatory function of MSCs.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Department of Rheumatology, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran.,Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Heydari
- Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Reza Moradi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Immunology, Mashhad University of Medical Sciences, Azadi Square, Kalantari Blvd, Pardi's campusMashhad, Iran.
| |
Collapse
|
15
|
Current Status of Polysaccharides-Based Drug Delivery Systems for Nervous Tissue Injuries Repair. Pharmaceutics 2023; 15:pharmaceutics15020400. [PMID: 36839722 PMCID: PMC9966335 DOI: 10.3390/pharmaceutics15020400] [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: 12/21/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Neurological disorders affecting both CNS and PNS still represent one of the most critical and challenging pathologies, therefore many researchers have been focusing on this field in recent decades. Spinal cord injury (SCI) and peripheral nerve injury (PNI) are severely disabling diseases leading to dramatic and, in most cases, irreversible sensory, motor, and autonomic impairments. The challenging pathophysiologic consequences involved in SCI and PNI are demanding the development of more effective therapeutic strategies since, as yet, a therapeutic strategy that can effectively lead to a complete recovery from such pathologies is not available. Drug delivery systems (DDSs) based on polysaccharides have been receiving more and more attention for a wide range of applications, due to their outstanding physical-chemical properties. This review aims at providing an overview of the most studied polysaccharides used for the development of DDSs intended for the repair and regeneration of a damaged nervous system, with particular attention to spinal cord and peripheral nerve injury treatments. In particular, DDSs based on chitosan and their association with alginate, dextran, agarose, cellulose, and gellan were thoroughly revised.
Collapse
|
16
|
Yang Y, Zhao RC, Zhang F. Potential mesenchymal stem cell therapeutics for treating primary biliary cholangitis: advances, challenges, and perspectives. Front Cell Dev Biol 2022; 10:933565. [PMID: 35923849 PMCID: PMC9339990 DOI: 10.3389/fcell.2022.933565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Primary biliary cholangitis (PBC) is a cholestatic autoimmune liver disease characterized by the gradual destruction of small intrahepatic bile ducts that eventually leads to liver cirrhosis, failure, and even carcinoma. The treatment options for PBC are limited, and the main treatment choices are the US Food and Drug Administration–approved ursodeoxycholic acid and obeticholic acid. However, many patients fail to respond adequately to these drugs and the adverse effects frequently lead to low life quality. For patients with end-stage PBC, liver transplantation remains the only effective treatment. Given their low immunogenicity, prominent immunomodulation property, differentiation potential, and tissue maintenance capacity, mesenchymal stem cells (MSCs) are emerging as new options for treating liver diseases, including PBC. Accumulating evidence from basic research to clinical studies supports the positive effects of MSC-based therapy for treating PBC. In this review, we characterized the underlying roles and mechanisms of MSCs for treating liver diseases and highlight recent basic and clinical advances in MSC-based therapy for treating PBC. Finally, the current challenges and perspectives for MSC-based therapy in clinical application are discussed, which could help accelerate the application of MSCs in clinical practice, especially for refractory diseases such as PBC.
Collapse
Affiliation(s)
- Yanlei Yang
- Clinical Biobank, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Medical Science Research Centre, Medical Science Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- School of Life Sciences, Shanghai University, Shanghai, China
- *Correspondence: Fengchun Zhang, ; Robert Chunhua Zhao,
| | - Fengchun Zhang
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Fengchun Zhang, ; Robert Chunhua Zhao,
| |
Collapse
|
17
|
Alvites RD, Branquinho MV, Sousa AC, Lopes B, Sousa P, Prada J, Pires I, Ronchi G, Raimondo S, Luís AL, Geuna S, Varejão ASP, Maurício AC. Effects of Olfactory Mucosa Stem/Stromal Cell and Olfactory Ensheating Cells Secretome on Peripheral Nerve Regeneration. Biomolecules 2022; 12:biom12060818. [PMID: 35740943 PMCID: PMC9220795 DOI: 10.3390/biom12060818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
Cell secretome has been explored as a cell-free technique with high scientific and medical interest for Regenerative Medicine. In this work, the secretome produced and collected from Olfactory Mucosa Mesenchymal Stem Cells and Olfactory Ensheating Cells was analyzed and therapeutically applied to promote peripheral nerve regeneration. The analysis of the conditioned medium revealed the production and secretion of several factors with immunomodulatory functions, capable of intervening beneficially in the phases of nerve regeneration. Subsequently, the conditioned medium was applied to sciatic nerves of rats after neurotmesis, using Reaxon® as tube-guides. Over 20 weeks, the animals were subjected to periodic functional assessments, and after this period, the sciatic nerves and cranial tibial muscles were evaluated stereologically and histomorphometrically, respectively. The results obtained allowed to confirm the beneficial effects resulting from the application of this therapeutic combination. The administration of conditioned medium from Olfactory Mucosal Mesenchymal Stem Cells led to the best results in motor performance, sensory recovery, and gait patterns. Stereological and histomorphometric evaluation also revealed the ability of this therapeutic combination to promote nervous and muscular histologic reorganization during the regenerative process. The therapeutic combination discussed in this work shows promising results and should be further explored to clarify irregularities found in the outcomes and to allow establishing the use of cell secretome as a new therapeutic field applied in the treatment of peripheral nerves after injury.
Collapse
Affiliation(s)
- Rui D. Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Mariana V. Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Ana C. Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Justina Prada
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Isabel Pires
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Ana L. Luís
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Artur Severo P. Varejão
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Correspondence: ; Tel.: +351-91-9071286 or +351-22-0428000
| |
Collapse
|
18
|
Yin L, An Y, Chen X, Yan HX, Zhang T, Lu XG, Yan JT. Local vibration therapy promotes the recovery of nerve function in rats with sciatic nerve injury. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:265-273. [PMID: 35153133 DOI: 10.1016/j.joim.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE It has been reported that local vibration therapy can benefit recovery after peripheral nerve injury, but the optimized parameters and effective mechanism were unclear. In the present study, we investigated the effect of local vibration therapy of different amplitudes on the recovery of nerve function in rats with sciatic nerve injury (SNI). METHODS Adult male Sprague-Dawley rats were subjected to SNI and then randomly divided into 5 groups: sham group, SNI group, SNI + A-1 mm group, SNI + A-2 mm group, and SNI + A-4 mm group (A refers to the amplitude; n = 10 per group). Starting on the 7th day after model initiation, local vibration therapy was given for 21 consecutive days with a frequency of 10 Hz and an amplitude of 1, 2 or 4 mm for 5 min. The sciatic function index (SFI) was assessed before surgery and on the 7th, 14th, 21st and 28th days after surgery. Tissues were harvested on the 28th day after surgery for morphological, immunofluorescence and Western blot analysis. RESULTS Compared with the SNI group, on the 28th day after surgery, the SFIs of the treatment groups were increased; the difference in the SNI + A-2 mm group was the most obvious (95% confidence interval [CI]: [5.86, 27.09], P < 0.001), and the cross-sectional areas of myocytes in all of the treatment groups were improved. The G-ratios in the SNI + A-1 mm group and SNI + A-2 mm group were reduced significantly (95% CI: [-0.12, -0.02], P = 0.007; 95% CI: [-0.15, -0.06], P < 0.001). In addition, the expressions of S100 and nerve growth factor proteins in the treatment groups were increased; the phosphorylation expressions of ERK1/2 protein in the SNI + A-2 mm group and SNI + A-4 mm group were upregulated (95% CI: [0.03, 0.96], P = 0.038; 95% CI: [0.01, 0.94], P = 0.047, respectively), and the phosphorylation expression of Akt in the SNI + A-1 mm group was upregulated (95% CI: [0.11, 2.07], P = 0.031). CONCLUSION Local vibration therapy, especially with medium amplitude, was able to promote the recovery of nerve function in rats with SNI; this result was linked to the proliferation of Schwann cells and the activation of the ERK1/2 and Akt signaling pathways.
Collapse
Affiliation(s)
- Lu Yin
- Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yun An
- Department of Tuina, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiao Chen
- Department of Rehabilitation Medicine, the Second Rehabilitation Hospital of Shanghai, Shanghai 200441, China
| | - Hui-Xin Yan
- Department of Tuina, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Tao Zhang
- Department of Tuina, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xin-Gang Lu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Jun-Tao Yan
- Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
19
|
Yang Y, Liu S, He C, Lv T, Zeng L, Zhang F, Chen H, Zhao RC. LncRNA LYPLAL1-AS1 rejuvenates human adipose-derived mesenchymal stem cell senescence via transcriptional MIRLET7B inactivation. Cell Biosci 2022; 12:45. [PMID: 35449031 PMCID: PMC9022335 DOI: 10.1186/s13578-022-00782-x] [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: 10/20/2021] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stem cell (MSC) senescence is a phenotype of aging. Long noncoding RNAs (lncRNAs) are emerging as potential key regulators of senescence. However, the role of lncRNAs in MSC senescence remains largely unknown. Results We performed transcriptome analysis in senescent human adipose-derived MSCs (hADSCs) and identified that the lncRNA LYPLAL1 antisense RNA1 (LYPLAL1-AS1) was significantly downregulated in senescent hADSCs. LYPLAL1-AS1 expression in peripheral blood was lower in middle-aged healthy donors than in young adult donors, and correlated negatively with age. Knockdown of LYPLAL1-AS1 accelerated hADSC senescence, while LYPLAL1-AS1 overexpression attenuated it. Chromatin isolation by RNA purification (ChIRP) sequencing indicated that LYPLAL1-AS1 bound to the MIRLET7B promoter region and suppressed its transcription activity, as demonstrated by dual-luciferase assay. miR-let-7b, the transcript of MIRLET7B, was upregulated during hADSC senescence and was regulated by LYPLAL1-AS1. Furthermore, miR-let-7b mimics promoted hADSC senescence, while the inhibitors repressed it. Finally, LYPLAL1-AS1 overexpression reversed miR-let-7b-induced hADSC senescence. Conclusions Our data demonstrate that LYPLAL1-AS1 rejuvenates hADSCs through the transcriptional inhibition of MIRLET7B. Our work provides new insights into the mechanism of MSC senescence and indicates lncRNA LYPLAL1-AS1 and miR-let-7b as potential therapeutic targets in aging. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00782-x.
Collapse
Affiliation(s)
- Yanlei Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China.,Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, China
| | - Suying Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China
| | - Chengmei He
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China
| | - Taibiao Lv
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China
| | - Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China.
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, China. .,School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
20
|
Wang Y, Yu T, Hu F. Hypocapnia Stimuli-Responsive Engineered Exosomes Delivering miR-218 Facilitate Sciatic Nerve Regeneration. Front Bioeng Biotechnol 2022; 10:825146. [PMID: 35211463 PMCID: PMC8861458 DOI: 10.3389/fbioe.2022.825146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Therapeutic strategies of microRNAs (miRNAs) and exosomes have been systematically explored as an enhancing application by paracrine and modulating cellular activity after internalization of recipient cells in vitro, and progressively developed to meet the requirements of peripheral nerve regeneration in vivo. However, how to obtain exosomes with superior properties and effectively deliver miRNAs becomes a key challenge. Hypocapnia environment might play unexpected outcomes in strengthening exosome function when culturing adipose-derived stem cells (ASCs). Previously, we discovered the intensive regulation of miR-218 on the differentiation of ASCs. In the present study, we analyzed the functional differences of secreted exosomes in response to hypocapnia stimulation, and explored the application in combination with miR-218 to facilitate sciatic nerve regeneration. Our results indicated that the delivery system of engineered exosomes derived from ASCs remarkably loads upregulated miR-218 and promotes cellular activity in the recipient cells (PC12 cells), and hypocapnia stimuli-responsive exosomes exhibit strengthening properties. Furthermore, in a sciatic nerve injury model, exosomes delivering miR-218 combined with engineered scaffold facilitated the regeneration of injured sciatic nerves. In the hypocapnia-stimulated exosome group, more encouraging promotion was revealed on the regeneration of motor and nerve fibers. Hypoc-miR-218-ASC exosomes are suggested as a promising cell-free strategy for peripheral nerve repair.
Collapse
Affiliation(s)
- Yingshuai Wang
- School of Lifescience and Technology, Weifang Medical University, Weifang, China
| | - Tao Yu
- School of Lifescience and Technology, Weifang Medical University, Weifang, China
| | - Feihu Hu
- School of Lifescience and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
21
|
Yan X, Liu Y, Yu S, Huang D, Hu R. Repair Effects of Bone Marrow Mesenchymal Stem Cells on Demyelination of Trigeminal Ganglion in Rats with Trigeminal Neuralgia. J Pain Res 2022; 15:613-622. [PMID: 35250305 PMCID: PMC8894102 DOI: 10.2147/jpr.s347907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The current study investigated the effects of bone marrow mesenchymal stem cells (BMSCs) on pain behavior in rats with trigeminal neuralgia induced by infraorbital nerve chronic constriction injury (ION-CCI), and the repair effects of BMSCs on pathological changes in trigeminal ganglion demyelination. Methods BMSCs or phosphate-buffered saline (PBS) alone were injected around trigeminal ganglion in ION-CCI rats via a rat brain stereotaxic apparatus. Mechanical pain threshold (von Frey test) and face grooming behavior were measured in each group. Recovery of demyelination of trigeminal ganglion was observed via electron microscopy 2 weeks later, and BMSC differentiation was observed via immunofluorescence. Results Rats in the BMSC group exhibited significant improvements in mechanical pain threshold and face grooming behavior compared with the PBS group. BMSCs could repair demyelinating changes in trigeminal ganglion in ION-CCI rats. Only cells expressing GFAP, S-100, and p75 were observed via immunofluorescence, and no PKH67-labeled BMSCs were observed in the trigeminal ganglion. No BMSC differentiation was observed in the trigeminal ganglion. Conclusion Injection of BMSCs around the trigeminal ganglion could relieve trigeminal neuralgia effectively and repair trigeminal ganglion demyelination. No differentiation of BMSCs injected around the trigeminal ganglion into Schwann cells was observed. The mechanism of trigeminal neuralgia demyelination repair requires further investigation.
Collapse
Affiliation(s)
- Xuebin Yan
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yi Liu
- Department of Anesthesia, The First Hospital of Changsha, Changsha, People’s Republic of China
| | - Shanzi Yu
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Dong Huang
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Rong Hu
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Correspondence: Rong Hu, Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China, Tel +86 18973162969, Email
| |
Collapse
|
22
|
Lopes B, Sousa P, Alvites R, Branquinho M, Sousa AC, Mendonça C, Atayde LM, Luís AL, Varejão ASP, Maurício AC. Peripheral Nerve Injury Treatments and Advances: One Health Perspective. Int J Mol Sci 2022; 23:ijms23020918. [PMID: 35055104 PMCID: PMC8779751 DOI: 10.3390/ijms23020918] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral nerve injuries (PNI) can have several etiologies, such as trauma and iatrogenic interventions, that can lead to the loss of structure and/or function impairment. These changes can cause partial or complete loss of motor and sensory functions, physical disability, and neuropathic pain, which in turn can affect the quality of life. This review aims to revisit the concepts associated with the PNI and the anatomy of the peripheral nerve is detailed to explain the different types of injury. Then, some of the available therapeutic strategies are explained, including surgical methods, pharmacological therapies, and the use of cell-based therapies alone or in combination with biomaterials in the form of tube guides. Nevertheless, even with the various available treatments, it is difficult to achieve a perfect outcome with complete functional recovery. This review aims to enhance the importance of new therapies, especially in severe lesions, to overcome limitations and achieve better outcomes. The urge for new approaches and the understanding of the different methods to evaluate nerve regeneration is fundamental from a One Health perspective. In vitro models followed by in vivo models are very important to be able to translate the achievements to human medicine.
Collapse
Affiliation(s)
- Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Mariana Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Carla Mendonça
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Luís Miguel Atayde
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana Lúcia Luís
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Artur S. P. Varejão
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
- CECAV, Centre for Animal Sciences and Veterinary Studies, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (P.S.); (R.A.); (M.B.); (A.C.S.); (C.M.); (L.M.A.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-91-9071286
| |
Collapse
|
23
|
Potential of Fibrin Glue and Mesenchymal Stem Cells (MSCs) to Regenerate Nerve Injuries: A Systematic Review. Cells 2022; 11:cells11020221. [PMID: 35053336 PMCID: PMC8773549 DOI: 10.3390/cells11020221] [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: 12/03/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/04/2023] Open
Abstract
Cell-based therapy is a promising treatment to favor tissue healing through less invasive strategies. Mesenchymal stem cells (MSCs) highlighted as potential candidates due to their angiogenic, anti-apoptotic and immunomodulatory properties, in addition to their ability to differentiate into several specialized cell lines. Cells can be carried through a biological delivery system, such as fibrin glue, which acts as a temporary matrix that favors cell-matrix interactions and allows local and paracrine functions of MSCs. Thus, the aim of this systematic review was to evaluate the potential of fibrin glue combined with MSCs in nerve regeneration. The bibliographic search was performed in the PubMed/MEDLINE, Web of Science and Embase databases, using the descriptors ("fibrin sealant" OR "fibrin glue") AND "stem cells" AND "nerve regeneration", considering articles published until 2021. To compose this review, 13 in vivo studies were selected, according to the eligibility criteria. MSCs favored axonal regeneration, remyelination of nerve fibers, as well as promoted an increase in the number of myelinated fibers, myelin sheath thickness, number of axons and expression of growth factors, with significant improvement in motor function recovery. This systematic review showed clear evidence that fibrin glue combined with MSCs has the potential to regenerate nervous system lesions.
Collapse
|
24
|
Siemionow M, Strojny MM, Kozlowska K, Brodowska S, Grau-Kazmierczak W, Cwykiel J. Application of Human Epineural Conduit Supported with Human Mesenchymal Stem Cells as a Novel Therapy for Enhancement of Nerve Gap Regeneration. Stem Cell Rev Rep 2021; 18:642-659. [PMID: 34787795 PMCID: PMC8930890 DOI: 10.1007/s12015-021-10301-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2021] [Indexed: 12/18/2022]
Abstract
Various therapeutic methods have been suggested to enhance nerve regeneration. In this study, we propose a novel approach for enhancement of nerve gap regeneration by applying human epineural conduit (hEC) supported with human mesenchymal stem cells (hMSC), as an alternative to autograft repair. Restoration of 20 mm sciatic nerve defect with hEC created from human sciatic nerve supported with hMSC was tested in 4 experimental groups (n = 6 each) in the athymic nude rat model (Crl:NIH-Foxn1rnu): 1 - No repair control, 2 - Autograft control, 3 - Matched diameter hEC filled with 1 mL saline, 4 - Matched diameter hEC supported with 3 × 106 hMSC. Assessments included: functional tests: toe-spread and pinprick, regeneration assessment by immunofluorescence staining: HLA-1, HLA-DR, NGF, GFAP, Laminin B, S-100, VEGF, vWF and PKH26 labeling; histomorphometric analysis of myelin thickness, axonal density, fiber diameter and myelinated nerve fibers percentage; Gastrocnemius Muscle Index (GMI) and muscle fiber area ratio. Best sensory and motor function recovery, as well as GMI and muscle fiber area ratio, were observed in the autograft group, and were comparable to the hEC with hMSC group (p = 0.038). Significant improvements of myelin thickness (p = 0.003), fiber diameter (p = 0.0296), and percentage of myelinated fibers (p < 0.0001) were detected in hEC group supported with hMSC compared to hEC with saline controls. At 12-weeks after nerve gap repair, hEC combined with hMSC revealed increased expression of neurotrophic and proangiogenic factors, which corresponded with improvement of function comparable with the autograft control. Application of our novel hEC supported with hMSC provides a potential alternative to the autograft nerve repair.
Collapse
Affiliation(s)
- Maria Siemionow
- Poznan University of Medical Sciences, Poznan, Poland. .,Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.
| | - Marcin Michal Strojny
- Poznan University of Medical Sciences, Poznan, Poland.,Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA
| | - Katarzyna Kozlowska
- Poznan University of Medical Sciences, Poznan, Poland.,Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA
| | - Sonia Brodowska
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Joanna Cwykiel
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
25
|
Saffari S, Saffari TM, Chan K, Borschel GH, Shin AY. Mesenchymal stem cells and local tacrolimus delivery synergistically enhance neurite extension. Biotechnol Bioeng 2021; 118:4477-4487. [PMID: 34396506 PMCID: PMC8744499 DOI: 10.1002/bit.27916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The aim of this study was to investigate the combined effect of mesenchymal stem cells (MSC) and local delivery of tacrolimus (FK506) on nerve regeneration when applied to nerve autografts and decellularized allografts. METHODS A three-dimensional in vitro compartmented cell culture system consisting of a neonatal dorsal root ganglion adjacent to a nerve graft was used to evaluate the regenerating neurites into the peripheral nerve scaffold. Nerve autografts and allografts were treated with (i) undifferentiated MSCs, (ii) FK506 (100 ng/mL) or (iii) both (N = 9/group). After 48 hours, neurite extension was measured to quantify nerve regeneration and stem cell viability was evaluated. RESULTS Stem cell viability was confirmed in all MSC-treated grafts. Neurite extension was superior in autografts treated with FK506, and MSCs and FK506 combined (p < 0.001 and p = 0.0001, respectively), and autografts treated with MSCs (p = 0.12) were comparable to untreated autografts. In allografts, FK506 treatment and combined treatment were superior to controls (p < 0.001 and p = 0.0001, respectively), and treatment with MSCs (p = 0.09) was comparable to controls. All autograft groups were superior compared to their respective allograft treatment group (p < 0.05) in neurite extension. CONCLUSIONS Alone, either MSC or FK506 treatment improved neurite outgrowth, and combined they further enhanced neurite extension in both autografts and allografts.
Collapse
Affiliation(s)
- Sara Saffari
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Plastic-, Reconstructive- and Hand Surgery, Radboud University, Nijmegen, the Netherlands
| | - Tiam M. Saffari
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Plastic-, Reconstructive- and Hand Surgery, Radboud University, Nijmegen, the Netherlands
| | - Katelyn Chan
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Gregory H. Borschel
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Neurosciences and Mental Health, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Division of Plastic Surgery, Indiana University and Riley Hospital for Children, Indianapolis, IN, USA
| | - Alexander Y. Shin
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote the Proliferation of Schwann Cells by Regulating the PI3K/AKT Signaling Pathway via Transferring miR-21. Stem Cells Int 2021; 2021:1496101. [PMID: 34552631 PMCID: PMC8452411 DOI: 10.1155/2021/1496101] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022] Open
Abstract
As an alternative mesenchymal stem cell- (MSC-) based therapy, MSC-derived extracellular vesicles (EVs) have shown promise in the field of regenerative medicine. We previously found that human umbilical cord mesenchymal stem cell-derived EVs (hUCMSC-EVs) improved functional recovery and nerve regeneration in a rat model of sciatic nerve transection. However, the underlying mechanisms are poorly understood. Here, we demonstrated for the first time that hUCMSC-EVs promoted the proliferation of Schwann cells by activating the PI3K/AKT signaling pathway. Furthermore, we showed that hUCMSC-EVs mediated Schwann cell proliferation via transfer of miR-21. Our findings highlight a novel mechanism of hUCMSC-EVs in treating peripheral nerve injury and suggest that hUCMSC-EVs may be an attractive option for clinical application in the treatment of peripheral nerve injury.
Collapse
|
27
|
Afsharzadeh N, Lavi Arab F, Sankian M, Samiei L, Tabasi NS, Afsharzadeh D, Nikkhah K, Mahmoudi M. Comparative assessment of proliferation and immunomodulatory potential of Hypericum perforatum plant and callus extracts on mesenchymal stem cells derived adipose tissue from multiple sclerosis patients. Inflammopharmacology 2021; 29:1399-1412. [PMID: 34510276 DOI: 10.1007/s10787-021-00838-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Mesenchymal stem cells-derived adipose tissue (AT-MSCs) are recognized for the treatment of inflammatory diseases including multiple sclerosis (MS). Hypericum perforatum (HP) is an anti-inflammatory pharmaceutical plant with bioactive compounds. Plant tissue culture is a technique to improve desired pharmacological potential. The aim of this study was to compare the anti-inflammatory and proliferative effects of callus with field-growing plant extracts of HP on AT-MSCs derived from MS patients. MATERIALS AND METHODS AT-MSCs were isolated and characterized. HP callus was prepared and exposure to light spectrum (blue, red, blue-red, and control). Total phenols, flavonoids, and hypericin of HP callus and plant extracts were measured. The effects of HP extracts concentrations on proliferation were evaluated by MTT assay. Co-culture of AT-MSCs: PBMCs were challenged by HP plant and callus extracts, and Tregs percentage was assessed by flow cytometry. RESULTS Identification of MSCs was performed. Data showed that blue light could stimulate total phenols, flavonoids, and hypericin. MTT test demonstrated that plant extract in concentrations (0.03, 1.2, 2.5 and 10 μg/ml) and HP callus extract in 10 μg/ml significantly increased. Both HP extracts lead to an increase in Tregs percentage in all concentrations. In particular, a comparison between HP plant and callus extracts revealed that Tregs enhanced 3-fold more than control groups in the concentration of 10 μg/ml callus. CONCLUSIONS High concentrations of HP extracts showed effectiveness on AT-MSCs proliferation and immunomodulatory properties with a certain consequence in callus extract. HP extracts may be considered as supplementary treatments for the patients who receiving MSCs transplantation.
Collapse
Affiliation(s)
- Negin Afsharzadeh
- Immunology Research Center, Bu-Ali Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Bu-Ali Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Samiei
- Research Center for Plant Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nafiseh Sadat Tabasi
- Immunology Research Center, Bu-Ali Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Danial Afsharzadeh
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Karim Nikkhah
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Yang Y, Liu S, He C, Chen Z, Lyu T, Zeng L, Wang L, Zhang F, Chen H, Zhao RC. Long Non-coding RNA Regulation of Mesenchymal Stem Cell Homeostasis and Differentiation: Advances, Challenges, and Perspectives. Front Cell Dev Biol 2021; 9:711005. [PMID: 34368161 PMCID: PMC8339964 DOI: 10.3389/fcell.2021.711005] [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: 05/17/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022] Open
Abstract
Given the self-renewal, multi-differentiation, immunoregulatory, and tissue maintenance properties, mesenchymal stem cells (MSCs) are promising candidates for stem cell-based therapies. Breakthroughs have been made in uncovering MSCs as key contributors to homeostasis and the regenerative repair of tissues and organs derived from three germ layers. MSC differentiation into specialized cell types is sophisticatedly regulated, and accumulating evidence suggests long non-coding RNAs (lncRNAs) as the master regulators of various biological processes including the maintenance of homeostasis and multi-differentiation functions through epigenetic, transcriptional, and post-translational mechanisms. LncRNAs are ubiquitous and generally referred to as non-coding transcripts longer than 200 bp. Most lncRNAs are evolutionary conserved and species-specific; however, the weak conservation of their sequences across species does not affect their diverse biological functions. Although numerous lncRNAs have been annotated and studied, they are nevertheless only the tip of the iceberg; the rest remain to be discovered. In this review, we characterize MSC functions in homeostasis and highlight recent advances on the functions and mechanisms of lncRNAs in regulating MSC homeostasis and differentiation. We also discuss the current challenges and perspectives for understanding the roles of lncRNAs in MSC functions in homeostasis, which could help develop promising targets for MSC-based therapies.
Collapse
Affiliation(s)
- Yanlei Yang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Suying Liu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chengmei He
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhilei Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Taibiao Lyu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liuting Zeng
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li Wang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fengchun Zhang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hua Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
29
|
Millesi F, Weiss T, Mann A, Haertinger M, Semmler L, Supper P, Pils D, Naghilou A, Radtke C. Defining the regenerative effects of native spider silk fibers on primary Schwann cells, sensory neurons, and nerve-associated fibroblasts. FASEB J 2021; 35:e21196. [PMID: 33210360 PMCID: PMC7894153 DOI: 10.1096/fj.202001447r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/25/2020] [Accepted: 10/30/2020] [Indexed: 01/09/2023]
Abstract
The search for a suitable material to promote regeneration after long-distance peripheral nerve defects turned the spotlight on spider silk. Nerve conduits enriched with native spider silk fibers as internal guiding structures previously demonstrated a regenerative outcome similar to autologous nerve grafts in animal studies. Nevertheless, spider silk is a natural material with associated limitations for clinical use. A promising alternative is the production of recombinant silk fibers that should mimic the outstanding properties of their native counterpart. However, in vitro data on the regenerative features that native silk fibers provide for cells involved in nerve regeneration are scarce. Thus, there is a lack of reference parameters to evaluate whether recombinant silk fiber candidates will be eligible for nerve repair in vivo. To gain insight into the regenerative effect of native spider silk, our study aims to define the behavioral response of primary Schwann cells (SCs), nerve-associated fibroblasts (FBs), and dorsal root ganglion (DRG) neurons cultured on native dragline silk from the genus Nephila and on laminin coated dishes. The established multi-color immunostaining panels together with confocal microscopy and live cell imaging enabled the analysis of cell identity, morphology, proliferation, and migration on both substrates in detail. Our findings demonstrated that native spider silk rivals laminin coating as it allowed attachment and proliferation and supported the characteristic behavior of all tested cell types. Axonal out-growth of DRG neurons occurred along longitudinally aligned SCs that formed sustained bundled structures resembling Bungner bands present in regenerating nerves. The migration of SCs along the silk fibers achieved the reported distance of regenerating axons of about 1 mm per day, but lacked directionality. Furthermore, rFBs significantly reduced the velocity of rSCs in co-cultures on silk fibers. In summary, this study (a) reveals features recombinant silk must possess and what modifications or combinations could be useful for enhanced nerve repair and (b) provides assays to evaluate the regenerative performance of silk fibers in vitro before being applied as internal guiding structure in nerve conduits in vivo.
Collapse
Affiliation(s)
- Flavia Millesi
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Tamara Weiss
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Anda Mann
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Maximilian Haertinger
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Lorenz Semmler
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Paul Supper
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Dietmar Pils
- Division of General SurgeryDepartment of SurgeryComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Aida Naghilou
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Christine Radtke
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| |
Collapse
|
30
|
Lavorato A, Raimondo S, Boido M, Muratori L, Durante G, Cofano F, Vincitorio F, Petrone S, Titolo P, Tartara F, Vercelli A, Garbossa D. Mesenchymal Stem Cell Treatment Perspectives in Peripheral Nerve Regeneration: Systematic Review. Int J Mol Sci 2021; 22:E572. [PMID: 33430035 PMCID: PMC7827385 DOI: 10.3390/ijms22020572] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic peripheral nerve lesions affect hundreds of thousands of patients every year; their consequences are life-altering and often devastating and cause alterations in movement and sensitivity. Spontaneous peripheral nerve recovery is often inadequate. In this context, nowadays, cell therapy represents one of the most innovative approaches in the field of nerve repair therapies. The purpose of this systematic review is to discuss the features of different types of mesenchymal stem cells (MSCs) relevant for peripheral nerve regeneration after nerve injury. The published literature was reviewed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A combination of the keywords "nerve regeneration", "stem cells", "peripheral nerve injury", "rat", and "human" were used. Additionally, a "MeSH" research was performed in PubMed using the terms "stem cells" and "nerve regeneration". The characteristics of the most widely used MSCs, their paracrine potential, targeted stimulation, and differentiation potentials into Schwann-like and neuronal-like cells are described in this paper. Considering their ability to support and stimulate axonal growth, their remarkable paracrine activity, their presumed differentiation potential, their extremely low immunogenicity, and their high survival rate after transplantation, ADSCs appear to be the most suitable and promising MSCs for the recovery of peripheral nerve lesion. Clinical considerations are finally reported.
Collapse
Affiliation(s)
- Andrea Lavorato
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (S.R.); (L.M.)
| | - Marina Boido
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (M.B.); (A.V.)
| | - Luisa Muratori
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (S.R.); (L.M.)
| | - Giorgia Durante
- Faculty of Medicine and Surgery, University of Turin, 10126 Turin, TO, Italy;
| | - Fabio Cofano
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Francesca Vincitorio
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Salvatore Petrone
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Paolo Titolo
- Traumatology–Reconstructive Microsurgery, Department of Orthopaedics and Traumatology, CTO Hospital, 10126 Turin, TO, Italy;
| | - Fulvio Tartara
- Neurosurgery Unit, Istituto Clinico Città Studi (ICCS), 20131 Milan, MI, Italy;
| | - Alessandro Vercelli
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (M.B.); (A.V.)
| | - Diego Garbossa
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| |
Collapse
|
31
|
Xiao S, Zhang F, Zheng Y, Liu Z, Wang D, Wei Z, Deng C. Synergistic effect of nanofat and mouse nerve-growth factor for promotion of sensory recovery in anterolateral thigh free flaps. Stem Cells Transl Med 2020; 10:181-189. [PMID: 33043628 PMCID: PMC7848322 DOI: 10.1002/sctm.20-0226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Anterolateral thigh (ALT) free flaps are widely used for reconstruction, but poor sensory recovery of the flap tissue can cause unsatisfactory outcomes and poor function. Adipose‐derived mesenchymal stem cells (ADSCs) promote neural regeneration, but the clinical use of stem‐cell therapy has been limited by lack of regulatory approval. Nanofat is an autologous product that is prepared mechanically from harvested fat. It is enriched in ADSCs and does not contain any exogenous substances. The developmental and adult neurobiology of nerve‐growth factor (NGF) are well investigated, and mouse (m)NGF has been used to promote recovery following peripheral nerve injury. We investigated the promotion of nanofat and mNGF as either mono‐ or combined therapy on the sensory recovery of ALT free flaps. We found that nanofat and mNGF had a synergistic effect on sensory recovery that was associated with stimulation of angiogenesis and neurogenesis. Nanofat combined with mNGF was better at promoting neural regeneration and improving sensory recovery than treatment with either agent alone. The results provide a theoretical rationale for further study of the clinical use of nanofat combined with mNGF to promote the sensory recovery of ALT free flaps.
Collapse
Affiliation(s)
- Shune Xiao
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Fengling Zhang
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Yongjian Zheng
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Zhiyuan Liu
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Dali Wang
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Zairong Wei
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| | - Chengliang Deng
- Department of Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiPeople's Republic of China
| |
Collapse
|
32
|
Schwann Cell Role in Selectivity of Nerve Regeneration. Cells 2020; 9:cells9092131. [PMID: 32962230 PMCID: PMC7563640 DOI: 10.3390/cells9092131] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries result in the loss of the motor, sensory and autonomic functions of the denervated segments of the body. Neurons can regenerate after peripheral axotomy, but inaccuracy in reinnervation causes a permanent loss of function that impairs complete recovery. Thus, understanding how regenerating axons respond to their environment and direct their growth is essential to improve the functional outcome of patients with nerve lesions. Schwann cells (SCs) play a crucial role in the regeneration process, but little is known about their contribution to specific reinnervation. Here, we review the mechanisms by which SCs can differentially influence the regeneration of motor and sensory axons. Mature SCs express modality-specific phenotypes that have been associated with the promotion of selective regeneration. These include molecular markers, such as L2/HNK-1 carbohydrate, which is differentially expressed in motor and sensory SCs, or the neurotrophic profile after denervation, which differs remarkably between SC modalities. Other important factors include several molecules implicated in axon-SC interaction. This cell–cell communication through adhesion (e.g., polysialic acid) and inhibitory molecules (e.g., MAG) contributes to guiding growing axons to their targets. As many of these factors can be modulated, further research will allow the design of new strategies to improve functional recovery after peripheral nerve injuries.
Collapse
|
33
|
Mittal K, Schrenk-Siemens K. Lessons from iPSC research: Insights on peripheral nerve disease. Neurosci Lett 2020; 738:135358. [PMID: 32898616 DOI: 10.1016/j.neulet.2020.135358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 01/13/2023]
Abstract
With the publication of their breakthrough discovery describing the induction of pluripotent stem cells (iPSCs) from mouse and human fibroblasts, Takahashi and Yamanaka have changed the scientific landscape. The possibility of deriving human pluripotent stem cells from almost any somatic cell has provided the unprecedented opportunity to study specific hereditary diseases in human cells. In the context of diseases affecting peripheral nerves, iPSC platforms are now being increasingly utilized to investigate the underlying pathology as well as regenerative strategies. Peripheral neuropathies result in peripheral nerve damage, leading to - among other things - the degeneration of affected nerve fibers accompanied by severe sensory, motor and autonomic symptoms, often including intense pain. The generation of iPSCs from hereditary forms of peripheral neuropathies and their directed differentiation into cell types most affected by the disease can be instrumental to better understanding the pathological mechanisms underlying these disorders and to investigating cell replacement strategies for repair. In this minireview, we highlight studies that have used iPSCs to investigate the therapeutic potential of iPSC-derived Schwann cell-like cells for nerve regeneration, as well as studies using patient iPSC derivatives to investigate their contribution to disease pathology.
Collapse
Affiliation(s)
- Kritika Mittal
- Department of Anatomy, University of Heidelberg, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| | - Katrin Schrenk-Siemens
- Department of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
34
|
Yang Z, Zheng C, Zhang F, Lin B, Cao M, Tian X, Zhang J, Zhang X, Shen J. Magnetic resonance imaging of enhanced nerve repair with mesenchymal stem cells combined with microenvironment immunomodulation in neurotmesis. Muscle Nerve 2020; 61:815-825. [PMID: 32170960 DOI: 10.1002/mus.26862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 02/04/2020] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The immuno-microenvironment of injured nerves adversely affects mesenchymal stem cell (MSC) therapy for neurotmesis. Magnetic resonance imaging (MRI) can be used noninvasively to monitor nerve degeneration and regeneration. The aim of this study was to investigate nerve repair after MSC transplantation combined with microenvironment immunomodulation in neurotmesis by using multiparametric MRI. METHODS Rats with sciatic nerve transection and surgical coaptation were treated with MSCs combined with immunomodulation or MSCs alone. Serial multiparametric MRI examinations were performed over an 8-week period after surgery. RESULTS Nerves treated with MSCs combined with immunomodulation showed better functional recovery, rapid recovery of nerve T2, fractional anisotropy and radial diffusivity values, and more rapid restoration of the fiber tracks than nerves treated with MSCs alone. DISCUSSION Transplantation of MSCs in combination with immunomodulation can exert a synergistic repair effect on neurotmesis, which can be monitored by multiparametric MRI.
Collapse
Affiliation(s)
- Zehong Yang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chushan Zheng
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Binglin Lin
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xuwei Tian
- Department of Radiology, The First People's Hospital of Kashgar, Kashgar, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xiao Zhang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
35
|
Liu Y, Wang H. Peripheral nerve injury induced changes in the spinal cord and strategies to counteract/enhance the changes to promote nerve regeneration. Neural Regen Res 2020; 15:189-198. [PMID: 31552884 PMCID: PMC6905333 DOI: 10.4103/1673-5374.265540] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Peripheral nerve injury leads to morphological, molecular and gene expression changes in the spinal cord and dorsal root ganglia, some of which have positive impact on the survival of neurons and nerve regeneration, while the effect of others is the opposite. It is crucial to take prompt measures to capitalize on the positive effects of these reactions and counteract the negative impact after peripheral nerve injury at the level of spinal cord, especially for peripheral nerve injuries that are severe, located close to the cell body, involve long distance for axons to regrow and happen in immature individuals. Early nerve repair, exogenous supply of neurotrophic factors and Schwann cells can sustain the regeneration inductive environment and enhance the positive changes in neurons. Administration of neurotrophic factors, acetyl-L-carnitine, N-acetyl-cysteine, and N-methyl-D-aspartate receptor antagonist MK-801 can help counteract axotomy-induced neuronal loss and promote regeneration, which are all time-dependent. Sustaining and reactivation of Schwann cells after denervation provides another effective strategy. FK506 can be used to accelerate axonal regeneration of neurons, especially after chronic axotomy. Exploring the axotomy-induced changes after peripheral nerve injury and applying protective and promotional measures in the spinal cord which help to retain a positive functional status for neuron cell bodies will inevitably benefit regeneration of the peripheral nerve and improve functional outcomes.
Collapse
Affiliation(s)
- Yan Liu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China; Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
36
|
Kubiak CA, Grochmal J, Kung TA, Cederna PS, Midha R, Kemp SWP. Stem-cell-based therapies to enhance peripheral nerve regeneration. Muscle Nerve 2019; 61:449-459. [PMID: 31725911 DOI: 10.1002/mus.26760] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022]
Abstract
Peripheral nerve injury remains a major cause of morbidity in trauma patients. Despite advances in microsurgical techniques and improved understanding of nerve regeneration, obtaining satisfactory outcomes after peripheral nerve injury remains a difficult clinical problem. There is a growing body of evidence in preclinical animal studies demonstrating the supportive role of stem cells in peripheral nerve regeneration after injury. The characteristics of both mesoderm-derived and ectoderm-derived stem cell types and their role in peripheral nerve regeneration are discussed, specifically focusing on the presentation of both foundational laboratory studies and translational applications. The current state of clinical translation is presented, with an emphasis on both ethical considerations of using stems cells in humans and current governmental regulatory policies. Current advancements in cell-based therapies represent a promising future with regard to supporting nerve regeneration and achieving significant functional recovery after debilitating nerve injuries.
Collapse
Affiliation(s)
- Carrie A Kubiak
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Joey Grochmal
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Theodore A Kung
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Paul S Cederna
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rajiv Midha
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephen W P Kemp
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
37
|
Carvalho CR, Oliveira JM, Reis RL. Modern Trends for Peripheral Nerve Repair and Regeneration: Beyond the Hollow Nerve Guidance Conduit. Front Bioeng Biotechnol 2019; 7:337. [PMID: 31824934 PMCID: PMC6882937 DOI: 10.3389/fbioe.2019.00337] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve repair and regeneration remains among the greatest challenges in tissue engineering and regenerative medicine. Even though peripheral nerve injuries (PNIs) are capable of some degree of regeneration, frail recovery is seen even when the best microsurgical technique is applied. PNIs are known to be very incapacitating for the patient, due to the deprivation of motor and sensory abilities. Since there is no optimal solution for tackling this problem up to this day, the evolution in the field is constant, with innovative designs of advanced nerve guidance conduits (NGCs) being reported every day. As a basic concept, a NGC should act as a physical barrier from the external environment, concomitantly acting as physical guidance for the regenerative axons across the gap lesion. NGCs should also be able to retain the naturally released nerve growth factors secreted by the damaged nerve stumps, as well as reducing the invasion of scar tissue-forming fibroblasts to the injury site. Based on the neurobiological knowledge related to the events that succeed after a nerve injury, neuronal subsistence is subjected to the existence of an ideal environment of growth factors, hormones, cytokines, and extracellular matrix (ECM) factors. Therefore, it is known that multifunctional NGCs fabricated through combinatorial approaches are needed to improve the functional and clinical outcomes after PNIs. The present work overviews the current reports dealing with the several features that can be used to improve peripheral nerve regeneration (PNR), ranging from the simple use of hollow NGCs to tissue engineered intraluminal fillers, or to even more advanced strategies, comprising the molecular and gene therapies as well as cell-based therapies.
Collapse
Affiliation(s)
- Cristiana R. Carvalho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| |
Collapse
|
38
|
Liu Y, Dong R, Zhang C, Yang Y, Xu Y, Wang H, Zhang M, Zhu J, Wang Y, Sun Y, Zhang Z. Therapeutic effects of nerve leachate-treated adipose-derived mesenchymal stem cells on rat sciatic nerve injury. Exp Ther Med 2019; 19:223-231. [PMID: 31853293 PMCID: PMC6909684 DOI: 10.3892/etm.2019.8203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023] Open
Abstract
Peripheral nerve injury (PNI) is a common condition, often resulting from physical nerve injury and trauma. Successful repair of the peripheral nerve is dependent on the regenerative activity of Schwann cells (SCs). Application of SC-like adipose-derived mesenchymal stem cells (ADSCs) may be a suitable cell-based therapy for PNI. In the present study, nerve leachate derived from the rat sciatic nerve was used to induce the differentiation of ADSCs. These cells were placed in an acellular biological scaffold, which was then grafted to a rat sciatic nerve to bridge a 1-cm gap. Sprague-Dawley rats were divided into four groups: Scaffold only, untreated ADSCs + scaffold, nerve leachate-treated ADSCs + scaffold and autograft. Two-months post-transplant, the structure and function of the regenerated nerves and the recovery of the innervated muscles was analyzed. After transplant, there was a significant increase in the average area (15.86%; P<0.05), density (23.13%; P<0.05) and thickness (43.24%; P<0.05) of regenerated nerve fibers in the nerve leachate-treated ADSCs + scaffold group compared with the untreated ADSCs + scaffold group. The nerve conduction velocity in the nerve leachate-treated ADSCs + scaffold and autograft groups was superior to that in the other groups. In the nerve leachate-treated ADSCs + scaffold group, the cross-sectional area of the gastrocnemius increased by 39.28% (P<0.05) and the cross-sectional area of collagen fibers decreased by 29.87% (P<0.05) compared with the ADSCs + scaffold group. Moreover, the therapeutic effect of nerve leachate-treated ADSCs + scaffold on PNI was similar to that of an autograft. These results suggest that nerve leachate-treated ADSCs may promote the repair of PNI.
Collapse
Affiliation(s)
- Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Ruiqi Dong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Chunyan Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Yuxiang Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Yaolu Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Haojie Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Mengyu Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Jiamin Zhu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Yuqin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China.,Engineering Research Center for Mutton Sheep Breeding of Henan Province, Luoyang, Henan 471023, P.R. China
| | - Yanhong Sun
- Department of Physiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010110, P.R. China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| |
Collapse
|