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 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Song X, Li R, Chu X, Li Q, Li R, Li Q, Tong KY, Gu X, Ming D. Multilevel analysis of the central-peripheral-target organ pathway: contributing to recovery after peripheral nerve injury. Neural Regen Res 2025; 20:2807-2822. [PMID: 39435615 PMCID: PMC11826472 DOI: 10.4103/nrr.nrr-d-24-00641] [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: 06/11/2024] [Revised: 08/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Peripheral nerve injury is a common neurological condition that often leads to severe functional limitations and disabilities. Research on the pathogenesis of peripheral nerve injury has focused on pathological changes at individual injury sites, neglecting multilevel pathological analysis of the overall nervous system and target organs. This has led to restrictions on current therapeutic approaches. In this paper, we first summarize the potential mechanisms of peripheral nerve injury from a holistic perspective, covering the central nervous system, peripheral nervous system, and target organs. After peripheral nerve injury, the cortical plasticity of the brain is altered due to damage to and regeneration of peripheral nerves; changes such as neuronal apoptosis and axonal demyelination occur in the spinal cord. The nerve will undergo axonal regeneration, activation of Schwann cells, inflammatory response, and vascular system regeneration at the injury site. Corresponding damage to target organs can occur, including skeletal muscle atrophy and sensory receptor disruption. We then provide a brief review of the research advances in therapeutic approaches to peripheral nerve injury. The main current treatments are conducted passively and include physical factor rehabilitation, pharmacological treatments, cell-based therapies, and physical exercise. However, most treatments only partially address the problem and cannot complete the systematic recovery of the entire central nervous system-peripheral nervous system-target organ pathway. Therefore, we should further explore multilevel treatment options that produce effective, long-lasting results, perhaps requiring a combination of passive (traditional) and active (novel) treatment methods to stimulate rehabilitation at the central-peripheral-target organ levels to achieve better functional recovery.
Collapse
Affiliation(s)
- Xizi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Ruixin Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Xiaolei Chu
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qi Li
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Ruihua Li
- Department of Hand Microsurgery, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qingwen Li
- School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Kai-Yu Tong
- Department of Biomedical Engineering, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| |
Collapse
|
3
|
Goldschmidt-Clermont PJ, Khan A, Jimsheleishvili G, Graham P, Brooks A, Silvera R, Goldschmidt AJ, Pearse DD, Dietrich WD, Levi AD, Guest JD. Treating amyotrophic lateral sclerosis with allogeneic Schwann cell-derived exosomal vesicles: a case report. Neural Regen Res 2025; 20:1207-1216. [PMID: 38922880 PMCID: PMC11438342 DOI: 10.4103/nrr.nrr-d-23-01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 06/28/2024] Open
Abstract
Schwann cells are essential for the maintenance and function of motor neurons, axonal networks, and the neuromuscular junction. In amyotrophic lateral sclerosis, where motor neuron function is progressively lost, Schwann cell function may also be impaired. Recently, important signaling and potential trophic activities of Schwann cell-derived exosomal vesicles have been reported. This case report describes the treatment of a patient with advanced amyotrophic lateral sclerosis using serial intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles, marking, to our knowledge, the first instance of such treatment. An 81-year-old male patient presented with a 1.5-year history of rapidly progressive amyotrophic lateral sclerosis. After initial diagnosis, the patient underwent a combination of generic riluzole, sodium phenylbutyrate for the treatment of amyotrophic lateral sclerosis, and taurursodiol. The patient volunteered to participate in an FDA-approved single-patient expanded access treatment and received weekly intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles to potentially restore impaired Schwann cell and motor neuron function. We confirmed that cultured Schwann cells obtained from the amyotrophic lateral sclerosis patient via sural nerve biopsy appeared impaired (senescent) and that exposure of the patient's Schwann cells to allogeneic Schwann cell-derived exosomal vesicles, cultured expanded from a cadaver donor improved their growth capacity in vitro. After a period of observation lasting 10 weeks, during which amyotrophic lateral sclerosis Functional Rating Scale-Revised and pulmonary function were regularly monitored, the patient received weekly consecutive infusions of 1.54 × 10 12 (×2), and then consecutive infusions of 7.5 × 10 12 (×6) allogeneic Schwann cell-derived exosomal vesicles diluted in 40 mL of Dulbecco's phosphate-buffered saline. None of the infusions were associated with adverse events such as infusion reactions (allergic or otherwise) or changes in vital signs. Clinical lab serum neurofilament and cytokine levels measured prior to each infusion varied somewhat without a clear trend. A more sensitive in-house assay suggested possible inflammasome activation during the disease course. A trend for clinical stabilization was observed during the infusion period. Our study provides a novel approach to address impaired Schwann cells and possibly motor neuron function in patients with amyotrophic lateral sclerosis using allogeneic Schwann cell-derived exosomal vesicles. Initial findings suggest that this approach is safe.
Collapse
Affiliation(s)
| | - Aisha Khan
- Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - George Jimsheleishvili
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Patricia Graham
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Adriana Brooks
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Risset Silvera
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Damien D. Pearse
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - James D. Guest
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Tajabadi Z, Dadkhah PA, Gholami Chahkand MS, Esmaeilpour Moallem F, Karimi MA, Amini-Salehi E, Karimi M. Exploring the role of exosomes in diabetic neuropathy: From molecular mechanisms to therapeutic potential. Biomed Pharmacother 2025; 185:117959. [PMID: 40056828 DOI: 10.1016/j.biopha.2025.117959] [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: 12/25/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
Diabetic neuropathy (DN) is a debilitating complication of diabetes mellitus (DM), characterized by progressive neuronal damage, sensory dysfunction, and impaired quality of life. Recent advances in exosome research have elucidated their crucial role in DN's pathogenesis, diagnosis, and treatment. Exosomes-nanoscale extracellular vesicles-function as vehicles for molecular cargo, including microRNAs (miRNAs), proteins, and lipids, which mediate intercellular communication and regulate key biological processes. Pathologically, hyperglycemia and hyperlipidemia induce the release of exosomes enriched with pathogenic miRNAs, such as miR-130a and miR-20b-3p, which disrupt neuronal function, axonal regeneration, and inflammatory pathways. Conversely, diagnostic studies highlight the utility of exosomal biomarkers like miR-7 and miR-221 in the early detection and monitoring of DN. Therapeutically, Schwann cell-derived and mesenchymal stromal cell (MSC)-derived exosomes demonstrate neuroprotective and reparative effects by enhancing mitochondrial function, modulating inflammation, and promoting axonal repair. Emerging approaches, including engineered exosomes and miRNA-enriched vesicles, further expand their therapeutic potential. Despite these advances, challenges such as standardization, large-scale production, and clinical validation remain in translating these findings into clinical practice. This review underscores the multifaceted roles of exosomes in DN and highlights their potential as innovative tools for precision diagnostics and targeted therapies, paving the way for future research and clinical applications.
Collapse
Affiliation(s)
- Zohreh Tajabadi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kyiv, Ukraine
| |
Collapse
|
5
|
Spinelli M, Fusco S, Grassi C. Therapeutic potential of stem cell-derived extracellular vesicles in neurodegenerative diseases associated with cognitive decline. Stem Cells 2025; 43:sxae074. [PMID: 39541178 DOI: 10.1093/stmcls/sxae074] [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: 05/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
In the central nervous system, cell-to-cell interaction is essential for brain plassticity and repair, and its alteration is critically involved in the development of neurodegenerative diseases. Neural stem cells are a plentiful source of biological signals promoting neuroplasticity and the maintenance of cognitive functions. Extracellular vesicles (EVs) represent an additional strategy for cells to release signals in the surrounding cellular environment or to exchange information among both neighboring and distant cells. In the last years, rising attention has been devoted to the ability of stem cell (SC)-derived EVs to counteract inflammatory and degenerative brain disorders taking advantage of their immunomodulatory capacities and regenerative potential. Here, we review the role of adult neurogenesis impairment in the cognitive decline associated with neurodegenerative diseases and describe the beneficial effects of SC-derived EVs on brain plasticity and repair also discussing the advantages of SC-derived EV administration vs SC transplantation in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Matteo Spinelli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
6
|
Duda M, Maciak P, Tylko G, Wartalski K, Trzcińska M, Romek M, Samiec M. Research note: Harnessing the potential of chicken blastodermal cells as a new frontier in poultry science and biotechnology - Establishment of embryonic stem cells and their differentiability towards Schwann cell-like cell lineages. Poult Sci 2025; 104:104701. [PMID: 39721266 PMCID: PMC11729637 DOI: 10.1016/j.psj.2024.104701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
The nervous system's regenerative potential has sparked interest in exploring novel approaches to generate Schwann cell-like cells (SC-LCs) from chicken blastoderm (B)-derived embryonic stem cells (B-ESCs). This study investigates the hypothesis that specific growth factors, when used during ex-ovo culture, can induce the differentiation of chicken B-ESCs into cells resembling Schwann cells (SCs). Blastodermal cells (BCs) were isolated from in vivo-fertilized eggs at stage X followed by 14-d proliferative culture (PRC) of B-ESCs and subsequent 14-d glial/neurolemmogenic differentiation culture (DFC). Western blot and immunofluorescence analyses were applied to identify ESC-related markers and SC-specific proteins. Ultimately, slender or triangular cells resembling early SCs, designated as SC-LCs, were generated. Pluripotency-related markers OCT4, SOX2, SSEA-4 and TRA-1-60 were detected in B-ESCs, while SC-specific markers such as GFAP and S-100β were identified in neurolemmogenically differentiated B-ESC derivatives (SC-LCs). The current study demonstrates, for the first time, the successful differentiation of chicken B-ESCs into SC-LCs through ex-ovo sequential culture. After PRC termination, B-ESCs exhibited pluripotent characteristics as shown by the presence of OCT4, SOX2, SSEA-4 and TRA-1-60 markers. Subsequent DFC led to the acquisition of SC-like morphology by B-ESCs, confirmed by the expression of SC-specific markers GFAP and S-100β in the resulting SC-LCs. These findings highlight the potential of B-ESCs as a valuable source for propagating SC-LCs, with implications for regenerative medicine and neural/glial tissue engineering applications. Further research exploring the functional attributes of B-ESC-derived SC-LCs is required to elucidate their therapeutic potential in nerve reconstruction/repair.
Collapse
Affiliation(s)
- Małgorzata Duda
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow 30-387, Poland
| | - Patrycja Maciak
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow 30-387, Poland
| | - Grzegorz Tylko
- Department of Cell Biology and Imaging, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow 30-387, Poland
| | - Kamil Wartalski
- Department of Histology, Jagiellonian University Medical College, Krakow 31-034, Poland
| | - Monika Trzcińska
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Balice 32-083, Poland
| | - Marek Romek
- Department of Cell Biology and Imaging, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow 30-387, Poland
| | - Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Balice 32-083, Poland.
| |
Collapse
|
7
|
Zhang L, Xie J, Dai W, Lu B, Yi S. Schwann cells in regeneration and cancer. Front Pharmacol 2025; 16:1506552. [PMID: 39981185 PMCID: PMC11840318 DOI: 10.3389/fphar.2025.1506552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
Schwann cells are specific peripheral glial cells with remarkable plasticity following peripheral nerve injury. Injury responses stimulate c-Jun activation in Schwann cells, drive epithelial-mesenchymal transition and cellular phenotypic changes, and induce the generation of reprogrammed repair Schwann cells to orchestrate peripheral nerve regeneration process. Schwann cells and/or Schwann cell-derived molecules are commonly used as supporting cells and/or neurotrophic factors to construct Schwann cell-based tissue-engineered nerve grafts for repairing severe peripheral nerve injury with long defects. Transplantation of Schwann cells and/or Schwann cell-derived molecules also serves as a helpful approach for the treatment of other injured tissues, such as the spinal cord, skin, digit tip, and bone. Schwann cells are not only associated with tissue regeneration but also involved in tumorigenesis and tumor progression. Schwann cells are the major cellular component of neurofibromatosis type 1 and the sole cell type in neurofibromatosis type 2 and schwannomatosis. In addition, Schwann cells also function as an important player in the tumor microenvironment and aid in the growth and invasiveness of many other solid cancers. In the present review, we outline the physiological and pathological activities of Schwann cells and discuss the functional roles of Schwann cells in homeostasis, regeneration, and cancer.
Collapse
Affiliation(s)
- Lan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Jiale Xie
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Wenyu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Bing Lu
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
8
|
Chen Y, Qi W, Wang Z, Niu F. Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics 2025; 17:147. [PMID: 40006514 PMCID: PMC11858990 DOI: 10.3390/pharmaceutics17020147] [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: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Exosomes have emerged as promising therapeutic agents in regenerative medicine. This review introduces a novel cell type-oriented perspective to systematically analyze exosomal properties in regenerative therapies. To our knowledge, this review is the first to comprehensively compare exosomes based on cellular source type, offering unprecedented insights into selecting optimal exosome producers for targeted regenerative applications. Factors beyond cellular origin influencing exosomal therapeutic efficacy, such as donor sites and collection methods, are also explored here. By synthesizing key advances, we propose promising research directions in the end. We aim to accelerate the development of more effective exosome-based regenerative therapies and highlight underexplored directions in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | - Feng Niu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan, Beijing 100144, China; (Y.C.)
| |
Collapse
|
9
|
Shouman S, Hesham N, Salem TZ. Viruses and neurodegeneration: a growing concern. J Transl Med 2025; 23:46. [PMID: 39800721 PMCID: PMC11727702 DOI: 10.1186/s12967-024-06025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
Neurodegenerative diseases (NDDs) cause a progressive loss of neurons. Since NDDs are multifactorial, the precise etiology varies on the basis of the type of disease and patient history. Cohort studies and case studies have demonstrated a potential link between viral infections and the onset or progression of NDDs. Recent findings concerning the mechanisms by which neuropathic infections occur have provided more insights into the importance of such connections. In this review, we aim to elaborate on the occurrence of the neuropathic effects of viruses from epidemiological, clinical, and biological perspectives while highlighting potential treatments and challenges. One of the key players in viral neuropathogenesis is neuroinflammation caused by the immune response to the virus; this can occur due to both neurotropic and nonneurotropic viruses. The COVID-19 pandemic has raised concerns about whether vaccines are essential for preventing viruses or whether vaccines may play a part in exacerbating or accelerating NDDs. By classifying viruses and the common NDDs associated with them and further delving into their cellular pathways, this review provides insights to advance the development of potential treatments and diagnostic methods.
Collapse
Affiliation(s)
- S Shouman
- Biomedical Sciences Program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt
| | - N Hesham
- Biomedical Sciences Program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt
- Molecular Biology and Virology Laboratory (MBVL), Center for X-Ray Determination of the Structure of Matter (CXDS), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt
| | - T Z Salem
- Biomedical Sciences Program, UST, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt.
- Molecular Biology and Virology Laboratory (MBVL), Center for X-Ray Determination of the Structure of Matter (CXDS), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt.
| |
Collapse
|
10
|
Errante EL, Tigre JY, Schaeffer EA, Costello MC, Kloehn AJ, Puerto A, Khan A, Pressman Y, Silvera R, Sanchez FJ, Noga BR, Dietrich WD, Levi AD, Burks SS. Comparison of cellular-based therapies following a long-segmental peripheral nerve defect in a rat model. PLoS One 2025; 20:e0313292. [PMID: 39774446 PMCID: PMC11706366 DOI: 10.1371/journal.pone.0313292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Peripheral nerve injury (PNI) is characterized by a loss of cellular and axonal integrity, often leading to limited functional recovery and pain. Many PNIs are not amenable to repair with traditional techniques; however, cell therapies, particularly Schwann cells (SCs), offer the promise of neural tissue replacement and functional improvement. Exosomes, which carry cellular signaling molecules, can be secreted by SCs and have shown promise in PNI. Our laboratory has had success using SCs in preclinical and clinical treatment settings. Transplanted cells have several known limitations, which exosomes mitigate. To that end, the current study investigated if implanted SC-derived exosomes in conduits, conduits with SCs, reverse autograft, or empty conduits comparably improve axonal regeneration and pain outcomes 16-weeks after repair of a long gap PNI in adult rats. Results show that there were no differences between groups in the von Frey filament testing or in the Hargreaves test. Electrophysiological testing showed a significant difference between the injured (ipsilateral) and uninjured (contralateral) limbs while histological assessment showed a significant difference between axonal counts in different areas of the conduit. Based on the results of the current study, more research is needed to understand the therapeutic role of exosomes in PNI.
Collapse
Affiliation(s)
- Emily L. Errante
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Joseph Yunga Tigre
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ericka A. Schaeffer
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Meredith C. Costello
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Andrew J. Kloehn
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Aiko Puerto
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Aisha Khan
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Yelena Pressman
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Risset Silvera
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Francisco J. Sanchez
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Brian R. Noga
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - S. Shelby Burks
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
11
|
Ahmad S, Christova T, Pye M, Narimatsu M, Song S, Wrana JL, Attisano L. Small Extracellular Vesicles Promote Axon Outgrowth by Engaging the Wnt-Planar Cell Polarity Pathway. Cells 2025; 14:56. [PMID: 39791757 PMCID: PMC11720052 DOI: 10.3390/cells14010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
In neurons, the acquisition of a polarized morphology is achieved upon the outgrowth of a single axon from one of several neurites. Small extracellular vesicles (sEVs), such as exosomes, from diverse sources are known to promote neurite outgrowth and thus may have therapeutic potential. However, the effect of fibroblast-derived exosomes on axon elongation in neurons of the central nervous system under growth-permissive conditions remains unclear. Here, we show that fibroblast-derived sEVs promote axon outgrowth and a polarized neuronal morphology in mouse primary embryonic cortical neurons. Mechanistically, we demonstrate that the sEV-induced increase in axon outgrowth requires endogenous Wnts and core PCP components including Prickle, Vangl, Frizzled, and Dishevelled. We demonstrate that sEVs are internalized by neurons, colocalize with Wnt7b, and induce relocalization of Vangl2 to the distal axon during axon outgrowth. In contrast, sEVs derived from neurons or astrocytes do not promote axon outgrowth, while sEVs from activated astrocytes inhibit elongation. Thus, our data reveal that fibroblast-derived sEVs promote axon elongation through the Wnt-PCP pathway in a manner that is dependent on endogenous Wnts.
Collapse
Affiliation(s)
- Samar Ahmad
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Tania Christova
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Melanie Pye
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Masahiro Narimatsu
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Siyuan Song
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Jeffrey L. Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Liliana Attisano
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| |
Collapse
|
12
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2025; 27:36-50. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
13
|
Li S, Zhang F, Wang G, Liu Q, Wang X, Chen Q, Chu D. Tau Isoform-Regulated Schwann Cell Proliferation and Migration Improve Peripheral Nerve Regeneration After Injury. Int J Mol Sci 2024; 25:12352. [PMID: 39596423 PMCID: PMC11594695 DOI: 10.3390/ijms252212352] [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: 10/09/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Tau is a microtubule-associated protein that plays a vital role in the mammalian nervous system. Alternative splicing of the MAPT gene leads to the formation of tau isoforms with varying N-terminal inserts and microtubule-binding repeats. Dysregulation of tau alternative splicing has been linked to diseases in the central nervous system, but the roles of tau isoforms in the peripheral nervous system remain unclear. Here, we investigated the alternative splicing of tau exons 4A and 10 in the sciatic nerve and Schwann cells during development and following injury. We discovered that low-molecular-weight (LMW) tau, resulting from the exclusion of exon 4A, and 3R tau, generated by the exclusion of exon 10, diminishes with aging in rat sciatic nerve and Schwann cells. High-molecular-weight (HMW) tau and 3R tau increase in the adult sciatic nerve post-injury. We constructed viruses that expressed HMW-4R, LMW-4R, HMW-3R, and LMW-3R and introduced them into cultured cells or the distal part of the injured sciatic nerve to assess their effects on Schwann cell migration and proliferation. We also examined the effects of the four isoforms on axon growth and debris clearance after sciatic nerve injury. Our results demonstrated that tau isoforms inhibit Schwann cell proliferation while promoting Schwann cell migration and sciatic nerve regeneration. Specifically, the 3R-tau isoforms were more effective than the 4R-tau isoforms in promoting nerve regeneration. In conclusion, our study reveals the roles of tau isoforms in the peripheral nervous system and provides insights into the development of new therapeutic strategies for peripheral nerve injuries.
Collapse
Affiliation(s)
- Shiying Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Fuqian Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Guifang Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Qianyan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Qianqian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (S.L.); (F.Z.); (G.W.); (Q.L.); (X.W.)
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| |
Collapse
|
14
|
Wang JL, Huang QM, Hu DX, Zhang WJ. Therapeutic effect of exosomes derived from Schwann cells in the repair of peripheral nerve injury. Life Sci 2024; 357:123086. [PMID: 39357794 DOI: 10.1016/j.lfs.2024.123086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Peripheral nerve injury (PNI) can cause nerve demyelination, neuronal apoptosis, axonal atrophy, inflammatory infiltration, glial scar formation, and other pathologies that can lead to sensory and motor dysfunction and seriously affect the psychosomatic health of patients. There is currently no effective treatment method, so exploring a promising treatment method is of great significance. Several studies have revealed the therapeutic roles of Schwann cells (SCs) and their exosomes in nerve injury repair. Exosomes are extracellular nanovesicles secreted by cells that act as key molecules in intercellular communication. Progress has been made in understanding the role of exosomes derived from SCs (SC-EXOs) in peripheral nerve regeneration, including the promotion of axonal regeneration and myelin formation, anti-inflammation, vascular regeneration, neuroprotection, and neuroregulation. Therefore, in this paper, we summarize the functional characteristics of SC-EXOs and discuss their potential therapeutic effects on PNI repair as well as some existing problems and future challenges.
Collapse
Affiliation(s)
- Jia-Ling Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Qi-Ming Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
15
|
Liu C, Li Q, Ma JX, Lu B, Criswell T, Zhang Y. Exosome-mediated renal protection: Halting the progression of fibrosis. Genes Dis 2024; 11:101117. [PMID: 39263535 PMCID: PMC11388648 DOI: 10.1016/j.gendis.2023.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/13/2024] Open
Abstract
Renal fibrosis is a complex and multifactorial process that involves inflammation, cell proliferation, collagen, and fibronectin deposition in the kidney, ultimately leading to chronic kidney disease and even end-stage renal disease. The main goal of treatment is to slow down or halt the progression of fibrosis and to improve or preserve kidney function. Despite significant progress made in understanding the underlying mechanisms of renal fibrosis, current therapies have limited renal protection as the disease progresses. Exosomes derived from stem cells are a newer area of research for the treatment of renal fibrosis. Exosomes as nano-sized extracellular vesicles carry proteins, lipids, and nucleic acids, which can be taken up by local or distant cells, serving as mediators of intercellular communication and as drug delivery vehicles. Exosomes deliver molecules that reduce inflammation, renal fibrosis and extracellular matrix protein production, and promote tissue regeneration in animal models of kidney disease. Additionally, they have several advantages over stem cells, such as being non-immunogenic, having low risk of tumor formation, and being easier to produce and store. This review describes the use of natural and engineered exosomes containing therapeutic agents capable of mediating anti-inflammatory and anti-fibrotic processes during both acute kidney injury and chronic kidney disease. Exosome-based therapies will be compared with stem cell-based treatments for tissue regeneration, with a focus on renal protection. Finally, future directions and strategies for improving the therapeutic efficacy of exosomes are discussed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Baisong Lu
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Tracy Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
16
|
Nishimura K, Sanchez-Molano J, Kerr N, Pressman Y, Silvera R, Khan A, Gajavelli S, Bramlett HM, Dietrich WD. Beneficial Effects of Human Schwann Cell-Derived Exosomes in Mitigating Secondary Damage After Penetrating Ballistic-Like Brain Injury. J Neurotrauma 2024; 41:2395-2412. [PMID: 38445369 PMCID: PMC11631803 DOI: 10.1089/neu.2023.0650] [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] [Indexed: 03/07/2024] Open
Abstract
There is a growing body of evidence that the delivery of cell-derived exosomes normally involved in intracellular communication can reduce secondary injury mechanisms after brain and spinal cord injury and improve outcomes. Exosomes are nanometer-sized vesicles that are released by Schwann cells and may have neuroprotective effects by reducing post-traumatic inflammatory processes as well as promoting tissue healing and functional recovery. The purpose of this study was to evaluate the beneficial effects of human Schwann-cell exosomes (hSC-Exos) in a severe model of penetrating ballistic-like brain injury (PBBI) in rats and investigate effects on multiple outcomes. Human Schwann cell processing protocols followed Current Good Manufacturing Practices (cGMP) with exosome extraction and purification steps approved by the Food and Drug Administration for an expanded access single ALS patient Investigational New Drug. Anesthetized male Sprague-Dawley rats (280-350g) underwent PBBI surgery or Sham procedures and, starting 30 min after injury, received either a dose of hSC-Exos or phosphate-buffered saline through the jugular vein. At 48h after PBBI, flow cytometry analysis of cortical tissue revealed that hSC-Exos administration reduced the number of activated microglia and levels of caspase-1, a marker of inflammasome activation. Neuropathological analysis at 21 days showed that hSC-Exos treatment after PBBI significantly reduced overall contusion volume and decreased the frequency of Iba-1 positive activated and amoeboid microglia by immunocytochemical analysis. This study revealed that the systemic administration of hSC-Exos is neuroprotective in a model of severe TBI and reduces secondary inflammatory injury mechanisms and histopathological damage. The administration of hSC-Exos represents a clinically relevant cell-based therapy to limit the detrimental effects of neurotrauma or other progressive neurological injuries by impacting multiple pathophysiological events and promoting neurological recovery.
Collapse
Affiliation(s)
- Kengo Nishimura
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Juliana Sanchez-Molano
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nadine Kerr
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yelena Pressman
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Risset Silvera
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Aisha Khan
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Helen M. Bramlett
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| | - W. Dalton Dietrich
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
17
|
Pedersen C, Chen VT, Herbst P, Zhang R, Elfert A, Krishan A, Azar DT, Chang JH, Hu WY, Kremsmayer TP, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Target specification and therapeutic potential of extracellular vesicles for regulating corneal angiogenesis, lymphangiogenesis, and nerve repair. Ocul Surf 2024; 34:459-476. [PMID: 39426677 PMCID: PMC11921040 DOI: 10.1016/j.jtos.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Extracellular vesicles, including exosomes, are small extracellular vesicles that range in size from 30 nm to 10 μm in diameter and have specific membrane markers. They are naturally secreted and are present in various bodily fluids, including blood, urine, and saliva, and through the variety of their internal cargo, they contribute to both normal physiological and pathological processes. These processes include immune modulation, neuronal synapse formation, cell differentiation, cancer metastasis, angiogenesis, lymphangiogenesis, progression of infectious disease, and neurodegenerative disorders like Alzheimer's and Parkinson's disease. In recent years, interest has grown in the use of exosomes as a potential drug delivery system for various diseases and injuries. Importantly, exosomes originating from a patient's own cells exhibit minimal immunogenicity and possess remarkable stability along with inherent and adjustable targeting capabilities. This review explores the roles of exosomes in angiogenesis, lymphangiogenesis, and nerve repair with a specific emphasis on these processes within the cornea. Furthermore, it examines exosomes derived from specific cell types, discusses the advantages of exosome-based therapies in modulating these processes, and presents some of the most established methods for exosome isolation. Exosome-based treatments are emerging as potential minimally invasive and non-immunogenic therapies that modulate corneal angiogenesis and lymphangiogenesis, as well as enhance and accelerate endogenous corneal nerve repair.
Collapse
Affiliation(s)
- Cameron Pedersen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria T Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Runze Zhang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Amr Elfert
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tobias P Kremsmayer
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victor H Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
18
|
Zhang Y, Zhao Y, Zhang J, Gao Y, Gao X, Li S, Chang C, Yang G. Proteomics of plasma-derived extracellular vesicles reveals S100A8 as a novel biomarker for Alzheimer's disease: A preliminary study. J Proteomics 2024; 308:105279. [PMID: 39159863 DOI: 10.1016/j.jprot.2024.105279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024]
Abstract
Extracellular vesicles (EVs) act as mediators for intercellular transfer of Aβ and tau proteins, promoting the propagation of these pathological misfolded proteins throughout the brain in Alzheimer's disease (AD). Levels of blood exosomal Aβ42, total Tau (t-Tau) and phosphorylated Tau (p-Tau) had a high correlation with their concentrations in cerebrospinal fluid (CSF), demonstrating that exosomal biomarkers have equal contribution as those in CSF for the diagnosis of AD. We aimed to comprehensively characterize the proteome of plasma-derived EVs to identify differentially expressed proteins (DEPs) and pathways in AD. Tandem mass tag (TMT) labeled quantitative proteomics was applied to analyze plasma-derived EV proteins in 9 AD patients and 9 healthy controls. 335 proteins were quantified, and 12 DEPs were identified including seven upregulated proteins and five down-regulated proteins. Oligomerized Aβ1-42 induced SH-SY5Y cell damage model was built to mimic the pathological changes of AD, and small interfering RNA (siRNA) against S100A8 was used to knock down S100A8 expression. Results displayed S100A8 was down regulated in plasma-derived EVs from AD patients, while enriched in EVs derived from Aβ1-42-induced SH-SY5Y cells. Furthermore, Aβ1-42-induced SH-SY5Y cells treated with S100A8 siRNA showed decreased Aβ levels in cell lysate and EVs, especially in EVs. SIGNIFICANCE: The investigation aimed to comprehensively characterize the proteome of plasma-derived EVs to identify DEPs and potential biomarker of AD. S100A8 was found down regulated in plasma-derived EVs from AD patients using TMT labeled quantitative proteomics. The diagnostic value of S100A8 was also confirmed using receiver operating characteristic curve (ROC) analysis. Furthermore, Aβ1-42-induced SH-SY5Y cells treated with S100A8 siRNA showed decreased Aβ levels in cell lysate and EVs, especially in EVs. The preliminary findings suggest that suppression of S100A8 expression inhibits Aβ aggregation both in cell lysate and EVs from Aβ1-42-induced SH-SY5Y cells, and S100A8 more likely regulates Aβ aggregation via EVs. Therefore, plasma-derived EV S100A8 might be a potential biomarker of AD. Manipulation of S100A8 expression may be a novel therapeutic strategy in the treatment of AD.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuan Zhao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jian Zhang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ya Gao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xuan Gao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuyue Li
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cui Chang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guofeng Yang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
19
|
D'Souza RS, Her YF, Hussain N, Karri J, Schatman ME, Calodney AK, Lam C, Buchheit T, Boettcher BJ, Chang Chien GC, Pritzlaff SG, Centeno C, Shapiro SA, Klasova J, Grider JS, Hubbard R, Ege E, Johnson S, Epstein MH, Kubrova E, Ramadan ME, Moreira AM, Vardhan S, Eshraghi Y, Javed S, Abdullah NM, Christo PJ, Diwan S, Hassett LC, Sayed D, Deer TR. Evidence-Based Clinical Practice Guidelines on Regenerative Medicine Treatment for Chronic Pain: A Consensus Report from a Multispecialty Working Group. J Pain Res 2024; 17:2951-3001. [PMID: 39282657 PMCID: PMC11402349 DOI: 10.2147/jpr.s480559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Injectable biologics have not only been described and developed to treat dermal wounds, cardiovascular disease, and cancer, but have also been reported to treat chronic pain conditions. Despite emerging evidence supporting regenerative medicine therapy for pain, many aspects remain controversial. Methods The American Society of Pain and Neuroscience (ASPN) identified the educational need for an evidence-based guideline on regenerative medicine therapy for chronic pain. The executive board nominated experts spanning multiple specialties including anesthesiology, physical medicine and rehabilitation, and sports medicine based on expertise, publications, research, and clinical practice. A steering committee selected preliminary questions, which were reviewed and refined. Evidence was appraised using the United States Preventive Services Task Force (USPSTF) criteria for evidence level and degree of recommendation. Using a modified Delphi approach, consensus points were distributed to all collaborators and each collaborator voted on each point. If collaborators provided a decision of "disagree" or "abstain", they were invited to provide a rationale in a non-blinded fashion to the committee chair, who incorporated the respective comments and distributed revised versions to the committee until consensus was achieved. Results Sixteen questions were selected for guideline development. Questions that were addressed included type of injectable biologics and mechanism, evidence in treating chronic pain indications (eg, tendinopathy, muscular pathology, osteoarthritis, intervertebral disc disease, neuropathic pain), role in surgical augmentation, dosing, comparative efficacy between injectable biologics, peri-procedural practices to optimize therapeutic response and quality of injectate, federal regulations, and complications with mitigating strategies. Conclusion In well-selected individuals with certain chronic pain indications, use of injectable biologics may provide superior analgesia, functionality, and/or quality of life compared to conventional medical management or placebo. Future high-quality randomized clinical trials are warranted with implementation of minimum reporting standards, standardization of preparation protocols, investigation of dose-response associations, and comparative analysis between different injectable biologics.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yeng F Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Jay Karri
- Departments of Orthopedic Surgery and Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael E Schatman
- Department of Anesthesiology, Perioperative Care, & Pain Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Christopher Lam
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Thomas Buchheit
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Brennan J Boettcher
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Scott G Pritzlaff
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Sacramento, CA, USA
| | | | - Shane A Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jay S Grider
- Department of Anesthesiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ryan Hubbard
- Department of Sports Medicine, Anderson Orthopedic Clinic, Arlington, VA, USA
| | - Eliana Ege
- Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Shelby Johnson
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Max H Epstein
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Mohamed Ehab Ramadan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alexandra Michelle Moreira
- Department of Physical Medicine & Rehabilitation, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | - Swarnima Vardhan
- Department of Internal Medicine, Yale New Haven Health - Bridgeport Hospital, Bridgeport, CT, USA
| | - Yashar Eshraghi
- Department of Anesthesiology & Critical Care Medicine, Ochsner Health System, New Orleans, LA, USA
| | - Saba Javed
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Newaj M Abdullah
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Paul J Christo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sudhir Diwan
- Department of Pain Medicine, Advanced Spine on Park Avenue, New York City, NY, USA
| | | | - Dawood Sayed
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy R Deer
- Department of Anesthesiology and Pain Medicine, West Virginia University School of Medicine, Charleston, WV, USA
| |
Collapse
|
20
|
Wang L, Lu X, Szalad A, Liu XS, Zhang Y, Wang X, Golembieski WA, Powell B, Mccann M, Lu M, Chopp M, Zhang ZG. Schwann cell-derived exosomes ameliorate peripheral neuropathy induced by ablation of dicer in Schwann cells. Front Cell Neurosci 2024; 18:1462228. [PMID: 39285940 PMCID: PMC11402728 DOI: 10.3389/fncel.2024.1462228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Background MicroRNAs (miRNAs) in Schwann cells (SCs) mediate peripheral nerve function. Ablating Dicer, a key gene in miRNA biogenesis, in SCs causes peripheral neuropathy. Exosomes from healthy SCs (SC-Exo) ameliorate diabetic peripheral neuropathy in part via miRNAs. Thus, using transgenic mice with conditional and inducible ablation of Dicer in proteolipid protein (PLP) expressing SCs (PLP-cKO), we examined whether SC-Exo could reduce peripheral neuropathy in PLP-cKO mice. Methods PLP-cKO mice at the age of 16 weeks (8 week post-Tamoxifen) were randomly treated with SC-Exo or saline weekly for 8 weeks. Age-and sex-matched wild-type (WT) littermates were used as controls. Peripheral neurological functions, sciatic nerve integrity, and myelination were analyzed. Quantitative RT-PCR and Western blot analyses were performed to examine miRNA and protein expression in sciatic nerve tissues, respectively. Results Compared to the WT mice, PLP-cKO mice exhibited a significant decrease in motor and sensory conduction velocities, thermal sensitivity, and motor coordination. PLP-cKO mice exhibited substantial demyelination and axonal damage of the sciatic nerve. Treatment of PLP-cKO mice with SC-Exo significantly ameliorated the peripheral neuropathy and sciatic nerve damage. PLP-cKO mice showed a substantial reduction in a set of Dicer-related miRNAs known to regulate myelination, axonal integrity, and inflammation such as miR-138, -146a and - 338 in the sciatic nerve. In addition, PLP-cKO mice exhibited significant reduction of myelin forming proteins, early growth response 2 (EGR2) and sex determining region Y-box10 (Sox10), but significantly increased myelination inhibitors, Notch1, c-Jun, and Sox2 and the axonal growth inhibitor phosphatase and tens in homolog (PTEN). However, SC-Exo treatment reversed the PLP-cKO altered miRNAs and proteins. Conclusion This study demonstrates that exogenous SC-Exo ameliorate peripheral neuropathy induced by Dicer ablation in PLP expressing SCs. The therapeutic benefit may be mediated by the SC-Exo altered miRNAs and their targeted genes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - XueRong Lu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Xian Shuang Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mikkala Mccann
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
21
|
Kajitani GS, Xavier G, Villena-Rueda BE, Karia BTR, Santoro ML. Extracellular vesicles in neurodegenerative, mental, and other neurological disorders: Perspectives into mechanisms, biomarker potential, and therapeutic implications. CURRENT TOPICS IN MEMBRANES 2024; 94:299-336. [PMID: 39370211 DOI: 10.1016/bs.ctm.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Extracellular vesicles (EVs) are produced, secreted, and targeted by most human cells, including cells that compose nervous system tissues. EVs carry several types of biomolecules, such as lipids, proteins and microRNA, and can function as signaling agents in physiological and pathological processes. In this chapter, we will focus on EVs and their cargo secreted by brain cells, especially neurons and glia, and how these aspects are affected in pathological conditions. The chapter covers neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis, as well as several psychiatric disorders, namely schizophrenia, autism spectrum disorder and major depressive disorder. This chapter also addresses other types of neurological dysfunctions, epilepsy and traumatic brain injury. EVs can cross the blood brain barrier, and thus brain EVs may be detected in more accessible peripheral tissue, such as circulating blood. Alterations in EV composition and contents can therefore impart valuable clues into the molecular etiology of these disorders, and serve biomarkers regarding disease prevalence, progression and treatment. EVs can also be used to carry drugs and biomolecules into brain tissue, considered as a promising drug delivery agent for neurological diseases. Therefore, although this area of research is still in its early development, it offers great potential in further elucidating and in treating neurological disorders.
Collapse
Affiliation(s)
- Gustavo Satoru Kajitani
- Department of Morphology and Genetics, Universidade Federal de São Paulo, Brazil; Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil
| | - Gabriela Xavier
- Center for Genomic Medicine, Massachusetts General Hospital, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, United States
| | - Beatriz Enguidanos Villena-Rueda
- Department of Morphology and Genetics, Universidade Federal de São Paulo, Brazil; Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil
| | - Bruno Takao Real Karia
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil; Department of Biochemistry, Universidade Federal de São Paulo, Brazil
| | - Marcos Leite Santoro
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil; Department of Biochemistry, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
22
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
23
|
Errante EL, Smartz T, Costello MC, Schaeffer EA, Kloehn AJ, Yunga Tigre J, Khan A, Pressman Y, Levi AD, Burks SS. Assessing Survival, Distribution, and Optimal Loading Technique of Schwann Cell-Derived Exosomes Into Second-generation Axon Guidance Channels. Mil Med 2024; 189:63-66. [PMID: 39160822 DOI: 10.1093/milmed/usae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/12/2023] [Accepted: 02/06/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Peripheral nerve injury (PNI) occurs in approximately 3% of all trauma patients and can be challenging to treat, particularly when injury is severe such as with a long-segmental gap. Although peripheral nerves can regenerate after injury, functional recovery is often insufficient, leading to deficits in the quality of life of patients with PNI. Although nerve autografts are the gold standard of care, there are several disadvantages to their use, namely a lack of autologous nerve material for repair. This has led to the pursuit of alternative treatment methods such as axon guidance channels (AGCs). Second-generation AGCs have been shown to be able to deliver growth-enhancing substrates for nerve repair directly to the injury site. Although our laboratory has had success with second-generation AGCs filled with Schwann cells (SCs), SCs have their own set of issues clinically. Because of this, we have begun to utilize SC-derived exosomes as an alternative, as they have the appropriate protein markers, associate to axons in high concentrations, and are able to improve nerve regeneration. However, it is unknown how SC-derived exosomes may react within second-generation AGCs; thus, the aim of the present study was to assess the ability of SC-derived exosomes to be loaded into a second-generation AGC and how they would distribute within it. MATERIALS AND METHODS A total of 4 dry second-generation AGCs were loaded with SC-derived exosomes that were derived from green fluorescent protein (GFP)-labeled SCs. They were subsequently frozen and sliced before imaging. RESULTS Here, we present findings that SC-derived exosomes can be loaded into second-generation AGCs through our established loading method utilizing negative pressure and are able to survive and equally distribute along the length of the AGC. CONCLUSIONS Although only 4 second-generation AGCs were utilized, these findings indicate a potential use for SC-derived exosomes within second-generation AGCs to treat severe PNI. Future research should focus on exploring this in greater detail and in different contexts to assess the ability of SC-derived exosomes to survive at the site of injury and treat PNI.
Collapse
Affiliation(s)
- Emily L Errante
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Taylor Smartz
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Meredith C Costello
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ericka A Schaeffer
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew J Kloehn
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joseph Yunga Tigre
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Aisha Khan
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yelena Pressman
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - S Shelby Burks
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
24
|
Rasouli A, Roshangar L, Hosseini M, Pourmohammadfazel A, Nikzad S. Beyond boundaries: The therapeutic potential of exosomes in neural microenvironments in neurological disorders. Neuroscience 2024; 553:98-109. [PMID: 38964450 DOI: 10.1016/j.neuroscience.2024.06.031] [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: 12/30/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Neurological disorders are a diverse group of conditions that can significantly impact individuals' quality of life. The maintenance of neural microenvironment homeostasis is essential for optimal physiological cellular processes. Perturbations in this delicate balance underlie various pathological manifestations observed across various neurological disorders. Current treatments for neurological disorders face substantial challenges, primarily due to the formidable blood-brain barrier and the intricate nature of neural tissue structures. These obstacles have resulted in a paucity of effective therapies and inefficiencies in patient care. Exosomes, nanoscale vesicles that contain a complex repertoire of biomolecules, are identifiable in various bodily fluids. They hold substantial promise in numerous therapeutic interventions due to their unique attributes, including targeted drug delivery mechanisms and the ability to cross the BBB, thereby enhancing their therapeutic potential. In this review, we investigate the therapeutic potential of exosomes across a range of neurological disorders, including neurodegenerative disorders, traumatic brain injury, peripheral nerve injury, brain tumors, and stroke. Through both in vitro and in vivo studies, our findings underscore the beneficial influence of exosomes in enhancing the neural microenvironment following neurological diseases, offering promise for improved neural recovery and management in these conditions.
Collapse
Affiliation(s)
- Arefe Rasouli
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammadbagher Hosseini
- Department of Pediatrics, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pourmohammadfazel
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
25
|
Arbade G, Jose JV, Gulbake A, Kadam S, Kashte SB. From stem cells to extracellular vesicles: a new horizon in tissue engineering and regenerative medicine. Cytotechnology 2024; 76:363-401. [PMID: 38933869 PMCID: PMC11196501 DOI: 10.1007/s10616-024-00631-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 04/07/2024] [Indexed: 06/28/2024] Open
Abstract
In the fields of tissue engineering and regenerative medicine, extracellular vesicles (EVs) have become viable therapeutic tools. EVs produced from stem cells promote tissue healing by regulating the immune system, enhancing cell proliferation and aiding remodeling processes. Recently, EV has gained significant attention from researchers due to its ability to treat various diseases. Unlike stem cells, stem cell-derived EVs show lower immunogenicity, are less able to overcome biological barriers, and have a higher safety profile. This makes the use of EVs derived from cell-free stem cells a promising alternative to whole-cell therapy. This review focuses on the biogenesis, isolation, and characterization of EVs and highlights their therapeutic potential for bone fracture healing, wound healing, and neuronal tissue repair and treatment of kidney and intestinal diseases. Additionally, this review discusses the potential of EVs for the treatment of cancer, COVID-19, and HIV. In summary, the use of EVs derived from stem cells offers a new horizon for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER G), Guwahati, Assam 781101 India
| | - Sachin Kadam
- Sophisticated Analytical and Technical Help Institute, Indian Institute of Technology, Delhi, New Delhi 110016 India
| | - Shivaji B. Kashte
- Department of Stem Cell and Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to be University), Kolhapur, MS 416006 India
| |
Collapse
|
26
|
Liao S, Chen Y, Luo Y, Zhang M, Min J. The phenotypic changes of Schwann cells promote the functional repair of nerve injury. Neuropeptides 2024; 106:102438. [PMID: 38749170 DOI: 10.1016/j.npep.2024.102438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/17/2024]
Abstract
Functional recovery after nerve injury is a significant challenge due to the complex nature of nerve injury repair and the non-regeneration of neurons. Schwann cells (SCs), play a crucial role in the nerve injury repair process because of their high plasticity, secretion, and migration abilities. Upon nerve injury, SCs undergo a phenotypic change and redifferentiate into a repair phenotype, which helps in healing by recruiting phagocytes, removing myelin fragments, promoting axon regeneration, and facilitating myelin formation. However, the repair phenotype can be unstable, limiting the effectiveness of the repair. Recent research has found that transplantation of SCs can be an effective treatment option, therefore, it is essential to comprehend the phenotypic changes of SCs and clarify the related mechanisms to develop the transplantation therapy further.
Collapse
Affiliation(s)
- Shufen Liao
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Yan Chen
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Yin Luo
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Mengqi Zhang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Jun Min
- Neurology Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
27
|
Ahmed LA, Al-Massri KF. Exploring the Role of Mesenchymal Stem Cell-Derived Exosomes in Diabetic and Chemotherapy-Induced Peripheral Neuropathy. Mol Neurobiol 2024; 61:5916-5927. [PMID: 38252384 PMCID: PMC11249772 DOI: 10.1007/s12035-024-03916-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 12/31/2023] [Indexed: 01/23/2024]
Abstract
Diabetic and chemotherapy-induced peripheral neuropathies are known for long-term complications that are associated with uncontrolled hyperglycemia and cancer treatment, respectively. Peripheral neuropathy often requires long-term therapy and could persist after treatment provoking detrimental effects on the patient's quality of life. Despite continuous drug discoveries, development of efficient therapies is still needed for the significant management of diabetic and chemotherapy-induced peripheral neuropathy. Exosomes are nanosized extracellular vesicles that show great promise recently in tissue regeneration and injury repair compared to their parent stem cells. Herein, we provided a summary for the use of mesenchymal stem cell-derived exosomes in diabetic and chemotherapy-induced peripheral neuropathy in addition to recent advancements and ways proposed for the enhancement of their efficacy in these diseases.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt.
| | - Khaled F Al-Massri
- Department of Pharmacy and Biotechnology, Faculty of Medicine and Health Sciences, University of Palestine, Gaza, Palestine
| |
Collapse
|
28
|
Xin GD, Liu XY, Fan XD, Zhao GJ. Exosomes repairment for sciatic nerve injury: a cell-free therapy. Stem Cell Res Ther 2024; 15:214. [PMID: 39020385 PMCID: PMC11256477 DOI: 10.1186/s13287-024-03837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Sciatic nerve injury (SNI) is a common type of peripheral nerve injury typically resulting from trauma, such as contusion, sharp force injuries, drug injections, pelvic fractures, or hip dislocations. It leads to both sensory and motor dysfunctions, characterized by pain, numbness, loss of sensation, muscle atrophy, reduced muscle tone, and limb paralysis. These symptoms can significantly diminish a patient's quality of life. Following SNI, Wallerian degeneration occurs, which activates various signaling pathways, inflammatory factors, and epigenetic regulators. Despite the availability of several surgical and nonsurgical treatments, their effectiveness remains suboptimal. Exosomes are extracellular vesicles with diameters ranging from 30 to 150 nm, originating from the endoplasmic reticulum. They play a crucial role in facilitating intercellular communication and have emerged as highly promising vehicles for drug delivery. Increasing evidence supports the significant potential of exosomes in repairing SNI. This review delves into the pathological progression of SNI, techniques for generating exosomes, the molecular mechanisms behind SNI recovery with exosomes, the effectiveness of combining exosomes with other approaches for SNI repair, and the changes and future outlook for utilizing exosomes in SNI recovery.
Collapse
Affiliation(s)
- Guang-Da Xin
- Nephrology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China
| | - Xue-Yan Liu
- Cardiology Department, China-Japan Union Hospital of Jilin Universit, Changchun, Jilin Province, 130000, China
| | - Xiao-Di Fan
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China
| | - Guan-Jie Zhao
- Nephrology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130000, China.
| |
Collapse
|
29
|
Zhu F, Wang T, Wang G, Yan C, He B, Qiao B. The Exosome-Mediated Bone Regeneration: An Advanced Horizon Toward the Isolation, Engineering, Carrying Modalities, and Mechanisms. Adv Healthc Mater 2024; 13:e2400293. [PMID: 38426417 DOI: 10.1002/adhm.202400293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Exosomes, nanoparticles secreted by various cells, composed of a bilayer lipid membrane, and containing bioactive substances such as proteins, nucleic acids, metabolites, etc., have been intensively investigated in tissue engineering owing to their high biocompatibility and versatile biofunction. However, there is still a lack of a high-quality review on bone defect regeneration potentiated by exosomes. In this review, the biogenesis and isolation methods of exosomes are first introduced. More importantly, the engineered exosomes of the current state of knowledge are discussed intensively in this review. Afterward, the biomaterial carriers of exosomes and the mechanisms of bone repair elucidated by compelling evidence are presented. Thus, future perspectives and concerns are revealed to help devise advanced modalities based on exosomes to overcome the challenges of bone regeneration. It is totally believed this review will attract special attention from clinicians and provide promising ideas for their future works.
Collapse
Affiliation(s)
- Fukang Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Taiyou Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Guangjian Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Department of Orthopaedics, The People's Hospital of Rongchang District, Chongqing, 402460, P. R. China
| | - Caiping Yan
- Department of Orthopaedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Bin He
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Bo Qiao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400010, P. R. China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400010, P. R. China
| |
Collapse
|
30
|
Saijilafu, Ye LC, Zhang JY, Xu RJ. The top 100 most cited articles on axon regeneration from 2003 to 2023: a bibliometric analysis. Front Neurosci 2024; 18:1410988. [PMID: 38988773 PMCID: PMC11233811 DOI: 10.3389/fnins.2024.1410988] [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: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
Objective In this study, we used a bibliometric and visual analysis to evaluate the characteristics of the 100 most cited articles on axon regeneration. Methods The 100 most cited papers on axon regeneration published between 2003 and 2023 were identified by searching the Web of Science Core Collection database. The extracted data included the title, author, keywords, journal, publication year, country, and institution. A bibliometric analysis was subsequently undertaken. Results The examined set of 100 papers collectively accumulated a total of 39,548 citations. The number of citations for each of the top 100 articles ranged from 215 to 1,604, with a median value of 326. The author with the most contributions to this collection was He, Zhigang, having authored eight papers. Most articles originated in the United States (n = 72), while Harvard University was the institution with the most cited manuscripts (n = 19). Keyword analysis unveiled several research hotspots, such as chondroitin sulfate proteoglycan, alternative activation, exosome, Schwann cells, axonal protein synthesis, electrical stimulation, therapeutic factors, and remyelination. Examination of keywords in the articles indicated that the most recent prominent keyword was "local delivery." Conclusion This study offers bibliometric insights into axon regeneration, underscoring that the United States is a prominent leader in this field. Our analysis highlights the growing relevance of local delivery systems in axon regeneration. Although these systems have shown promise in preclinical models, challenges associated with long-term optimization, agent selection, and clinical translation remain. Nevertheless, the continued development of local delivery technologies represents a promising pathway for achieving axon regeneration; however, additional research is essential to fully realize their potential and thereby enhance patient outcomes.
Collapse
Affiliation(s)
- Saijilafu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ling-Chen Ye
- Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jing-Yu Zhang
- Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Ren-Jie Xu
- Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
31
|
Stassart RM, Gomez-Sanchez JA, Lloyd AC. Schwann Cells as Orchestrators of Nerve Repair: Implications for Tissue Regeneration and Pathologies. Cold Spring Harb Perspect Biol 2024; 16:a041363. [PMID: 38199866 PMCID: PMC11146315 DOI: 10.1101/cshperspect.a041363] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.
Collapse
Affiliation(s)
- Ruth M Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
- Instituto de Neurociencias CSIC-UMH, Sant Joan de Alicante 03550, Spain
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
32
|
Rust R, Holm MM, Egger M, Weinmann O, van Rossum D, Walter FR, Santa-Maria AR, Grönnert L, Maurer MA, Kraler S, Akhmedov A, Cideciyan R, Lüscher TF, Deli MA, Herrmann IK, Schwab ME. Nogo-A is secreted in extracellular vesicles, occurs in blood and can influence vascular permeability. J Cereb Blood Flow Metab 2024; 44:938-954. [PMID: 38000040 PMCID: PMC11318402 DOI: 10.1177/0271678x231216270] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023]
Abstract
Nogo-A is a transmembrane protein with multiple functions in the central nervous system (CNS), including restriction of neurite growth and synaptic plasticity. Thus far, Nogo-A has been predominantly considered a cell contact-dependent ligand signaling via cell surface receptors. Here, we show that Nogo-A can be secreted by cultured cells of neuronal and glial origin in association with extracellular vesicles (EVs). Neuron- and oligodendrocyte-derived Nogo-A containing EVs inhibited fibroblast spreading, and this effect was partially reversed by Nogo-A receptor S1PR2 blockage. EVs purified from HEK cells only inhibited fibroblast spreading upon Nogo-A over-expression. Nogo-A-containing EVs were found in vivo in the blood of healthy mice and rats, as well as in human plasma. Blood Nogo-A concentrations were elevated after acute stroke lesions in mice and rats. Nogo-A active peptides decreased barrier integrity in an in vitro blood-brain barrier model. Stroked mice showed increased dye permeability in peripheral organs when tested 2 weeks after injury. In the Miles assay, an in vivo test to assess leakage of the skin vasculature, a Nogo-A active peptide increased dye permeability. These findings suggest that blood borne, possibly EV-associated Nogo-A could exert long-range regulatory actions on vascular permeability.
Collapse
Affiliation(s)
- Ruslan Rust
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| | - Mea M Holm
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Matteo Egger
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | | | | | - Fruzsina R Walter
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | | | - Lisa Grönnert
- Brain Research Institute, University of Zürich, Switzerland
| | | | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | | | - Rose Cideciyan
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Maria A Deli
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | - Inge K Herrmann
- Particles Biology Interactions Laboratory, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, Switzerland
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| |
Collapse
|
33
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
34
|
Sun J, Zeng Q, Wu Z, Li Z, Gao Q, Liao Z, Li H, Ling C, Chen C, Wang H, Zhang B. Enhancing intraneural revascularization following peripheral nerve injury through hypoxic Schwann-cell-derived exosomes: an insight into endothelial glycolysis. J Nanobiotechnology 2024; 22:283. [PMID: 38789980 PMCID: PMC11127458 DOI: 10.1186/s12951-024-02536-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Endothelial cell (EC)-driven intraneural revascularization (INRV) and Schwann cells-derived exosomes (SCs-Exos) both play crucial roles in peripheral nerve injury (PNI). However, the interplay between them remains unclear. We aimed to elucidate the effects and underlying mechanisms of SCs-Exos on INRV following PNI. RESULTS We found that GW4869 inhibited INRV, as well as that normoxic SCs-Exos (N-SCs-Exos) exhibited significant pro-INRV effects in vivo and in vitro that were potentiated by hypoxic SCs-Exos (H-SCs-Exos). Upregulation of glycolysis emerged as a pivotal factor for INRV after PNI, as evidenced by the observation that 3PO administration, a glycolytic inhibitor, inhibited the INRV process in vivo and in vitro. H-SCs-Exos more significantly enhanced extracellular acidification rate/oxygen consumption rate ratio, lactate production, and glycolytic gene expression while simultaneously suppressing acetyl-CoA production and pyruvate dehydrogenase E1 subunit alpha (PDH-E1α) expression than N-SCs-Exos both in vivo and in vitro. Furthermore, we determined that H-SCs-Exos were more enriched with miR-21-5p than N-SCs-Exos. Knockdown of miR-21-5p significantly attenuated the pro-glycolysis and pro-INRV effects of H-SCs-Exos. Mechanistically, miR-21-5p orchestrated EC metabolism in favor of glycolysis by targeting von Hippel-Lindau/hypoxia-inducible factor-1α and PDH-E1α, thereby enhancing hypoxia-inducible factor-1α-mediated glycolysis and inhibiting PDH-E1α-mediated oxidative phosphorylation. CONCLUSION This study unveiled a novel intrinsic mechanism of pro-INRV after PNI, providing a promising therapeutic target for post-injury peripheral nerve regeneration and repair.
Collapse
Affiliation(s)
- Jun Sun
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China
| | - Qiuhua Zeng
- Department of Radiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Zhimin Wu
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China
| | - Zhangyu Li
- Department of Neurosurgery, School of Medicine, the First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361102, China
| | - Qun Gao
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhi Men South St, Beijing, 100044, China
| | - Zhi Liao
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China
| | - Hao Li
- Department of Neurosurgery, Guangzhou Panyu Central Hospital, No.8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong, PR China
| | - Cong Ling
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China
| | - Chuan Chen
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China.
| | - Hui Wang
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China.
| | - Baoyu Zhang
- Department of Neurosurgery, the Third Affiliated Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, 510630, PR China
| |
Collapse
|
35
|
Zhang X, Zhang W, Sun H, Wang H. The effects of exosomes originating from different cell sources on the differentiation of bone marrow mesenchymal stem cells into schwann cells. J Nanobiotechnology 2024; 22:220. [PMID: 38698449 PMCID: PMC11067703 DOI: 10.1186/s12951-024-02450-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) can differentiate into Schwann cells (SCs) during peripheral nerve injury; in our previous research, we showed that SC-derived exosomes (SC-exos) played a direct induction role while fibroblast-derived exosomes (Fb-exos) had no obvious induction role. The induction role of neural stem cell (NSC)-derived exosomes (NSC-exos) has also been widely confirmed. However, no studies have compared the induction effects of these three types of cells at the same time. Therefore, by investigating the effect of these three cell-derived exosomes upon the induction of BMSCs to differentiate into SCs, this study explored the role of different exosomes in promoting the differentiation of stem cells into SCs cells, and conducted a comparison between the two groups by RNA sequencing to further narrow the range of target genes and related gene pathways in order to study their related mechanisms. MATERIALS AND METHODS We extracted exosomes from SCs, fibroblasts (Fb) and neural stem cells (NSC) and then investigated the ability of these exosomes to induce differentiation into BMSCs under different culture conditions. The expression levels of key proteins and gene markers were detected in induced cells by fluorescence immunoassays, western blotting and polymerase chain reaction (PCR); then, we statistically compared the relative induction effects under different conditions. Finally, we analyzed the three types of exosomes by RNA-seq to predict target genes and related gene pathways. RESULTS BMSCs were cultured by three media: conventional (no induction), pre-induction or pre-induction + original induction medium (ODM) with exosomes of the same cell origin under different culture conditions. When adding the three different types of exosomes separately, the overall induction of BMSCs to differentiate into SCs was significantly increased (P < 0.05). The induction ability was ranked as follows: pre-induction + ODM + exosome group > pre-induction + exosome group > non-induction + exosome group. Using exosomes from different cell sources under the same culture conditions, we observed the following trends under the three culture conditions: RSC96-exos group ≥ NSC-exos group > Fb-exos group. The overall ability to induce BMSCs into SCs was significantly greater in the RSC96-exos group and the NSC-exos group. Although there was no significant difference in induction efficiency when comparing these two groups, the overall induction ability of the RSC96-exos group was slightly higher than that of the NSC-exos group. By combining the differentiation induction results with the RNA-seq data, the three types of exosomes were divided into three comparative groups: RSC vs. NSC, RSC vs. Fb and NSC vs. Fb. We identified 203 differentially expressed mRNA target genes in these three groups. Two differentially expressed genes were upregulated simultaneously, namely riboflavin kinase (RFK, ENSRNOG00000022273) and ribosomal RNA processing 36 (Rrp36, ENSRNOG00000017836). We did not identify any co-upregulated target genes for the miRNAs, but did identify one target gene of the lncRNAs, namely ENSRNOG00000065005. Analysis identified 90 GO terms related to nerves and axons in the mRNAs; in addition, KEGG enrichment and GASA analysis identified 13 common differential expression pathways in the three groups. CONCLUSIONS Our analysis found that pre-induction + ODM + RSC96/NSC-exos culture conditions were most conducive with regards to induction and differentiation. RSC96-exos and NSC-exos exhibited significantly greater differentiation efficiency of BMSCs into SCs. Although there was no statistical difference, the data indicated a trend for RSC96-exos to be advantageous We identified 203 differentially expressed mRNAs between the three groups and two differentially expressed target mRNAs were upregulated, namely riboflavin kinase (RFK, ENSRNOG00000022273) and ribosomal RNA processing 36 (Rrp36, ENSRNOG00000017836). 90 GO terms were related to nerves and axons. Finally, we identified 13 common differentially expressed pathways across our three types of exosomes. It is hoped that the efficiency of BMSCs induction differentiation into SCs can be improved, bringing hope to patients and more options for clinical treatment.
Collapse
Affiliation(s)
- Xianxiang Zhang
- Department of Otolaryngology and Head and Neck Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101101, China
| | - Weiwei Zhang
- Department of Otolaryngology and Head and Neck Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101101, China
| | - Hao Sun
- Department of Otolaryngology and Head and Neck Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101101, China
| | - Hui Wang
- Department of Otolaryngology and Head and Neck Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101101, China.
| |
Collapse
|
36
|
Shurin MR, Wheeler SE, Shurin GV, Zhong H, Zhou Y. Schwann cells in the normal and pathological lung microenvironment. Front Mol Biosci 2024; 11:1365760. [PMID: 38638689 PMCID: PMC11024312 DOI: 10.3389/fmolb.2024.1365760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
The lungs are a key organ in the respiratory system. They are regulated by a complex network of nerves that control their development, structure, function, and response to various pathological stimuli. Accumulating evidence suggests the involvement of a neural mechanism in different pathophysiological conditions in the lungs and the development and progression of common respiratory diseases. Lung diseases are the chief source of death globally. For instance, lung cancer is the second most commonly diagnosed malignancy, after prostate cancer in men and breast cancer in women, and is the most lethal cancer worldwide. However, although airway nerves are accepted as a mechanistically and therapeutically important feature that demands appropriate emphasizing in the context of many respiratory diseases, significantly less is known about the role of the neuroglial cells in lung physiology and pathophysiology, including lung cancer. New data have uncovered some cellular and molecular mechanisms of how Schwann cells, as fundamental components of the peripheral nervous system, may regulate lung cancer cells' survival, spreading, and invasiveness in vitro and in vivo. Schwann cells control the formation and maintenance of the lung cancer microenvironment and support metastasis formation. It was also reported that the number of lung cancer-associated Schwann cells correlates with patients' survival. Different factors secreted by Schwann cells, including microRNA, are known to sharpen the lung cancer environment by regulating the tumor-neuro-immune axis. Further clinical and experimental studies are required to elucidate the detailed role of Schwann cells in creating and maintaining pulmonary tumor-neuro-immune axis, which will advance our understanding of the pathogenesis of lung cancer and may inform therapeutic hypotheses aiming neoplasms and metastases in the lung.
Collapse
Affiliation(s)
- Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Sarah E. Wheeler
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Galina V. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Liu B, Alimi OA, Wang Y, Kong Y, Kuss M, Krishnan MA, Hu G, Xiao Y, Dong J, DiMaio DJ, Duan B. Differentiated mesenchymal stem cells-derived exosomes immobilized in decellularized sciatic nerve hydrogels for peripheral nerve repair. J Control Release 2024; 368:24-41. [PMID: 38367864 PMCID: PMC11411504 DOI: 10.1016/j.jconrel.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Peripheral nerve injury (PNI) and the limitations of current treatments often result in incomplete sensory and motor function recovery, which significantly impact the patient's quality of life. While exosomes (Exo) derived from stem cells and Schwann cells have shown promise on promoting PNI repair following systemic administration or intraneural injection, achieving effective local and sustained Exo delivery holds promise to treat local PNI and remains challenging. In this study, we developed Exo-loaded decellularized porcine nerve hydrogels (DNH) for PNI repair. We successfully isolated Exo from differentiated human adipose-derived mesenchymal stem cells (hADMSC) with a Schwann cell-like phenotype (denoted as dExo). These dExo were further combined with polyethylenimine (PEI), and DNH to create polyplex hydrogels (dExo-loaded pDNH). At a PEI content of 0.1%, pDNH showed cytocompatibility for hADMSCs and supported neurite outgrowth of dorsal root ganglions. The sustained release of dExos from dExo-loaded pDNH persisted for at least 21 days both in vitro and in vivo. When applied around injured nerves in a mouse sciatic nerve crush injury model, the dExo-loaded pDNH group significantly improved sensory and motor function recovery and enhanced remyelination compared to dExo and pDNH only groups, highlighting the synergistic regenerative effects. Interestingly, we observed a negative correlation between the number of colony-stimulating factor-1 receptor (CSF-1R) positive cells and the extent of PNI regeneration at the 21-day post-surgery stage. Subsequent in vitro experiments demonstrated the potential involvement of the CSF-1/CSF-1R axis in Schwann cells and macrophage interaction, with dExo effectively downregulating CSF-1/CSF-1R signaling.
Collapse
Affiliation(s)
- Bo Liu
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Olawale A Alimi
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yanfei Wang
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mena Asha Krishnan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| |
Collapse
|
38
|
Zhou Y, Zhang Y, Xu J, Wang Y, Yang Y, Wang W, Gu A, Han B, Shurin GV, Zhong R, Shurin MR, Zhong H. Schwann cell-derived exosomes promote lung cancer progression via miRNA-21-5p. Glia 2024; 72:692-707. [PMID: 38192185 DOI: 10.1002/glia.24497] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Schwann cells (SCs), the primary glial cells of the peripheral nervous system, which have been identified in many solid tumors, play an important role in cancer development and progression by shaping the tumor immunoenvironment and supporting the development of metastases. Using different cellular, molecular, and genetic approaches with integrated bioinformatics analysis and functional assays, we revealed the role of human SC-derived exosomal miRNAs in lung cancer progression in vitro and in vivo. We found that exosomal miRNA-21 from SCs up-regulated the proliferation, motility, and invasiveness of human lung cancer cells in vitro, which requires functional Rab small GTPases Rab27A and Rab27B in SCs for exosome release. We also revealed that SC exosomal miRNA-21-5p regulated the functional activation of tumor cells by targeting metalloprotease inhibitor RECK in tumor cells. Integrated bioinformatic analyses showed that hsa-miRNA-21-5p is associated with poor prognosis in patients with lung adenocarcinoma and can promote lung cancer progression through multiple signaling pathways including the MAPK, PI3K/Akt, and TNF signaling. Furthermore, in mouse xenograft models, SC exosomes and SC exosomal hsa-miRNA-21-5p augmented human lung cancer cell growth and lymph node metastasis in vivo. Together our data revealed, for the first time, that SC-secreted exosomes and exosomal miRNA-21-5p promoted the proliferation, motility, and spreading of human lung cancer cells in vitro and in vivo. Thus, exosomal miRNA-21 may play an oncogenic role in SC-accelerated progression of lung cancer and this pathway may serve as a new therapeutic target for further evaluation.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Yang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weimin Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiqin Gu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baohui Han
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Caamaño D, Cabezas J, Aguilera C, Martinez I, Wong YS, Sagredo DS, Ibañez B, Rodriguez S, Castro FO, Rodriguez-Alvarez L. DNA Content in Embryonic Extracellular Vesicles Is Independent of the Apoptotic Rate in Bovine Embryos Produced In Vitro. Animals (Basel) 2024; 14:1041. [PMID: 38612280 PMCID: PMC11011075 DOI: 10.3390/ani14071041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Pre-implantation embryos release extracellular vesicles containing different molecules, including DNA. The presence of embryonic DNA in E-EVs released into the culture medium during in vitro embryo production could be useful for genetic diagnosis. However, the vesicles containing DNA might be derived from embryos suffering from apoptosis, i.e., embryos of bad quality. This work intended to confirm that embryos release DNA that is useful for genotyping by evaluating the effect of embryonic apoptosis on DNA content in E-EVs. Bovine embryos were produced by parthenogenesis and in vitro fertilization (IVF). On Day 5, morulae were transferred to individual cultures in an EV-depleted SOF medium. On Day 7, embryos were used to evaluate cellular apoptosis, and each culture medium was collected to evaluate E-EV concentration, characterization, and DNA quantification. While no effect of the origin of the embryo on the apoptotic rate was found, arrested morulae had a higher apoptotic rate. E-EVs containing DNA were identified in all samples, and the concentration of those vesicles was not affected by the origin or quality of the embryos. However, the concentration of DNA was higher in EVs released by the arrested parthenogenetic embryos. There was a correlation between the concentration of E-EVs, the concentration of DNA-positive E-EVs, and the concentration of DNA. There was no negative effect of apoptotic rate on DNA-positive E-EVs and DNA concentration; however, embryos of the best quality with a low apoptotic rate still released EVs containing DNA. This study confirms that the presence of DNA in E-EVs is independent of embryo quality. Therefore, E-EVs could be used in liquid biopsy for noninvasive genetic diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Lleretny Rodriguez-Alvarez
- Laboratory of Animal Biotechnology, Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Mendez 595, Chillán 3780000, Chile; (D.C.); (J.C.); (C.A.); (I.M.); (Y.S.W.); (D.S.S.); (B.I.); (S.R.); (F.O.C.)
| |
Collapse
|
40
|
Izhiman Y, Esfandiari L. Emerging role of extracellular vesicles and exogenous stimuli in molecular mechanisms of peripheral nerve regeneration. Front Cell Neurosci 2024; 18:1368630. [PMID: 38572074 PMCID: PMC10989355 DOI: 10.3389/fncel.2024.1368630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Peripheral nerve injuries lead to significant morbidity and adversely affect quality of life. The peripheral nervous system harbors the unique trait of autonomous regeneration; however, achieving successful regeneration remains uncertain. Research continues to augment and expedite successful peripheral nerve recovery, offering promising strategies for promoting peripheral nerve regeneration (PNR). These include leveraging extracellular vesicle (EV) communication and harnessing cellular activation through electrical and mechanical stimulation. Small extracellular vesicles (sEVs), 30-150 nm in diameter, play a pivotal role in regulating intercellular communication within the regenerative cascade, specifically among nerve cells, Schwann cells, macrophages, and fibroblasts. Furthermore, the utilization of exogenous stimuli, including electrical stimulation (ES), ultrasound stimulation (US), and extracorporeal shock wave therapy (ESWT), offers remarkable advantages in accelerating and augmenting PNR. Moreover, the application of mechanical and electrical stimuli can potentially affect the biogenesis and secretion of sEVs, consequently leading to potential improvements in PNR. In this review article, we comprehensively delve into the intricacies of cell-to-cell communication facilitated by sEVs and the key regulatory signaling pathways governing PNR. Additionally, we investigated the broad-ranging impacts of ES, US, and ESWT on PNR.
Collapse
Affiliation(s)
- Yara Izhiman
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Electrical and Computer Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
41
|
Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, Xu C, Cao M, Chen C, Xu S, Zhang Z, Li M, Zhu Y, Peng N. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release 2024; 367:265-282. [PMID: 38253204 DOI: 10.1016/j.jconrel.2024.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Peripheral nerve injury (PNI) remains a severe clinical problem with debilitating consequences. Mesenchymal stem cell (MSC)-based therapy is promising, but the problems of poor engraftment and insufficient neurotrophic effects need to be overcome. Herein, we isolated platelet-rich plasma-derived exosomes (PRP-Exos), which contain abundant bioactive molecules, and investigated their potential to increase the regenerative capacity of MSCs. We observed that PRP-Exos significantly increased MSC proliferation, viability, and mobility, decreased MSC apoptosis under stress, maintained MSC stemness, and attenuated MSC senescence. In vivo, PRP-Exo-treated MSCs (pExo-MSCs) exhibited an increased retention rate and heightened therapeutic efficacy, as indicated by increased axonal regeneration, remyelination, and recovery of neurological function in a PNI model. In vitro, pExo-MSCs coculture promoted Schwann cell proliferation and dorsal root ganglion axon growth. Moreover, the increased neurotrophic behaviour of pExo-MSCs was mediated by trophic factors, particularly glia-derived neurotrophic factor (GDNF), and PRP-Exos activated the PI3K/Akt signalling pathway in MSCs, leading to the observed phenotypes. These findings demonstrate that PRP-Exos may be novel agents for increasing the ability of MSCs to promote neural repair and regeneration in patients with PNI.
Collapse
Affiliation(s)
- Yongyi Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China; State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; No.962 Hospital of the PLA Joint Logistic Support Force, Harbin 150080, China
| | - Dan Yi
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Quan Hong
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiangbei Cao
- Departments of Anaesthesiology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaodong Geng
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinwei Liu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuang Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengyu Cao
- Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Chen
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuaixuan Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Molin Li
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yaqiong Zhu
- Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| | - Nan Peng
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
42
|
Mattera V, Occhiuzzi F, Correale J, Pasquini JM. Remyelinating effect driven by transferrin-loaded extracellular vesicles. Glia 2024; 72:338-361. [PMID: 37860913 DOI: 10.1002/glia.24478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023]
Abstract
Extracellular vesicles (EVs) are involved in diverse cellular functions, playing a significant role in cell-to-cell communication in both physiological conditions and pathological scenarios. Therefore, EVs represent a promising therapeutic strategy. Oligodendrocytes (OLs) are myelinating glial cells developed from oligodendrocyte progenitor cells (OPCs) and damaged in chronic demyelinating diseases such as multiple sclerosis (MS). Glycoprotein transferrin (Tf) plays a critical role in iron homeostasis and has pro-differentiating effects on OLs in vivo and in vitro. In the current work, we evaluated the use of EVs as transporters of Tf to the central nervous system (CNS) through the intranasal (IN) route. For the in vitro mechanistic studies, we used rat plasma EVs. Our results show that EVTf enter OPCs through clathrin-caveolae and cholesterol-rich lipid raft endocytic pathways, releasing the cargo and exerting a pro-maturation effect on OPCs. These effects were also observed in vivo using the animal model of demyelination induced by cuprizone (CPZ). In this model, IN administered Tf-loaded EVs isolated from mouse plasma reached the brain parenchyma, internalizing into OPCs, promoting their differentiation, and accelerating remyelination. Furthermore, in vivo experiments demonstrated that EVs protected the Tf cargo and significantly reduced the amount of Tf required to induce remyelination as compared to soluble Tf. Collectively, these findings unveil EVs as functional nanocarriers of Tf to induce remyelination.
Collapse
Affiliation(s)
- Vanesa Mattera
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Federico Occhiuzzi
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Jorge Correale
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| | - Juana M Pasquini
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
43
|
Blasko F, Horvathova L. The relationship between the tumor and its innervation: historical, methodical, morphological, and functional assessments - A minireview. Endocr Regul 2024; 58:68-82. [PMID: 38563296 DOI: 10.2478/enr-2024-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
The acceptance of the tumor as a non-isolated structure within the organism has opened a space for the study of a wide spectrum of potential direct and indirect interactions, not only between the tumor tissue and its vicinity, but also between the tumor and its macroenvironment, including the nervous system. Although several lines of evidence have implicated the nervous system in tumor growth and progression, for many years, researchers believed that tumors lacked innervation and the notion of indirect neuro-neoplastic interactions via other systems (e.g., immune, or endocrine) predominated. The original idea that tumors are supplied not only by blood and lymphatic vessels, but also autonomic and sensory nerves that may influence cancer progression, is not a recent phenomenon. Although in the past, mainly due to the insufficiently sensitive methodological approaches, opinions regarding the presence of nerves in tumors were inconsistent. However, data from the last decade have shown that tumors are able to stimulate the formation of their own innervation by processes called neo-neurogenesis and neo-axonogenesis. It has also been shown that tumor infiltrating nerves are not a passive, but active components of the tumor microenvironment and their presence in the tumor tissue is associated with an aggressive tumor phenotype and correlates with poor prognosis. The aim of the present review was to 1) summarize the available knowledge regarding the course of tumor innervation, 2) present the potential mechanisms and pathways for the possible induction of new nerve fibers into the tumor microenvironment, and 3) highlight the functional significance/consequences of the nerves infiltrating the tumors.
Collapse
Affiliation(s)
- Filip Blasko
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Lubica Horvathova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
44
|
Xia B, Gao X, Qian J, Li S, Yu B, Hao Y, Wei B, Ma T, Wu H, Yang S, Zheng Y, Gao X, Guo L, Gao J, Yang Y, Zhang Y, Wei Y, Xue B, Jin Y, Luo Z, Zhang J, Huang J. A Novel Superparamagnetic Multifunctional Nerve Scaffold: A Remote Actuation Strategy to Boost In Situ Extracellular Vesicles Production for Enhanced Peripheral Nerve Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305374. [PMID: 37652460 DOI: 10.1002/adma.202305374] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/03/2023] [Indexed: 09/02/2023]
Abstract
Extracellular vesicles (EVs) have inherent advantages over cell-based therapies in regenerative medicine because of their cargos of abundant bioactive cues. Several strategies are proposed to tune EVs production in vitro. However, it remains a challenge for manipulation of EVs production in vivo, which poses significant difficulties for EVs-based therapies that aim to promote tissue regeneration, particularly for long-term treatment of diseases like peripheral neuropathy. Herein, a superparamagnetic nanocomposite scaffold capable of controlling EVs production on-demand is constructed by incorporating polyethyleneglycol/polyethyleneimine modified superparamagnetic nanoparticles into a polyacrylamide/hyaluronic acid double-network hydrogel (Mag-gel). The Mag-gel is highly sensitive to a rotating magnetic field (RMF), and can act as mechano-stimulative platform to exert micro/nanoscale forces on encapsulated Schwann cells (SCs), an essential glial cell in supporting nerve regeneration. By switching the ON/OFF state of the RMF, the Mag-gel can scale up local production of SCs-derived EVs (SCs-EVs) both in vitro and in vivo. Further transcriptome sequencing indicates an enrichment of transcripts favorable in axon growth, angiogenesis, and inflammatory regulation of SCs-EVs in the Mag-gel with RMF, which ultimately results in optimized nerve repair in vivo. Overall, this research provides a noninvasive and remotely time-scheduled method for fine-tuning EVs-based therapies to accelerate tissue regeneration, including that of peripheral nerves.
Collapse
Affiliation(s)
- Bing Xia
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
- Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xue Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jiaqi Qian
- College of Chemical Engineering, Fuzhou University, Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Shengyou Li
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Beibei Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Bin Wei
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Haining Wu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Shijie Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xueli Gao
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Lingli Guo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jianbo Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yujie Yang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yongfeng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710032, P. R. China
| | - Yitao Wei
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Borui Xue
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yan Jin
- Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| |
Collapse
|
45
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
46
|
Ghosh M, Pearse DD. Schwann Cell-Derived Exosomal Vesicles: A Promising Therapy for the Injured Spinal Cord. Int J Mol Sci 2023; 24:17317. [PMID: 38139147 PMCID: PMC10743801 DOI: 10.3390/ijms242417317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles released by cells into their extracellular milieu. Within these nanovesicles reside a multitude of bioactive molecules, which orchestrate essential biological processes, including cell differentiation, proliferation, and survival, in the recipient cells. These bioactive properties of exosomes render them a promising choice for therapeutic use in the realm of tissue regeneration and repair. Exosomes possess notable positive attributes, including a high bioavailability, inherent safety, and stability, as well as the capacity to be functionalized so that drugs or biological agents can be encapsulated within them or to have their surface modified with ligands and receptors to imbue them with selective cell or tissue targeting. Remarkably, their small size and capacity for receptor-mediated transcytosis enable exosomes to cross the blood-brain barrier (BBB) and access the central nervous system (CNS). Unlike cell-based therapies, exosomes present fewer ethical constraints in their collection and direct use as a therapeutic approach in the human body. These advantageous qualities underscore the vast potential of exosomes as a treatment option for neurological injuries and diseases, setting them apart from other cell-based biological agents. Considering the therapeutic potential of exosomes, the current review seeks to specifically examine an area of investigation that encompasses the development of Schwann cell (SC)-derived exosomal vesicles (SCEVs) as an approach to spinal cord injury (SCI) protection and repair. SCs, the myelinating glia of the peripheral nervous system, have a long history of demonstrated benefit in repair of the injured spinal cord and peripheral nerves when transplanted, including their recent advancement to clinical investigations for feasibility and safety in humans. This review delves into the potential of utilizing SCEVs as a therapy for SCI, explores promising engineering strategies to customize SCEVs for specific actions, and examines how SCEVs may offer unique clinical advantages over SC transplantation for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
47
|
Fuentes-Flores A, Geronimo-Olvera C, Girardi K, Necuñir-Ibarra D, Patel SK, Bons J, Wright MC, Geschwind D, Hoke A, Gomez-Sanchez JA, Schilling B, Rebolledo DL, Campisi J, Court FA. Senescent Schwann cells induced by aging and chronic denervation impair axonal regeneration following peripheral nerve injury. EMBO Mol Med 2023; 15:e17907. [PMID: 37860842 DOI: 10.15252/emmm.202317907] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Following peripheral nerve injury, successful axonal growth and functional recovery require Schwann cell (SC) reprogramming into a reparative phenotype, a process dependent upon c-Jun transcription factor activation. Unfortunately, axonal regeneration is greatly impaired in aged organisms and following chronic denervation, which can lead to poor clinical outcomes. While diminished c-Jun expression in SCs has been associated with regenerative failure, it is unclear whether the inability to maintain a repair state is associated with the transition into an axonal growth inhibition phenotype. We here find that reparative SCs transition into a senescent phenotype, characterized by diminished c-Jun expression and secretion of inhibitory factors for axonal regeneration in aging and chronic denervation. In both conditions, the elimination of senescent SCs by systemic senolytic drug treatment or genetic targeting improved nerve regeneration and functional recovery, increased c-Jun expression and decreased nerve inflammation. This work provides the first characterization of senescent SCs and their influence on axonal regeneration in aging and chronic denervation, opening new avenues for enhancing regeneration and functional recovery after peripheral nerve injuries.
Collapse
Affiliation(s)
- Andrés Fuentes-Flores
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Cristian Geronimo-Olvera
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Karina Girardi
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - David Necuñir-Ibarra
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Megan C Wright
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ahmet Hoke
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Instituto de Neurociencias de Alicante, UMH-CSIC, San Juan de Alicante, Spain
| | | | - Daniela L Rebolledo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
48
|
Wang Y, Shi G, Huang TCT, Li J, Long Z, Reisdorf R, Shin AY, Amadio P, Behfar A, Zhao C, Moran SL. Enhancing Functional Recovery after Segmental Nerve Defect Using Nerve Allograft Treated with Plasma-Derived Exosome. Plast Reconstr Surg 2023; 152:1247-1258. [PMID: 36912739 DOI: 10.1097/prs.0000000000010389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Nerve injuries can result in detrimental functional outcomes. Currently, autologous nerve graft offers the best outcome for segmental peripheral nerve injury. Allografts are alternatives, but do not have comparable results. This study evaluated whether plasma-derived exosome can improve nerve regeneration and functional recovery when combined with decellularized nerve allografts. METHODS The effect of exosomes on Schwann cell proliferation and migration were evaluated. A rat model of sciatic nerve repair was used to evaluate the effect on nerve regeneration and functional recovery. A fibrin sealant was used as the scaffold for exosome. Eighty-four Lewis rats were divided into autograft, allograft, and allograft with exosome groups. Gene expression of nerve regeneration factors was analyzed on postoperative day 7. At 12 and 16 weeks, rats were subjected to maximum isometric tetanic force and compound muscle action potential. Nerve specimens were then analyzed by means of histology and immunohistochemistry. RESULTS Exosomes were readily taken up by Schwann cells that resulted in improved Schwann cell viability and migration. The treated allograft group had functional recovery (compound muscle action potential, isometric tetanic force) comparable to that of the autograft group. Similar results were observed in gene expression analysis of nerve regenerating factors. Histologic analysis showed no statistically significant differences between treated allograft and autograft groups in terms of axonal density, fascicular area, and myelin sheath thickness. CONCLUSIONS Plasma-derived exosome treatment of decellularized nerve allograft may provide comparable clinical outcomes to that of an autograft. This can be a promising strategy in the future as an alternative for segmental peripheral nerve repair. CLINICAL RELEVANCE STATEMENT Off-the-shelf exosomes may improve recovery in nerve allografts.
Collapse
Affiliation(s)
- Yicun Wang
- From the Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University
- Division of Plastic Surgery, Department of Surgery
- Department of Orthopedic Surgery
| | - Guidong Shi
- Department of Orthopedic Surgery
- Tianjin Medical University
| | | | - Jialun Li
- Division of Plastic Surgery, Department of Surgery
- Department of Plastic Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology
| | | | | | | | | | - Atta Behfar
- Center for Regenerative Medicine
- Department of Cardiovascular Medicine, Mayo Clinic
| | | | | |
Collapse
|
49
|
You M, Xing H, Yan M, Zhang J, Chen J, Chen Y, Liu X, Zhu J. Schwann Cell-Derived Exosomes Ameliorate Paclitaxel-Induced Peripheral Neuropathy Through the miR-21-Mediated PTEN Signaling Pathway. Mol Neurobiol 2023; 60:6840-6851. [PMID: 37498480 DOI: 10.1007/s12035-023-03488-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023]
Abstract
Paclitaxel-induced peripheral neuropathy (PIPN) is a neurological disorder resulting from paclitaxel (PTX) treatment for cancer patients. There are currently no drugs available that can definitively prevent or treat PIPN. Exosomes are cell-secreted nanoscale vesicles that mediate communication between neurons and glial cells. We found that Schwann cell-derived exosomes (SC-EXOs) robustly improved PIPN both in vitro and in vivo. In vivo studies showed that SC-EXOs were able to alleviate PTX-induced mechanical nociceptive sensitization in rats. Pathomorphological analysis showed that SC-EXOs ameliorated PTX-induced plantar intraepidermal nerve fiber loss and dorsal root ganglion (DRG) injury. Additionally, the results of in vitro studies showed that SC-EXOs had significant protective effects on the DRG cells damaged by PTX, and did not affect the antitumor effect of PTX against Hela cells. Further, mechanism research revealed that SC-EXOs promoted axonal regeneration and protected damaged neurons by upregulating miR-21 to repress the phosphatase and tensin homolog (PTEN) pathway, which could improve PIPN. Taken together, these findings suggest that SC-EXOs ameliorate PTX-induced peripheral neuropathy via the miR-21-mediated PTEN signaling pathway, which provides a novel strategy for the treatment of PIPN.
Collapse
Affiliation(s)
- Min You
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haizhu Xing
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ming Yan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Jie Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Jiayi Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yang Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Xiaoli Liu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Jing Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Chen YS, Harn HJ, Hong ZX, Huang YC, Lin YT, Zheng HX, Chen PY, Yang HH, Chen PR, Tsai HC, Lin SZ, Ho TJ, Chiou TW. Preconditioning of exosomes derived from human olfactory ensheathing cells improved motor coordination and balance in an SCA3/MJD mouse model: A new therapeutic approach. Eur J Pharm Sci 2023; 191:106608. [PMID: 37832855 DOI: 10.1016/j.ejps.2023.106608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Exosome therapy is a novel trend in regeneration medicine. However, identifying a suitable biomarker that can associate the therapeutic efficacy of exosomes with SCA3/MJD is essential. In this study, parental cells were preconditioned with butylidenephthalide (Bdph) for exosome preparation to evaluate the therapeutic effect of SCA3/MJD. The therapeutic agent hsa-miRNA-6780-5p was enriched up to 98-fold in exosomes derived from butylidenephthalide (Bdph)-preconditioned human olfactory ensheathing cells (hOECs) compared with that in naïve hOECs exosomes. The particle sizes of exosomes derived from naïve hOECs and those derived from hOECs preconditioned with Bdph were approximately 113.0 ± 3.5 nm and 128.9 ± 0.7 nm, respectively. A liposome system was used to demonstrate the role of hsa-miRNA-6780-5p, wherein hsa-miRNA-6780-5p was found to enhance autophagy and inhibit the expression of spinocerebellar ataxia type 3 (SCA3) disease proteins with the polyglutamine (polyQ) tract. Exosomes with enriched hsa-miRNA-6780-5p were further applied to HEK-293-84Q cells, leading to decreased expression of polyQ and increased autophagy. The results were reversed when 3MA, an autophagy inhibitor, was added to the cells treated with hsa-miRNA-6780-5p-enriched exosomes, indicating that the decreased polyQ expression was modulated via autophagy. SCA3 mice showed improved motor coordination behavior when they intracranially received exosomes enriched with hsa-miRNA-6780-5p. SCA3 mouse cerebellar tissues treated with hsa-miRNA-6780-5p-enriched exosomes showed decreased expression of polyQ and increased expression of LC3II/I, an autophagy marker. In conclusion, our findings can serve as a basis for developing an alternative therapeutic strategy for SCA3 disease treatment using miRNA-enriched exosomes derived from chemically preconditioned cells.
Collapse
Affiliation(s)
- Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan, ROC; Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC; Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Horng-Jyh Harn
- Department of Pathology, Hualien Tzu Chi Hospital, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Zhen-Xiang Hong
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan, ROC
| | - Yi-Chen Huang
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd, Shoufeng, Hualien 974301, Taiwan, ROC
| | - Yi-Tung Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan, ROC
| | - Hui-Xuan Zheng
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd, Shoufeng, Hualien 974301, Taiwan, ROC
| | - Pei-Yu Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan, ROC
| | - Hsueh-Hui Yang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Peir-Rong Chen
- Department of Otolaryngology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, Taiwan, ROC
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Sec. 3, Chung-Yang Rd., Hualien, Taiwan, ROC.
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd, Shoufeng, Hualien 974301, Taiwan, ROC.
| |
Collapse
|