1
|
Li D, Xie X, Ou Y, Sun P, Lin J, Yu C, Huang H, Huang L, Yang C, Kuang W, Zhou C. Bone marrow mesenchymal stem cells-derived exosomal miR-24-3p alleviates spinal cord injury by targeting MAPK9 to inhibit the JNK/c-Jun/c-Fos pathway. Arch Biochem Biophys 2025; 769:110434. [PMID: 40274174 DOI: 10.1016/j.abb.2025.110434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Spinal cord injury (SCI) is a very harmful neurological disease that can cause serious damage to sensation, movement, and autonomic nervous function below the affected area. Apoptosis and inflammatory response play important roles in the pathological process of spinal cord injury. The exosomes secreted by bone marrow mesenchymal stem cells (BMSCs) may play a protective role against spinal cord injury. However, the detailed mechanism behind this is not fully understood. The main objective of this study was to investigate the anti-inflammatory and anti-apoptotic effects of bone marrow mesenchymal stem cell exosomes (BMSCs-EXO) in SCI in vitro and in vivo and their mechanisms. The study demonstrated that bone marrow mesenchymal stem cells reduced apoptosis and inflammation and promoted axon growth in LPS-treated PC12 cells. The miRDB predicted that miR-24-3p targets MAPK9(JNK2). Transcriptome sequencing and Western blot confirmed that miR-24-3p inhibits the JNK/c-Jun/c-Fos pathway by targeting MAPK9. In vivo experiments, injection of BMSC exosomes overexpressing miR-24-3p from the tail vein attenuated the SCI exercise injury-related behavior in rats. In conclusion, this study indicates that bone marrow MSC-derived exosomes can mitigate SCI-related injury by suppressing apoptosis and inflammation, with miR-24-3p playing a crucial role, potentially offering a novel therapeutic approach for SCI treatment.
Collapse
Affiliation(s)
- Dailong Li
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Xuyuan Xie
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Orthopedics, The First People's Hospital of Zhaoqing, Zhaoqing, 526000, China
| | - Yuxuan Ou
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Peiwen Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jiezhao Lin
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Orthopedic Surgery, Shantou Central Hospital, Shantou, 515000, China
| | - Cheng Yu
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Haoran Huang
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Longcheng Huang
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Changjian Yang
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wenhao Kuang
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Chusong Zhou
- Department of Spinal Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
2
|
Bellák T, Fekécs Z, Török D, Kristóf R, Esezoobo O, Marton A, Vizler C, Nógrádi A, Pajer K. Systemic stem cell treatment rescues injured motoneurons by reducing L-selectin expression on leukocytes. Stem Cell Res Ther 2025; 16:237. [PMID: 40361249 PMCID: PMC12076817 DOI: 10.1186/s13287-025-04283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/19/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Avulsion injury results in motoneuron death due to the increased cytotoxicity developing after the injury. We have earlier shown that intraspinally grafted immortalized NE-4C neuroectodermal stem cells derived from 9-day old mouse forebrain vesicles produced a secretome, which induced decreased microglia/macrophage reaction, and promoted the neuroprotection and regeneration following avulsion injury. Here we intended to prove the motoneuron rescuing effect of intravenously grafted NE-4C stem cells and reveal the mechanism of action used by the grafted cells. METHODS In our experimental model the left lumbar 4 (L4) ventral root of the spinal cord was avulsed and then reimplanted into the L4 spinal segment. Treated animals received various doses of NE-4C stem cells intravenously and the survival and regeneration of the affected motoneurons was checked by morphological and functional analysis. The molecular changes within the treated cord were followed by the ELISA Proteome Profiler rat cytokine array and qPCR analysis. To mimic the effect of stem cells fucoidan treatment (a specific selectin inhibitor, 50 and 100 mg/kg bw) was applied for two weeks intraperitoneally. RESULTS High doses of intravenous stem cell treatment (4 × 105 and 1 × 106 cells) induced the reinnervation of the reimplanted ventral root by surviving injured motoneurons (up to 38% of the total L4 pool). Proteome Profiler analysis showed that systemic stem cell treatment downregulated the level of L-selectin, that promotes leukocyte rolling on vascular endothelium. Both systemic stem cell and fucoidan treatment reduced macrophage and microglial densities in the affected spinal segment and administration of fucoidan downregulated inflammatory cytokine and inflammasome levels along with improved morphological and functional reinnervation. CONCLUSIONS Blocking L-selectin, similarly to systemic NE-4C stem cell treatment decreases the neuroinflammation in the injured spinal cord segment after ventral root avulsion and induces significant motoneuron survival and functional reinnervation of the denervated hind limb muscles.
Collapse
Affiliation(s)
- Tamás Bellák
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Zoltán Fekécs
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Dénes Török
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Rebeka Kristóf
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Omoikhoje Esezoobo
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Present address: Galway University Hospitals, Galway, Ireland
| | - Annamária Marton
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Center, Szeged, Hungary
| | - Csaba Vizler
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Center, Szeged, Hungary
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
3
|
Balzani MFV, Coser LDO, de Oliveira ALR. Immunomodulation by 4-Hydroxy-TEMPO (TEMPOL) and Dimethyl Fumarate (DMF) After Ventral Root Crush (VRC) in C57BL/6J Mice: A Flow Cytometry Analysis. BIOLOGY 2025; 14:473. [PMID: 40427663 PMCID: PMC12109085 DOI: 10.3390/biology14050473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 04/11/2025] [Indexed: 05/29/2025]
Abstract
Spinal motor nerve root lesions can happen after avulsion or crush, generating acute motoneuron death and synaptic loss, consequently, causing motor and sensory dysfunctions. Local response is mediated by astroglial and microglial cells, giving rise to a pro-inflammatory profile. TEMPOL and DMF are drugs that have been studied in our laboratory after spinal cord nerve root injuries and have demonstrated significant results in terms of neuroprotection and immunomodulation, decreasing the inflammation process. In the present work, a flow cytometry approach was used to evaluate cellular responses to injury and immunomodulation. For this, injured animals received TEMPOL, DMF or vehicle once a day for 7, 14 or 28 days of treatment. Flow cytometry multiparametric analysis allowed the quantification of different pro- and anti-inflammatory glial, macrophage and lymphocyte markers. Contrasting with the vehicle treated counterpart, TEMPOL and DMF led to downregulation of pro-inflammatory cytokines in astrocytes and microglia subpopulations, but did not show significant results in increasing anti-inflammatory phenotypes. As for macrophage and lymphocyte subpopulations, both treatments showed a balance between pro- and anti-inflammatory phenotypes. Therefore, it was concluded that both drugs exhibit immunomodulatory action, contributing to a pro-regenerative profile in the tissue.
Collapse
Affiliation(s)
- Maria Fernanda Vannucci Balzani
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
| | - Lilian de Oliveira Coser
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
| | - Alexandre Leite Rodrigues de Oliveira
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
- Center for Gender-Specific Biology and Medicine (CGBM)–Brazil, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil
| |
Collapse
|
4
|
Hua R, Zhao C, Xu Z, Liu D, Shen W, Yuan W, Li Y, Ma J, Wang Z, Feng S. ROS-responsive nanoparticle delivery of ferroptosis inhibitor prodrug to facilitate mesenchymal stem cell-mediated spinal cord injury repair. Bioact Mater 2024; 38:438-454. [PMID: 38770428 PMCID: PMC11103787 DOI: 10.1016/j.bioactmat.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Spinal cord injury (SCI) is a traumatic condition that results in impaired motor and sensory function. Ferroptosis is one of the main causes of neural cell death and loss of neurological function in the spinal cord, and ferroptosis inhibitors are effective in reducing inflammation and repairing SCI. Although human umbilical cord mesenchymal stem cells (Huc-MSCs) can ameliorate inflammatory microenvironments and promote neural regeneration in SCI, their efficacy is greatly limited by the local microenvironment after SCI. Therefore, in this study, we constructed a drug-release nanoparticle system with synergistic Huc-MSCs and ferroptosis inhibitor, in which we anchored Huc-MSCs by a Tz-A6 peptide based on the CD44-targeting sequence, and combined with the reactive oxygen species (ROS)-responsive drug nanocarrier mPEG-b-Lys-BECI-TCO at the other end for SCI repair. Meanwhile, we also modified the classic ferroptosis inhibitor Ferrostatin-1 (Fer-1) and synthesized a new prodrug Feborastatin-1 (Feb-1). The results showed that this treatment regimen significantly inhibited the ferroptosis and inflammatory response after SCI, and promoted the recovery of neurological function in rats with SCI. This study developed a combination therapy for the treatment of SCI and also provides a new strategy for the construction of a drug-coordinated cell therapy system.
Collapse
Affiliation(s)
- Renshuai Hua
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhengyu Xu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Derong Liu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Wenyuan Shen
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Orthopedics, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, China
| | - Wenlu Yuan
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yan Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jun Ma
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhishuo Wang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shiqing Feng
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Orthopedics, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, China
| |
Collapse
|
5
|
Fujiwara S, Nakano-Doi A, Sawano T, Kubo S, Doe N, Nakagomi T. Administration of Human-Derived Mesenchymal Stem Cells Activates Locally Stimulated Endogenous Neural Progenitors and Reduces Neurological Dysfunction in Mice after Ischemic Stroke. Cells 2024; 13:939. [PMID: 38891071 PMCID: PMC11171641 DOI: 10.3390/cells13110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Increasing evidence shows that the administration of mesenchymal stem cells (MSCs) is a promising option for various brain diseases, including ischemic stroke. Studies have demonstrated that MSC transplantation after ischemic stroke provides beneficial effects, such as neural regeneration, partially by activating endogenous neural stem/progenitor cells (NSPCs) in conventional neurogenic zones, such as the subventricular and subgranular zones. However, whether MSC transplantation regulates the fate of injury-induced NSPCs (iNSPCs) regionally activated at injured regions after ischemic stroke remains unclear. Therefore, mice were subjected to ischemic stroke, and mCherry-labeled human MSCs (h-MSCs) were transplanted around the injured sites of nestin-GFP transgenic mice. Immunohistochemistry of brain sections revealed that many GFP+ cells were observed around the grafted sites rather than in the regions in the subventricular zone, suggesting that transplanted mCherry+ h-MSCs stimulated GFP+ locally activated endogenous iNSPCs. In support of these findings, coculture studies have shown that h-MSCs promoted the proliferation and neural differentiation of iNSPCs extracted from ischemic areas. Furthermore, pathway analysis and gene ontology analysis using microarray data showed that the expression patterns of various genes related to self-renewal, neural differentiation, and synapse formation were changed in iNSPCs cocultured with h-MSCs. We also transplanted h-MSCs (5.0 × 104 cells/µL) transcranially into post-stroke mouse brains 6 weeks after middle cerebral artery occlusion. Compared with phosphate-buffered saline-injected controls, h-MSC transplantation displayed significantly improved neurological functions. These results suggest that h-MSC transplantation improves neurological function after ischemic stroke in part by regulating the fate of iNSPCs.
Collapse
Affiliation(s)
- Shuichi Fujiwara
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| | - Toshinori Sawano
- Department of Biomedical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-8577, Japan;
| | - Shuji Kubo
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
| | - Nobutaka Doe
- Department of Rehabilitation, Hyogo Medical University (Kobe Campus), 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan;
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| |
Collapse
|
6
|
Sun Y, Liu Q, Qin Y, Xu Y, Zhao J, Xie Y, Li C, Qin T, Jin Y, Jiang L, Cao Y, Lu H, Hu J. Exosomes derived from CD271 +CD56 + bone marrow mesenchymal stem cell subpopoulation identified by single-cell RNA sequencing promote axon regeneration after spinal cord injury. Theranostics 2024; 14:510-527. [PMID: 38169566 PMCID: PMC10758065 DOI: 10.7150/thno.89008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale: Spinal cord injury (SCI) results in neural tissue damage. However, the limited regenerative capacity of adult mammals' axons upon SCI leads to persistent neurological dysfunction. Thus, exploring the pathways that can enhance axon regeneration in injured spinal cord is of great significance. Methods: Through the utilization of single-cell RNA sequencing in this research, a distinct subpopulation of bone marrow mesenchymal stem cells (BMSCs) that exhibits the capacity to facilitate axon regeneration has been discovered. Subsequently, the CD271+CD56+ BMSCs subpopulation was isolated using flow cytometry, and the exosomes derived from this subpopulation (CD271+CD56+ BMSC-Exos) were extracted and incorporated into a hydrogel to create a sustained release system. The aim was to investigate the therapeutic effects of CD271+CD56+ BMSC-Exos and elucidate the underlying mechanisms involved in promoting axon regeneration and neural function recovery. Results: The findings indicate that CD271+CD56+ BMSC-Exos share similar physical and chemical properties with conventional exosomes. Importantly, in an SCI model, in situ implantation of CD271+CD56+ BMSC-Exos hydrogel resulted in increased expression of NF and synaptophysin, markers associated with axon regeneration and synapse formation, respectively. This intervention also contributed to improved neural function recovery. In vitro experiments demonstrated that CD271+CD56+ BMSC-Exos treatment significantly enhanced axon extension distance and increased the number of branches in dorsal root ganglion axons. Moreover, further investigation into the molecular mechanisms underlying CD271+CD56+ BMSC-Exos-mediated axon regeneration revealed the crucial involvement of the miR-431-3p/RGMA axis. Conclusion: In summary, the implantation of CD271+CD56+ BMSC-Exos hydrogel presents a promising and effective therapeutic approach for SCI.
Collapse
Affiliation(s)
- Yi Sun
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Quanbo Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yiming Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yan Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Jinyun Zhao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yong Xie
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Chengjun Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Tian Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yuxin Jin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Liyuan Jiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yong Cao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| |
Collapse
|
7
|
Paes SM, Castro MVD, Barbosa RM, Politti Cartarozzi L, Coser LDO, Kempe PRG, Decarli MC, Moraes ÂM, Barraviera B, Ferreira Júnior RS, Oliveira ALRD. Human dental pulp stem cell monolayer and spheroid therapy after spinal motor root avulsion in adult rats. Brain Res 2023; 1802:148229. [PMID: 36592804 DOI: 10.1016/j.brainres.2022.148229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
Spinal cord injuries result in severe neurological deficits and neuronal loss, with poor functional recovery. Mesenchymal stem cells have shown promising results; therefore the present objective of this work was to compare motor recovery after treatment with human dental pulp stem cells (hDPSC) cultivated in monolayer (2D) or as spheroids (3D), following avulsion and reimplantation of spinal motor roots in adult rats. Thus, 72 adult female Lewis rats were divided into 4 groups: avulsion (AV); avulsion followed by reimplantation (AR); avulsion associated with reimplant and 2D cell therapy (AR + 2D), and avulsion associated with reimplant and 3D cell therapy (AR + 3D). The application of the cells in 2D and 3D was performed by microsurgery, with subsequent functional assessment using a walking track test (Catwalk system), immunohistochemistry, neuronal survival, and qRT-PCR in 1-, 4-, and 12-weeks post-injury. The animals in the AR + 2D and AR + 3D groups showed the highest neuronal survival rates, and immunofluorescence revealed downregulation of GFAP, and Iba-1, with preservation of synaptophysin, indicating a reduction in glial reactivity, combined with the maintenance of pre-synaptic inputs. There was an increase in anti-inflammatory (IL-4, TGFβ) and a reduction of pro-inflammatory factors (IL-6, TNFα) in animals treated with reimplantation and hDPSC. As for the functional recovery, in all analyzed parameters, the AR + 2D group performed better and was superior to the avulsion alone. Overall, our results indicate that the 2D and 3D cell therapy approaches provide successful immunomodulation and motor recovery, consistent with advanced therapies after spinal cord injury.
Collapse
Affiliation(s)
- Sabrina Moreira Paes
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mateus Vidigal de Castro
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Rafael Maza Barbosa
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil; School of Chemical Engineering, University of Campinas, UNICAMP, Av. Albert Einstein, 500, Brazil
| | - Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Lilian de Oliveira Coser
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paula Regina Gelinski Kempe
- Laboratory of Nerve Regeneration, Institute of Biology (IB), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Monize Caiado Decarli
- School of Chemical Engineering, University of Campinas, UNICAMP, Av. Albert Einstein, 500, Brazil
| | - Ângela Maria Moraes
- School of Chemical Engineering, University of Campinas, UNICAMP, Av. Albert Einstein, 500, Brazil
| | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Rui Seabra Ferreira Júnior
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | |
Collapse
|
8
|
Poly (L-Lactic Acid) Cell-Laden Scaffolds Applied on Swine Model of Tracheal Fistula. J Surg Res 2022; 277:319-334. [PMID: 35552075 DOI: 10.1016/j.jss.2022.03.024] [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: 06/24/2021] [Revised: 02/20/2022] [Accepted: 03/21/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Tracheal fistula (TF) treatments may involve temporary orthosis and further ablative procedures, which can lead to infection. Thus, TF requires other therapy alternatives development. The hypothesis of this work was to demonstrate the feasibility of a tissue-engineered alternative for small TF in a preclinical model. Also, its association with suture filaments enriched with adipose tissue-derived mesenchymal stromal stem cells (AT-MSCs) was assessed to determine whether it could optimize the regenerative process. METHODS Poly (L-Lactic acid) (PLLA) membranes were manufactured by electrospinning and had morphology analyzed by scanning electron microscopy. AT-MSCs were cultured in these scaffolds and in vitro assays were performed (cytotoxicity, cellular adhesion, and viability). Subsequently, these cellular constructs were implanted in an animal small TF model. The association with suture filaments containing attached AT-MSCs was present in one animal group. After 30 d, animals were sacrificed and regenerative potential was evaluated, mainly related to the extracellular matrix remodeling, by performing histopathological (Hematoxylin-Eosin and trichrome Masson) and immunohistochemistry (Collagen I/II/III, matrix metalloproteinases-2, matrix metalloproteinases-9, vascular endothelial growth factor, and interleukin-10) analyses. RESULTS PLLA membranes presented porous fibers, randomly oriented. In vitro assays results showed that AT-MSCs attached were viable and maintained an active metabolism. Swine implanted with AT-MSCs attached to membranes and suture filaments showed aligned collagen fibers and a better regenerative progress in 30 d. CONCLUSIONS PLLA membranes with AT-MSCs attached were useful to the extracellular matrix restoration and have a high potential for small TF treatment. Also, their association with suture filaments enriched with AT-MSCs was advantageous.
Collapse
|
9
|
Nito C, Suda S, Nitahara-Kasahara Y, Okada T, Kimura K. Dental-Pulp Stem Cells as a Therapeutic Strategy for Ischemic Stroke. Biomedicines 2022; 10:biomedicines10040737. [PMID: 35453487 PMCID: PMC9032844 DOI: 10.3390/biomedicines10040737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Regenerative medicine aims to restore human functions by regenerating organs and tissues using stem cells or living tissues for the treatment of organ and tissue defects or dysfunction. Clinical trials investigating the treatment of cerebral infarction using mesenchymal stem cells, a type of somatic stem cell therapy, are underway. The development and production of regenerative medicines using somatic stem cells is expected to contribute to the treatment of cerebral infarction, a central nervous system disease for which there is no effective treatment. Numerous experimental studies have shown that cellular therapy, including the use of human dental pulp stem cells, is an attractive strategy for patients with ischemic brain injury. This review describes the basic research, therapeutic mechanism, clinical trials, and future prospects for dental pulp stem cell therapy, which is being investigated in Japan in first-in-human clinical trials for the treatment of patients with acute cerebral ischemia.
Collapse
Affiliation(s)
- Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (S.S.); (K.K.)
- Collaborative Research Center, Laboratory for Clinical Research, Nippon Medical School, Tokyo 113-8603, Japan
- Correspondence: ; Tel.: +81-3-3822-2131; Fax: +81-3-5814-6176
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (S.S.); (K.K.)
| | - Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; (Y.N.-K.); (T.O.)
| | - Takashi Okada
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; (Y.N.-K.); (T.O.)
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (S.S.); (K.K.)
| |
Collapse
|
10
|
Vellosillo L, Pascual-Guerra J, Muñoz MP, Rodríguez-Navarro JA, González-Nieto D, Barrio LC, Lobo MDVT, Paíno CL. Oligodendroglia Generated From Adult Rat Adipose Tissue by Direct Cell Conversion. Front Cell Dev Biol 2022; 10:741499. [PMID: 35223826 PMCID: PMC8873586 DOI: 10.3389/fcell.2022.741499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/19/2022] [Indexed: 11/28/2022] Open
Abstract
Obtaining oligodendroglial cells from dispensable tissues would be of great interest for autologous or immunocompatible cell replacement therapy in demyelinating diseases, as well as for studying myelin-related pathologies or testing therapeutic approaches in culture. We evaluated the feasibility of generating oligodendrocyte precursor cells (OPCs) from adult rat adipose tissue by expressing genes encoding transcription factors involved in oligodendroglial development. Adipose-derived mesenchymal cells were lentivirally transduced with tetracycline-inducible Sox10, Olig2, Zfp536, and/or Nkx6.1 transgenes. Immunostaining with the OPC-specific O4 monoclonal antibody was used to mark oligodendroglial induction. O4- and myelin-associated glycoprotein (MAG)-positive cells emerged after 3 weeks when using the Sox10 + Olig2 + Zfp536 combination, followed in the ensuing weeks by GFAP-, O1 antigen-, p75NTR (low-affinity NGF receptor)-, and myelin proteins-positive cells. The O4+ cell population progressively expanded, eventually constituting more than 70% of cells in culture by 5 months. Sox10 transgene expression was essential for generating O4+ cells but was insufficient for inducing a full oligodendroglial phenotype. Converted cells required continuous transgene expression to maintain their glial phenotype. Some vestigial characteristics of mesenchymal cells were maintained after conversion. Growth factor withdrawal and triiodothyronine (T3) supplementation generated mature oligodendroglial phenotypes, while FBS supplementation produced GFAP+- and p75NTR+-rich cultures. Converted cells also showed functional characteristics of neural-derived OPCs, such as the expression of AMPA, NMDA, kainate, and dopaminergic receptors, as well as similar metabolic responses to differentiation-inducing drugs. When co-cultured with rat dorsal root ganglion neurons, the converted cells differentiated and ensheathed multiple axons. We propose that functional oligodendroglia can be efficiently generated from adult rat mesenchymal cells by direct phenotypic conversion.
Collapse
Affiliation(s)
- Lara Vellosillo
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Center for Biomedical Technology (CTB), Universidad Politécnica, Madrid, Spain
| | - Jorge Pascual-Guerra
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Maria Paz Muñoz
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Antonio Rodríguez-Navarro
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | | | - Luis Carlos Barrio
- Unidad de Neurología Experimental, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Maria del Val Toledo Lobo
- Departamento de Biomedicina y Biotecnología, IRYCIS, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Carlos Luis Paíno
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Center for Biomedical Technology (CTB), Universidad Politécnica, Madrid, Spain
- *Correspondence: Carlos Luis Paíno,
| |
Collapse
|
11
|
Kim WK, Kim WH, Kweon OK, Kang BJ. Heat-Shock Proteins Can Potentiate the Therapeutic Ability of Cryopreserved Mesenchymal Stem Cells for the Treatment of Acute Spinal Cord Injury in Dogs. Stem Cell Rev Rep 2022; 18:1461-1477. [PMID: 35001344 DOI: 10.1007/s12015-021-10316-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are applied in the treatment of spinal cord injury (SCI) because of their neural tissue restoring ability. In the clinical setting, intravenous injection of cryopreserved cells is essential for the immediate treatment of SCI, exhibiting the disadvantage of reduced cell properties. METHODS In this study, we potentiated the characteristics of cryopreserved MSCs by heat-shock (HS) treatment to induce the expression of HS protein (HSP) HSP70/HSP27 and further improved antioxidant capacity by overexpressing HSP32 (heme oxygenase-1 [HO-1]). We randomly assigned 12 beagle dogs with acute SCI into three groups and transplanted cells intravenously: (i) F-MSCs (MSCs in frozen/thawed conditions); (ii) F-HSP-MSCs (HS-treated MSCs in frozen/thawed conditions); and (iii) F-HSP-HO-MSCs (HO-1-overexpressing and HS-treated MSCs in frozen/thawed conditions). RESULTS The potentiated MSCs exhibited increased growth factor-, anti-inflammatory-, antioxidant-, homing- and stemness-related gene expression. In the animal experiments, the HSP-induced groups showed significant improvement in hind-limb locomotion, highly expressed neural markers, less intervened fibrotic changes, and improved myelination. In particular, the HO-1-overexpression group was more prominent, controlling the initial inflammatory response with high antioxidant capabilities, suggesting that antioxidation was important to prevent secondary injury. Accordingly, HSPs not only successfully increased the ability of frozen MSCs but also demonstrated excellent neural protection and regeneration capacity in the case of acute SCI. CONCLUSIONS The application of HSP-induced cryopreserved MSCs in first-aid treatment for acute SCI is considered to help early neural sparing and further hind-limb motor function restoration.
Collapse
Affiliation(s)
- Woo Keyoung Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.,BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, South Korea
| | - Wan Hee Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.,BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, South Korea
| | - Oh-Kyeong Kweon
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea. .,BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
12
|
Cartarozzi LP, Perez M, Fernandes GG, Chiarotto GB, Luzo ÂCM, Campos AC, Kirchhoff F, de Oliveira ALR. Neuroprotection and gliosis attenuation by intravenous application of human mesenchymal stem cells (hMSC) following ventral root crush in mice. Mol Cell Neurosci 2021; 118:103694. [PMID: 34954382 DOI: 10.1016/j.mcn.2021.103694] [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: 06/09/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 11/15/2022] Open
Abstract
Rupture and stretching of spinal roots are common incidents that take place in high-energy accidents. The proximal axotomy of motoneurons by crushing of ventral roots is directly related to the degeneration of half of the lesioned population within the first two weeks. Moreover, only a small percentage of surviving motoneurons can successfully achieve regeneration after such a proximal lesion, and new treatments are necessary to improve this scenario. In this sense, mesenchymal stem cells (MSC) are of great interest once they secrete a broad spectrum of bioactive molecules that are immunomodulatory and can restore the environment after a lesion. The present work aimed at studying the effects of human mesenchymal stem cells (hMSC) therapy after ventral root crush (VRC) in mice. We evaluated motoneuron survival, glial reaction, and synapse preservation at the ventral horn. For this purpose, C57BL/6 J were submitted to a crush procedure of L4 to L6 ventral roots and treated with a single intravenous injection of adipose-derived hMSC. Evaluation of the results was carried out at 7, 14, and 28 days after injury. Analysis of motoneuron survival and astrogliosis showed that hMSC treatment resulted in higher motoneuron preservation (motoneuron survival ipsi/contralateral ratio: VRC group = 53%, VRC + hMSC group = 66%; p < 0.01), combined with reduction of astrogliosis (ipsi/contralateral GFAP immunolabeling: VRC group = 470%, VRC + hMSC group = 250%; p < 0.001). The morphological classification and Sholl analysis of microglial activation revealed that hMSC treatment reduced type V and increased type II profiles, indicating an enhancement of surveying over activated microglial cells. The glial reactivity modulation directly influenced synaptic inputs in apposition to axotomized motoneurons. In the hMSC-treated group, synaptic maintenance was increased (ipsi/contralateral synaptophysin immunolabeling: VRC group = 53%, VRC + hMSC group = 64%; p < 0.05). Overall, the present data show that intravenous injection of hMSC has neuroprotective and anti-inflammatory effects, decreasing reactive astrogliosis, and microglial reaction. Also, such cell therapy results in motoneuron preservation, combined with significant maintenance of spinal cord circuits, in particular those related to the ventral horn.
Collapse
Affiliation(s)
- Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil
| | - Matheus Perez
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Gabriel Gripp Fernandes
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Gabriela Bortolança Chiarotto
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil
| | - Ângela Cristina Malgeiros Luzo
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Alline Cristina Campos
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Building 48, 66421 Homburg, Germany
| | - Alexandre Leite Rodrigues de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
13
|
Li H, Yuan F, Du Y, Pan T, Wen W, Li S, Wang L, Lu A. Umbilical cord blood stem cells transplantation in a patient with severe progressive supranuclear palsy: a case report. J Med Case Rep 2021; 15:574. [PMID: 34844635 PMCID: PMC8628425 DOI: 10.1186/s13256-021-03139-z] [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: 05/21/2020] [Accepted: 10/01/2021] [Indexed: 12/05/2022] Open
Abstract
Background Progressive supranuclear palsy is a neurodegenerative condition that worsens over time. Given the lack of targeted treatments, patients with severe progressive supranuclear palsy have very low life expectancy. Case presentation We present a case of a 61-year-old Chinese man with severe progressive supranuclear palsy and treated with umbilical cord blood stem cells transplantation. After the umbilical cord blood stem cells therapy, his neurologic symptoms stopped deteriorating, his muscle rigidity was mildly improved, and he remains alive for more than 8 years. Conclusions Umbilical cord blood stem cells transplantation may be an alternative therapy for patients with severe progressive supranuclear palsy.
Collapse
Affiliation(s)
- Huiping Li
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Fang Yuan
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yaming Du
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Tao Pan
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Wanxin Wen
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Shaoxue Li
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Lixin Wang
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Aili Lu
- Department of Neurocritical Care, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
14
|
Warnecke A, Harre J, Shew M, Mellott AJ, Majewski I, Durisin M, Staecker H. Successful Treatment of Noise-Induced Hearing Loss by Mesenchymal Stromal Cells: An RNAseq Analysis of Protective/Repair Pathways. Front Cell Neurosci 2021; 15:656930. [PMID: 34887728 PMCID: PMC8650824 DOI: 10.3389/fncel.2021.656930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are an adult derived stem cell-like population that has been shown to mediate repair in a wide range of degenerative disorders. The protective effects of MSCs are mainly mediated by the release of growth factors and cytokines thereby modulating the diseased environment and the immune system. Within the inner ear, MSCs have been shown protective against tissue damage induced by sound and a variety of ototoxins. To better understand the mechanism of action of MSCs in the inner ear, mice were exposed to narrow band noise. After exposure, MSCs derived from human umbilical cord Wharton's jelly were injected into the perilymph. Controls consisted of mice exposed to sound trauma only. Forty-eight hours post-cell delivery, total RNA was extracted from the cochlea and RNAseq performed to evaluate the gene expression induced by the cell therapy. Changes in gene expression were grouped together based on gene ontology classification. A separate cohort of animals was treated in a similar fashion and allowed to survive for 2 weeks post-cell therapy and hearing outcomes determined. Treatment with MSCs after severe sound trauma induced a moderate hearing protective effect. MSC treatment resulted in an up-regulation of genes related to immune modulation, hypoxia response, mitochondrial function and regulation of apoptosis. There was a down-regulation of genes related to synaptic remodeling, calcium homeostasis and the extracellular matrix. Application of MSCs may provide a novel approach to treating sound trauma induced hearing loss and may aid in the identification of novel strategies to protect hearing.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Clinic for Otolaryngology–Head & Neck Surgery, Hanover Medical School, Hanover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation (EXC 2177/1), Oldenburg, Germany
| | - Jennifer Harre
- Clinic for Otolaryngology–Head & Neck Surgery, Hanover Medical School, Hanover, Germany
- Cluster of Excellence “Hearing4all” of the German Research Foundation (EXC 2177/1), Oldenburg, Germany
| | - Matthew Shew
- Department of Otolaryngology–Head & Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | | | - Igor Majewski
- Clinic for Otolaryngology–Head & Neck Surgery, Hanover Medical School, Hanover, Germany
| | - Martin Durisin
- Clinic for Otolaryngology–Head & Neck Surgery, Hanover Medical School, Hanover, Germany
| | - Hinrich Staecker
- Department of Otolaryngology–Head & Neck Surgery, University of Kansas School of Medicine, Kansas City, KS, United States
| |
Collapse
|
15
|
Lale Ataei M, Karimipour M, Shahabi P, Pashaei-Asl R, Ebrahimie E, Pashaiasl M. The Restorative Effect of Human Amniotic Fluid Stem Cells on Spinal Cord Injury. Cells 2021; 10:cells10102565. [PMID: 34685545 PMCID: PMC8534241 DOI: 10.3390/cells10102565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition within the neural system which is clinically manifested by sensory-motor dysfunction, leading, in some cases, to neural paralysis for the rest of the patient’s life. In the current study, mesenchymal stem cells (MSCs) were isolated from the human amniotic fluid, in order to study their juxtacrine and paracrine activities. Flow cytometry analysis was performed to identify the MSCs. A conditioned medium (CM) was collected to measure the level of BDNF, IL-1β, and IL-6 proteins using the ELISA assay. Following the SCI induction, MSCs and CM were injected into the lesion site, and also CM was infused intraperitoneally in the different groups. Two weeks after SCI induction, the spinal cord samples were examined to evaluate the expression of the doublecortin (DCX) and glial fibrillary acid protein (GFAP) markers using immunofluorescence staining. The MSCs’ phenotype was confirmed upon the expression and un-expression of the related CD markers. Our results show that MSCs increased the expression level of the DCX and decreased the level of the GFAP relative to the injury group (p < 0.001). Additionally, the CM promoted the DCX expression rate (p < 0.001) and decreased the GFAP expression rate (p < 0.01) as compared with the injury group. Noteworthily, the restorative potential of the MSCs was higher than that of the CM (p < 0.01). Large-scale meta-analysis of transcriptomic data highlighted PAK5, ST8SIA3, and NRXN1 as positively coexpressed genes with DCX. These genes are involved in neuroactive ligand–receptor interaction. Overall, our data revealed that both therapeutic interventions could promote the regeneration and restoration of the damaged neural tissue by increasing the rate of neuroblasts and decreasing the astrocytes.
Collapse
Affiliation(s)
- Maryam Lale Ataei
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| | - Roghiyeh Pashaei-Asl
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran 1417653911, Iran;
| | - Esmaeil Ebrahimie
- School of Life Sciences, College of Science, Health and Engineering, La Trobe University, Melbourne, VIC 3086, Australia;
- Genomics Research Platform, Research & Industry Engagement, La Trobe University, Melbourne, VIC 3086, Australia
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA 5371, Australia
- School of BioSciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Maryam Pashaiasl
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz 5166614766, Iran
- Women’s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
- Correspondence: ; Tel.: +98-41-33348573
| |
Collapse
|
16
|
Perez M, Cartarozzi LP, Chiarotto GB, Guimarães FS, Oliveira ALRD. Short and long-term neuroprotective effects of cannabidiol after neonatal peripheral nerve axotomy. Neuropharmacology 2021; 197:108726. [PMID: 34303725 DOI: 10.1016/j.neuropharm.2021.108726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/05/2021] [Accepted: 07/18/2021] [Indexed: 11/18/2022]
Abstract
Neonatal rat sciatic nerve crush mimics obstetric axonotmesis, leading to extensive loss of motor and sensory neurons. The present study aimed to investigate the neuroprotective potential of cannabidiol (CBD) and the role of cannabinoid receptors after sciatic nerve crush in neonatal rats. For that, two-day-old Wistar rats were used, organized into the following experimental groups: sciatic nerve crush plus CBD treatment (CBD), crush plus vehicle treatment (VE), crush + CBD + AM251 treatment (AM251 - CB1 inverse agonist), crush + CBD + AM630 treatment (AM630 - CB2 antagonist). Spinal motoneuron survival was evaluated by Nissl staining of the lumbar spinal cord, 5- and 56-days following injury. CBD treatment enhanced neuronal survival by ~54 % both 5 days and 8 weeks after injury. However, AM251 and AM630 treatment decreased neuronal rescue by 30 % when compared to the CBD group, suggesting that CBD acts partially through such pathways. However, in the long term, only the CB1 blockade reverted CBD positive results. Synaptic preservation was evaluated by anti-synaptophysin immunolabeling. Five days after the lesion, CBD treatment preserved ~35 % of synapses in the ventral horn, and such effect was partially reversed by CB1 inactivation. Additionally, CBD treatment reduced astroglial reaction both at 5 days (39 %, compared to VE) and 8 weeks (31 %, compared to VE) after lesion. The microglial response was acutely reduced by 62 % after CBD treatment. Overall, the results herein show that CBD is neuroprotective, increasing neuronal survival and reducing glial reaction after neonatal axotomy. Such effects require CB1 and CB2 receptors to be effective, in turn influencing neuroprotection, glial reactivity, and functional recovery.
Collapse
Affiliation(s)
- Matheus Perez
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970, Campinas, SP, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907, Ribeirão Preto, SP, Brazil
| | - Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970, Campinas, SP, Brazil
| | - Gabriela Bortolança Chiarotto
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970, Campinas, SP, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907, Ribeirão Preto, SP, Brazil
| | - Alexandre Leite Rodrigues de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, 13083-970, Campinas, SP, Brazil.
| |
Collapse
|
17
|
Bonilla P, Hernandez J, Giraldo E, González-Pérez MA, Alastrue-Agudo A, Elkhenany H, Vicent MJ, Navarro X, Edel M, Moreno-Manzano V. Human-Induced Neural and Mesenchymal Stem Cell Therapy Combined with a Curcumin Nanoconjugate as a Spinal Cord Injury Treatment. Int J Mol Sci 2021; 22:5966. [PMID: 34073117 PMCID: PMC8198521 DOI: 10.3390/ijms22115966] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
We currently lack effective treatments for the devastating loss of neural function associated with spinal cord injury (SCI). In this study, we evaluated a combination therapy comprising human neural stem cells derived from induced pluripotent stem cells (iPSC-NSC), human mesenchymal stem cells (MSC), and a pH-responsive polyacetal-curcumin nanoconjugate (PA-C) that allows the sustained release of curcumin. In vitro analysis demonstrated that PA-C treatment protected iPSC-NSC from oxidative damage in vitro, while MSC co-culture prevented lipopolysaccharide-induced activation of nuclear factor-κB (NF-κB) in iPSC-NSC. Then, we evaluated the combination of PA-C delivery into the intrathecal space in a rat model of contusive SCI with stem cell transplantation. While we failed to observe significant improvements in locomotor function (BBB scale) in treated animals, histological analysis revealed that PA-C-treated or PA-C and iPSC-NSC + MSC-treated animals displayed significantly smaller scars, while PA-C and iPSC-NSC + MSC treatment induced the preservation of β-III Tubulin-positive axons. iPSC-NSC + MSC transplantation fostered the preservation of motoneurons and myelinated tracts, while PA-C treatment polarized microglia into an anti-inflammatory phenotype. Overall, the combination of stem cell transplantation and PA-C treatment confers higher neuroprotective effects compared to individual treatments.
Collapse
Affiliation(s)
- Pablo Bonilla
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
| | - Joaquim Hernandez
- Neuroplasticity and Regeneration Group, Department Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona and CIBERNED, 08193 Bellaterra, Spain; (J.H.); (X.N.)
| | - Esther Giraldo
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
- Department of Biotechnology, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Miguel A. González-Pérez
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
| | - Ana Alastrue-Agudo
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
| | - Hoda Elkhenany
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria 22785, Egypt
| | - María J. Vicent
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain;
| | - Xavier Navarro
- Neuroplasticity and Regeneration Group, Department Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona and CIBERNED, 08193 Bellaterra, Spain; (J.H.); (X.N.)
| | - Michael Edel
- Laboratory of Regenerative Medicine, Institut Barraquer, 08021 Barcelona, Spain;
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (P.B.); (E.G.); (M.A.G.-P.); (A.A.-A.); (H.E.)
| |
Collapse
|
18
|
Rodríguez-Sánchez DN, Pinto GBA, Cartarozzi LP, de Oliveira ALR, Bovolato ALC, de Carvalho M, da Silva JVL, Dernowsek JDA, Golim M, Barraviera B, Ferreira RS, Deffune E, Bertanha M, Amorim RM. 3D-printed nerve guidance conduits multi-functionalized with canine multipotent mesenchymal stromal cells promote neuroregeneration after sciatic nerve injury in rats. Stem Cell Res Ther 2021; 12:303. [PMID: 34051869 PMCID: PMC8164252 DOI: 10.1186/s13287-021-02315-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/29/2021] [Indexed: 01/09/2023] Open
Abstract
Background Nerve injuries are debilitating, leading to long-term motor deficits. Remyelination and axonal growth are supported and enhanced by growth factor and cytokines. Combination of nerve guidance conduits (NGCs) with adipose-tissue-derived multipotent mesenchymal stromal cells (AdMSCs) has been performing promising strategy for nerve regeneration. Methods 3D-printed polycaprolactone (PCL)-NGCs were fabricated. Wistar rats subjected to critical sciatic nerve damage (12-mm gap) were divided into sham, autograft, PCL (empty NGC), and PCL + MSCs (NGC multi-functionalized with 106 canine AdMSCs embedded in heterologous fibrin biopolymer) groups. In vitro, the cells were characterized and directly stimulated with interferon-gamma to evaluate their neuroregeneration potential. In vivo, the sciatic and tibial functional indices were evaluated for 12 weeks. Gait analysis and nerve conduction velocity were analyzed after 8 and 12 weeks. Morphometric analysis was performed after 8 and 12 weeks following lesion development. Real-time PCR was performed to evaluate the neurotrophic factors BDNF, GDNF, and HGF, and the cytokine and IL-10. Immunohistochemical analysis for the p75NTR neurotrophic receptor, S100, and neurofilament was performed with the sciatic nerve. Results The inflammatory environment in vitro have increased the expression of neurotrophins BDNF, GDNF, HGF, and IL-10 in canine AdMSCs. Nerve guidance conduits multi-functionalized with canine AdMSCs embedded in HFB improved functional motor and electrophysiological recovery compared with PCL group after 12 weeks. However, the results were not significantly different than those obtained using autografts. These findings were associated with a shift in the regeneration process towards the formation of myelinated fibers. Increased immunostaining of BDNF, GDNF, and growth factor receptor p75NTR was associated with the upregulation of BDNF, GDNF, and HGF in the spinal cord of the PCL + MSCs group. A trend demonstrating higher reactivity of Schwann cells and axonal branching in the sciatic nerve was observed, and canine AdMSCs were engrafted at 30 days following repair. Conclusions 3D-printed NGCs multi-functionalized with canine AdMSCs embedded in heterologous fibrin biopolymer as cell scaffold exerted neuroregenerative effects. Our multimodal approach supports the trophic microenvironment, resulting in a pro-regenerative state after critical sciatic nerve injury in rats.
Collapse
Affiliation(s)
- Diego Noé Rodríguez-Sánchez
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Giovana Boff Araujo Pinto
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Luciana Politti Cartarozzi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Ana Livia Carvalho Bovolato
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University, Botucatu, SP, Brazil
| | - Marcio de Carvalho
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Jorge Vicente Lopes da Silva
- Renato Archer Information Technology Center (CTI), Three-dimensional Technologies Research Group, Campinas, SP, Brazil
| | - Janaina de Andréa Dernowsek
- Renato Archer Information Technology Center (CTI), Three-dimensional Technologies Research Group, Campinas, SP, Brazil
| | - Marjorie Golim
- Hemocenter division of Botucatu Medical School, São Paulo State University, Botucatu, SP, Brazil
| | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Elenice Deffune
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University, Botucatu, SP, Brazil
| | - Mathues Bertanha
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University, Botucatu, SP, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
19
|
Zhou H, Lu S, Li K, Yang Y, Hu C, Wang Z, Wang Q, He Y, Wang X, Ye D, Guan Q, Zang J, Liu C, Qu S, Luan Z. Study on the Safety of Human Oligodendrocyte Precursor Cell Transplantation in Young Animals and Its Efficacy on Myelination. Stem Cells Dev 2021; 30:587-600. [PMID: 33823616 PMCID: PMC8165470 DOI: 10.1089/scd.2021.0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) can differentiate into myelinating oligodendrocytes during embryonic development, thereby representing an important potential source for myelin repair or regeneration. To the best of our knowledge, there are very few OPCs from human sources (human-derived OPCs [hOPCs]). In this study, we aimed to evaluate the safety and remyelination capacity of hOPCs developed in our laboratory, transplanted into the lateral ventricles of young animals. Several acute and chronic toxicity experiments were conducted in which different doses of hOPCs were transplanted into the lateral ventricles of Sprague–Dawley rats of different ages. The toxicity, biodistribution, and tumor formation ability of the injected hOPCs were examined by evaluating the rats' vital signs, developmental indicators, neural reflexes, as well as by hematology, immunology, and pathology. In addition, the hOPCs were transplanted into the corpus callosum of the shiverer mouse to verify cell myelination efficacy. Overall, our results show that transplanted hOPCs into young mice are nontoxic to their organ function or immune system. The transplanted cells engrafted in the brain and did not appear in other organs, nor did they cause tissue proliferation or tumor formation. In terms of efficacy, the transplanted hOPCs were able to form myelin in the corpus callosum, alleviate the trembling phenotype of shiverer mice, and promote normal development. The transplantation of hOPCs is safe; they can effectively form myelin in the brain, thereby providing a theoretical basis for the future clinical transplantation of hOPCs.
Collapse
Affiliation(s)
- Haipeng Zhou
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Siliang Lu
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Ke Li
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yinxiang Yang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Caiyan Hu
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhaoyan Wang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Wang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Ying He
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Xiaohua Wang
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dou Ye
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Guan
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jing Zang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Chang Liu
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Suqing Qu
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zuo Luan
- The Second Clinical College, Southern Medical University, Guangzhou, China.,Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
Andrzejewska A, Dabrowska S, Lukomska B, Janowski M. Mesenchymal Stem Cells for Neurological Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002944. [PMID: 33854883 PMCID: PMC8024997 DOI: 10.1002/advs.202002944] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/23/2020] [Indexed: 05/13/2023]
Abstract
Neurological disorders are becoming a growing burden as society ages, and there is a compelling need to address this spiraling problem. Stem cell-based regenerative medicine is becoming an increasingly attractive approach to designing therapies for such disorders. The unique characteristics of mesenchymal stem cells (MSCs) make them among the most sought after cell sources. Researchers have extensively studied the modulatory properties of MSCs and their engineering, labeling, and delivery methods to the brain. The first part of this review provides an overview of studies on the application of MSCs to various neurological diseases, including stroke, traumatic brain injury, spinal cord injury, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, Parkinson's disease, and other less frequently studied clinical entities. In the second part, stem cell delivery to the brain is focused. This fundamental but still understudied problem needs to be overcome to apply stem cells to brain diseases successfully. Here the value of cell engineering is also emphasized to facilitate MSC diapedesis, migration, and homing to brain areas affected by the disease to implement precision medicine paradigms into stem cell-based therapies.
Collapse
Affiliation(s)
- Anna Andrzejewska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Sylwia Dabrowska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Barbara Lukomska
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
| | - Miroslaw Janowski
- NeuroRepair DepartmentMossakowski Medical Research CentrePASWarsaw02‐106Poland
- Center for Advanced Imaging ResearchDepartment of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of MarylandBaltimoreMD21201‐1595USA
- Tumor Immunology and Immunotherapy ProgramUniversity of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of MarylandBaltimoreMD21201‐1595USA
| |
Collapse
|
21
|
Badyra B, Sułkowski M, Milczarek O, Majka M. Mesenchymal stem cells as a multimodal treatment for nervous system diseases. Stem Cells Transl Med 2020; 9:1174-1189. [PMID: 32573961 PMCID: PMC7519763 DOI: 10.1002/sctm.19-0430] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological disorders are a massive challenge for modern medicine. Apart from the fact that this group of diseases is the second leading cause of death worldwide, the majority of patients have no access to any possible effective and standardized treatment after being diagnosed, leaving them and their families helpless. This is the reason why such great emphasis is being placed on the development of new, more effective methods to treat neurological patients. Regenerative medicine opens new therapeutic approaches in neurology, including the use of cell-based therapies. In this review, we focus on summarizing one of the cell sources that can be applied as a multimodal treatment tool to overcome the complex issue of neurodegeneration-mesenchymal stem cells (MSCs). Apart from the highly proven safety of this approach, beneficial effects connected to this type of treatment have been observed. This review presents modes of action of MSCs, explained on the basis of data from vast in vitro and preclinical studies, and we summarize the effects of using these cells in clinical trial settings. Finally, we stress what improvements have already been made to clarify the exact mechanism of MSCs action, and we discuss potential ways to improve the introduction of MSC-based therapies in clinics. In summary, we propose that more insightful and methodical optimization, by combining careful preparation and administration, can enable use of multimodal MSCs as an effective, tailored cell therapy suited to specific neurological disorders.
Collapse
Affiliation(s)
- Bogna Badyra
- Department of TransplantationJagiellonian University Medical CollegeCracowPoland
| | - Maciej Sułkowski
- Department of TransplantationJagiellonian University Medical CollegeCracowPoland
| | - Olga Milczarek
- Department of Children NeurosurgeryJagiellonian University Medical CollegeCracowPoland
| | - Marcin Majka
- Department of TransplantationJagiellonian University Medical CollegeCracowPoland
| |
Collapse
|
22
|
Suda S, Nito C, Yokobori S, Sakamoto Y, Nakajima M, Sowa K, Obinata H, Sasaki K, Savitz SI, Kimura K. Recent Advances in Cell-Based Therapies for Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21186718. [PMID: 32937754 PMCID: PMC7555943 DOI: 10.3390/ijms21186718] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is the most prevalent cardiovascular disease worldwide, and is still one of the leading causes of death and disability. Stem cell-based therapy is actively being investigated as a new potential treatment for certain neurological disorders, including stroke. Various types of cells, including bone marrow mononuclear cells, bone marrow mesenchymal stem cells, dental pulp stem cells, neural stem cells, inducible pluripotent stem cells, and genetically modified stem cells have been found to improve neurological outcomes in animal models of stroke, and there are some ongoing clinical trials assessing their efficacy in humans. In this review, we aim to summarize the recent advances in cell-based therapies to treat stroke.
Collapse
Affiliation(s)
- Satoshi Suda
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
- Correspondence: ; Tel.: +81-3-3822-2131; Fax: +81-3-3822-4865
| | - Chikako Nito
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Yuki Sakamoto
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Masataka Nakajima
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Kota Sowa
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Hirofumi Obinata
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Kazuma Sasaki
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Sean I. Savitz
- Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX 77030, USA;
| | - Kazumi Kimura
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| |
Collapse
|
23
|
Yang Y, Cao TT, Tian ZM, Gao H, Wen HQ, Pang M, He WJ, Wang NX, Chen YY, Wang Y, Li H, Lin JW, Kang Z, Li MM, Liu B, Rong LM. Subarachnoid transplantation of human umbilical cord mesenchymal stem cell in rodent model with subacute incomplete spinal cord injury: Preclinical safety and efficacy study. Exp Cell Res 2020; 395:112184. [PMID: 32707134 DOI: 10.1016/j.yexcr.2020.112184] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022]
Abstract
Functional multipotency renders human umbilical cord mesenchymal stem cell (hUC-MSC) a promising candidate for the treatment of spinal cord injury (SCI). However, its safety and efficacy have not been fully understood for clinical translation. In this study, we performed cellular, kinematic, physiological, and anatomical analyses, either in vitro or in vivo, to comprehensively evaluate the safety and efficacy associated with subarachnoid transplantation of hUC-MSCs in rats with subacute incomplete SCI. Concerning safety, hUC-MSCs were shown to have normal morphology, excellent viability, steady proliferation, typical biomarkers, stable karyotype in vitro, and no tumorigenicity both in vitro and in vivo. Following subarachnoid transplantation of hUC-MSCs in the subject rodents, the biodistribution of hUC-MSCs was restricted to the spinal cord, and no toxicity to immune system or organ function was observed. Body weight, organ weight, and the ratio of the latter upon the former between stem cell-transplanted rats and placebo-injected rats revealed no statistical differences. Regarding efficacy, hUC-MSCs could differentiate into osteoblasts, chondrocytes, adipocytes and neural progenitor cells in vitro. While in vivo studies revealed that subarachnoid transplantation of stem cells resulted in significant improvement in locomotion, earlier automatic micturition recovery and reduced lesion size, which correlated with increased regeneration of tracking fiber and reduced parenchymal inflammation. In vivo luminescence imaging showed that a few of the transplanted luciferase-labeled hUC-MSCs tended to migrate towards the lesion epicenter. Shortened latency and enhanced amplitude were also observed in both motor and sensory evoked potentials, indicating improved signal conduction in the damaged site. Immunofluorescent staining confirmed that a few of the administrated hUC-MSCs integrated into the spinal cord parenchyma and differentiated into astrocytes and oligodendrocytes, but not neurons. Moreover, decreased astrogliosis, increased remyelination, and neuron regeneration could be observed. To the best of our knowledge, this preclinical study provides detailed safety and efficacy evidence regarding intrathecal transplantation of hUC-MSCs in treating SCI for the first time and thus, supports its initiation in the following clinical trial.
Collapse
Affiliation(s)
- Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Ting-Ting Cao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhen-Ming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Han Gao
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Hui-Quan Wen
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Wei-Jie He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Nan-Xiang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yu-Yong Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - He Li
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China
| | - Jun-Wei Lin
- Department of Obstetrics, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhuang Kang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Mang-Mang Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China.
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| | - Li-Min Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
24
|
Rocha LA, Gomes ED, Afonso JL, Granja S, Baltazar F, Silva NA, Shoichet MS, Sousa RA, Learmonth DA, Salgado AJ. In vitro Evaluation of ASCs and HUVECs Co-cultures in 3D Biodegradable Hydrogels on Neurite Outgrowth and Vascular Organization. Front Cell Dev Biol 2020; 8:489. [PMID: 32612997 PMCID: PMC7308435 DOI: 10.3389/fcell.2020.00489] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular disruption following spinal cord injury (SCI) decisively contributes to the poor functional recovery prognosis facing patients with the condition. Using a previously developed gellan gum hydrogel to which the adhesion motif GRGDS was grafted (GG-GRGDS), this work aimed to understand the ability of adipose-derived stem cells (ASCs) to impact vascular organization of human umbilical vein endothelial cells (HUVECs), and how this in turn affects neurite outgrowth of dorsal root ganglia (DRG) explants. Our data shows that culturing these cells together lead to a synergistic effect as showed by increased stimulation of neuritogenesis on DRG. Importantly, HUVECs were only able to assemble into vascular-like structures when cultured in the presence of ASCs, which shows the capacity of these cells in reorganizing the vascular milieu. Analysis of selected neuroregulatory molecules showed that the co-culture upregulated the secretion of several neurotrophic factors. On the other hand, ASCs, and ASCs + HUVECs presented a similar profile regarding the presence of angiotrophic molecules herein analyzed. Finally, the implantation of GG-GRGDS hydrogels encapsulating ASCs in the chick chorioallantoic membrane (CAM) lead to increases in vascular recruitment toward the hydrogels in comparison to GG-GRGDS alone. This indicates that the combination of ASCs with GG-GRGDS hydrogels could promote re-vascularization in trauma-related injuries in the central nervous system and thus control disease progression and induce functional recovery.
Collapse
Affiliation(s)
- Luís A Rocha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal.,Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - João L Afonso
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Fatima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Molly S Shoichet
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rui A Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - David A Learmonth
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| |
Collapse
|
25
|
Wang X, Ye L, Zhang K, Gao L, Xiao J, Zhang Y. Upregulation of microRNA-200a in bone marrow mesenchymal stem cells enhances the repair of spinal cord injury in rats by reducing oxidative stress and regulating Keap1/Nrf2 pathway. Artif Organs 2020; 44:744-752. [PMID: 31995644 DOI: 10.1111/aor.13656] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/13/2020] [Accepted: 01/22/2020] [Indexed: 12/13/2022]
Abstract
Spinal cord injury (SCI) is a common disease with high incidence, disability rate and treatment cost. microRNA (miR)-200a is reported to inhibit Keap1 to activate Nrf2 signaling. This study aimed to explore the effects of lentivirus-mediated miR-200a gene-modified bone marrow mesenchymal stem cells (BMSCs) transplantation on the repair of SCI in a rat model. BMSCs were isolated from the bone marrow of Sprague-Dawley rats. MiR-200a targeting to Keap1 was identified by luciferase reporter gene assay. The expressions of Keap1, nuclear factor erythroid 2-related factor 2 (Nrf2), NAD(P)H-dependent quinone oxidoreductase 1 (NQO-1), heme oxygenase-1 (HO-1) and glutamate-cysteine ligase catalytic subunit (GCLC) were detected by Western blotting in SCI rats. The locomotor capacity of the rats was evaluated using the Basso, Beattie, and Bresnahan scale. The levels of malondialdehyde (MDA), activities of superoxide dismutase (SOD), and catalase (CAT) were measured. miR-200a inhibited Keap-1 3' UTR activity in BMSCs. Transplantation of BMSCs with overexpression of miR-200a or si-Keap1 increased locomotor function recovery of rats after SCI, while decreased MDA level, increased SOD, CAT activities, and Nrf2 expression together with its downstream HO-1, NQO1, GCLC protein expressions in SCI rat. These results indicated that overexpressed miR-200a in BMSCs promoted SCI repair, which may be through regulating antioxidative signaling pathway.
Collapse
Affiliation(s)
- Xianxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lu Gao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jin Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yiquan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
26
|
Silvestro S, Bramanti P, Trubiani O, Mazzon E. Stem Cells Therapy for Spinal Cord Injury: An Overview of Clinical Trials. Int J Mol Sci 2020; 21:E659. [PMID: 31963888 PMCID: PMC7013533 DOI: 10.3390/ijms21020659] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) is a traumatic lesion that causes disability with temporary or permanent sensory and/or motor deficits. The pharmacological approach still in use for the treatment of SCI involves the employment of corticosteroid drugs. However, SCI remains a very complex disorder that needs future studies to find effective pharmacological treatments. SCI actives a strong inflammatory response that induces a loss of neurons followed by a cascade of events that lead to further spinal cord damage. Many experimental studies demonstrate the therapeutic effect of stem cells in SCI due to their capacity to differentiate into neuronal cells and by releasing neurotrophic factors. Therefore, they appear to be a valid strategy to use in the field of regenerative medicine. The purpose of this paper is to provide an overview of clinical trials, recorded in clinical trial.gov during 2005-2019, aimed to evaluate the use of stem cell-based therapy in SCI. The results available thus far show the safety and efficacy of stem cell therapy in patients with SCI. However, future trials are needed to investigate the safety and efficacy of stem cell transplantation.
Collapse
Affiliation(s)
- Serena Silvestro
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| | - Placido Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy;
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| |
Collapse
|
27
|
Zhu Q, Lu P. Stem Cell Transplantation for Amyotrophic Lateral Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1266:71-97. [PMID: 33105496 DOI: 10.1007/978-981-15-4370-8_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuronal degeneration disease, in which the death of motor neurons causes lost control of voluntary muscles. The consequence is weakness of muscles with a wide range of disabilities and eventually death. Most patients died within 5 years after diagnosis, and there is no cure for this devastating neurodegenerative disease up to date. Stem cells, including non-neural stem cells and neural stem cells (NSCs) or neural progenitor cells (NPCs), are very attractive cell sources for potential neuroprotection and motor neuron replacement therapy which bases on the idea that transplant-derived and newly differentiated motor neurons can replace lost motor neurons to re-establish voluntary motor control of muscles in ALS. Our recent studies show that transplanted NSCs or NPCs not only survive well in injured spinal cord, but also function as neuronal relays to receive regenerated host axonal connection and extend their own axons to host for connectivity, including motor axons in ventral root. This reciprocal connection between host neurons and transplanted neurons provides a strong rationale for neuronal replacement therapy for ALS to re-establish voluntary motor control of muscles. In addition, a variety of new stem cell resources and the new methodologies to generate NSCs or motor neuron-specific progenitor cells have been discovered and developed. Together, it provides the basis for motor neuron replacement therapy with NSCs or NPCs in ALS.
Collapse
Affiliation(s)
- Qiang Zhu
- Ludwig Institute, University of California - San Diego, La Jolla, CA, USA
| | - Paul Lu
- Veterans Administration San Diego Healthcare System, San Diego, CA, USA. .,Department of Neurosciences, University of California - San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Mukhamedshina Y, Shulman I, Ogurcov S, Kostennikov A, Zakirova E, Akhmetzyanova E, Rogozhin A, Masgutova G, James V, Masgutov R, Lavrov I, Rizvanov A. Mesenchymal Stem Cell Therapy for Spinal Cord Contusion: A Comparative Study on Small and Large Animal Models. Biomolecules 2019; 9:E811. [PMID: 31805639 PMCID: PMC6995633 DOI: 10.3390/biom9120811] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/05/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Here, we provide a first comparative study of the therapeutic potential of allogeneic mesenchymal stem cells derived from bone marrow (BM-MSCs), adipose tissue (AD-MSCs), and dental pulp (DP-MSCs) embedded in fibrin matrix, in small (rat) and large (pig) spinal cord injury (SCI) models during subacute period of spinal contusion. Results of behavioral, electrophysiological, and histological assessment as well as immunohistochemistry and real-time polymerase chain reaction analysis suggest that application of AD-MSCs combined with a fibrin matrix within the subacute period in rats (2 weeks after injury), provides significantly higher post-traumatic regeneration compared to a similar application of BM-MSCs or DP-MSCs. Within the rat model, use of AD-MSCs resulted in a marked change in: (1) restoration of locomotor activity and conduction along spinal axons; (2) reduction of post-traumatic cavitation and enhancing tissue retention; and (3) modulation of microglial and astroglial activation. The effect of an autologous application of AD-MSCs during the subacute period after spinal contusion was also confirmed in pigs (6 weeks after injury). Effects included: (1) partial restoration of the somatosensory spinal pathways; (2) reduction of post-traumatic cavitation and enhancing tissue retention; and (3) modulation of astroglial activation in dorsal root entry zone. However, pigs only partially replicated the findings observed in rats. Together, these results indicate application of AD-MSCs embedded in fibrin matrix at the site of SCI during the subacute period can facilitate regeneration of nervous tissue in rats and pigs. These results, for the first time, provide robust support for the use of AD-MSC to treat subacute SCI.
Collapse
Affiliation(s)
- Yana Mukhamedshina
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Histology, Cytology, and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Iliya Shulman
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Sergei Ogurcov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Alexander Kostennikov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Elena Zakirova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Elvira Akhmetzyanova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Alexander Rogozhin
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Neurology, Kazan State Medical Academy–Branch Campus of the Federal State Budgetary Edicational Institution of Father Professional Education «Russian Medical Academy of Continuous Professional Education», 420012 Kazan, Russia
| | - Galina Masgutova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Victoria James
- Division of Biomedical Science, School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK;
| | - Ruslan Masgutov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Igor Lavrov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Albert Rizvanov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| |
Collapse
|
29
|
Nogueira PAS, Pereira MP, Soares JJG, de Assis Silva Gomes J, Ribeiro DL, Razolli DS, Velloso LA, Neto MB, Zanon RG. Swimming reduces fatty acids-associated hypothalamic damage in mice. J Chem Neuroanat 2019; 103:101713. [PMID: 31726089 DOI: 10.1016/j.jchemneu.2019.101713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/20/2019] [Accepted: 11/09/2019] [Indexed: 12/28/2022]
Abstract
The arcuate and the paraventricular and lateral hypothalamic nuclei, related to hunger and satiety control, are generally compromised by excess fatty acids. In this situation, fatty acids cause inflammation via TLR4 (toll like receptor 4) and the nuclei become less responsive to the hormones leptin and insulin, contributing to the development of obesity. In this work, these nuclei were analyzed in animals fed with high-fat diet and submitted to swimming without and with load for two months. For this, frontal sections of the hypothalamus were immunolabelled with GFAP (glial fibrillary acidic protein), synaptophysin, IL-6 (interleukin 6) and TLR4. Also, proteins extracted from the hypothalamus were analyzed using Western blotting (GFAP and synaptophysin), fluorometric analysis for caspases 3 and 7, and CBA (cytometric bead array) for Th1, Th2, and Th17 profiles. The high-fat diet significantly caused overweight and, in the hypothalamus, decreased synapses and increased astrocytic reactivity. The swimming with load, especially 80 % of the maximum load, reduced those consequences. The high-fat diet increased TLR4 in the arcuate nucleus and the swimming exercise with 80 % of the maximum load showed a tendency of reducing this expression. Swimming did not significantly influence the inflammatory or anti-inflammatory cytokines in the hypothalamus or in plasma. The high-fat diet in sedentary animals increased the expression of caspases 3 and 7 and swimming practice reduced this increment to levels compatible with animals fed on a normal diet. The set of results conclude that the impact of swimming on the damage caused in the hypothalamus by a high-fat diet is positive. The different aspects analyzed in here point to better cellular viability and conservation of the synapses in the hypothalamic nuclei of overweight animals that practiced swimming with a load.
Collapse
Affiliation(s)
- Pedro Augusto Silva Nogueira
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil; Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil; Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil
| | - Miriam Pimenta Pereira
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | | | - Juliana de Assis Silva Gomes
- Laboratory of Cellular Interactions Biology, Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniele Lisboa Ribeiro
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Daniela Soares Razolli
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil; Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Licio Augusto Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil
| | - Morun Bernardino Neto
- Department of Basic and Environmental Sciences, University of São Paulo, Lorena, São Paulo, Brazil
| | - Renata Graciele Zanon
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil.
| |
Collapse
|
30
|
Faghih H, Javeri A, Amini H, Taha MF. Directed differentiation of human adipose tissue-derived stem cells to dopaminergic neurons in low-serum and serum-free conditions. Neurosci Lett 2019; 708:134353. [PMID: 31251959 DOI: 10.1016/j.neulet.2019.134353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/26/2022]
Abstract
Directing the fate of mesenchymal stem cells (MSCs) to dopaminergic neurons has great importance in both biomedical studies and cell therapy of Parkinson's disease. We recently generated dopamine-secreting cells from human adipose tissue-derived stem cells (hADSCs) by exposing the cells to a growth factor cocktail composed of SHH, bFGF, FGF8 and BDNF in low-serum condition. In the current study, we induced the cells by the same dopaminergic inducing cocktail in serum-free B27-supplemented Neurobasal medium. ADSCs differentiated in both conditions expressed several neuronal and dopaminergic markers. However, there were higher gene expression levels under the serum-free condition. Higher levels of TUJ1 and TH proteins were also detected in the cells exposed to the dopaminergic-inducing cocktail under serum-free Neurobasal condition. TH protein was expressed in about 28% and 60% of the cells differentiated in the low-serum and serum-free Neurobasal media, respectively. Moreover, the cells exposed to the dopaminergic-inducing cocktail in the serum-free Neurobasal condition released a more significant amount of dopamine in response to KCl-induced depolarization. Altogether, these findings show a greater efficiency of the serum-free Neurobasal condition for growth factor-directed differentiation of hADSCs to functional dopamine-secreting cells which may be valuable for transplantation therapy of Parkinson's disease in future.
Collapse
Affiliation(s)
- Hossein Faghih
- Department of Stem Cells and Regenerative medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Hossein Amini
- Department of Pharmacology, Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoumeh Fakhr Taha
- Department of Stem Cells and Regenerative medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
31
|
Mao Q, Nguyen PD, Shanti RM, Shi S, Shakoori P, Zhang Q, Le AD. Gingiva-Derived Mesenchymal Stem Cell-Extracellular Vesicles Activate Schwann Cell Repair Phenotype and Promote Nerve Regeneration. Tissue Eng Part A 2019; 25:887-900. [DOI: 10.1089/ten.tea.2018.0176] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Qin Mao
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Phuong D. Nguyen
- Division of Plastic and Reconstructive Surgery, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rabie M. Shanti
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Otolaryngology and Head and Neck Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shihong Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Pasha Shakoori
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Anh D. Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Allahdadi KJ, de Santana TA, Santos GC, Azevedo CM, Mota RA, Nonaka CK, Silva DN, Valim CXR, Figueira CP, dos Santos WLC, do Espirito Santo RF, Evangelista AF, Villarreal CF, dos Santos RR, de Souza BSF, Soares MBP. IGF-1 overexpression improves mesenchymal stem cell survival and promotes neurological recovery after spinal cord injury. Stem Cell Res Ther 2019; 10:146. [PMID: 31113444 PMCID: PMC6530133 DOI: 10.1186/s13287-019-1223-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/19/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Survival and therapeutic actions of bone marrow-derived mesenchymal stem cells (BMMSCs) can be limited by the hostile microenvironment present during acute spinal cord injury (SCI). Here, we investigated whether BMMSCs overexpressing insulin-like growth factor 1 (IGF-1), a cytokine involved in neural development and injury repair, improved the therapeutic effects of BMMSCs in SCI. METHODS Using a SCI contusion model in C57Bl/6 mice, we transplanted IGF-1 overexpressing or wild-type BMMSCs into the lesion site following SCI and evaluated cell survival, proliferation, immunomodulation, oxidative stress, myelination, and functional outcomes. RESULTS BMMSC-IGF1 transplantation was associated with increased cell survival and recruitment of endogenous neural progenitor cells compared to BMMSC- or saline-treated controls. Modulation of gene expression of pro- and anti-inflammatory mediators was observed after BMMSC-IGF1 and compared to saline- and BMMSC-treated mice. Treatment with BMMSC-IGF1 restored spinal cord redox homeostasis by upregulating antioxidant defense genes. BMMSC-IGF1 protected against SCI-induced myelin loss, showing more compact myelin 28 days after SCI. Functional analyses demonstrated significant gains in BMS score and gait analysis in BMMSC-IGF1, compared to BMMSC or saline treatment. CONCLUSIONS Overexpression of IGF-1 in BMMSC resulted in increased cell survival, immunomodulation, myelination, and functional improvements, suggesting that IGF-1 facilitates the regenerative actions of BMMSC in acute SCI.
Collapse
Affiliation(s)
- Kyan James Allahdadi
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- São Rafael Hospital, D’Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Thaís Alves de Santana
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | - Girlaine Café Santos
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | - Carine Machado Azevedo
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
| | - Roberta Alves Mota
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | - Carolina Kymie Nonaka
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- São Rafael Hospital, D’Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Daniela Nascimento Silva
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- São Rafael Hospital, D’Or Institute for Research and Education (IDOR), Salvador, Brazil
| | | | - Cláudio Pereira Figueira
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
| | - Washington Luis Conrado dos Santos
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | - Renan Fernandes do Espirito Santo
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | | | - Cristiane Flora Villarreal
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- Federal University of Bahia, UFBA, Salvador, BA Brazil
| | - Ricardo Ribeiro dos Santos
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ Brazil
| | - Bruno Solano Freitas de Souza
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ Brazil
- São Rafael Hospital, D’Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Milena Botelho Pereira Soares
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, BA Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia 40296-710 Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ Brazil
| |
Collapse
|
33
|
Rodríguez Sánchez DN, de Lima Resende LA, Boff Araujo Pinto G, de Carvalho Bovolato AL, Possebon FS, Deffune E, Amorim RM. Canine Adipose-Derived Mesenchymal Stromal Cells Enhance Neuroregeneration in a Rat Model of Sciatic Nerve Crush Injury. Cell Transplant 2019; 28:47-54. [PMID: 30369261 PMCID: PMC6322136 DOI: 10.1177/0963689718809045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Crush injuries in peripheral nerves are frequent and induce long-term disability with motor and sensory deficits. Due to axonal and myelin sheath disruptions, strategies for optimized axonal regeneration are needed. Multipotent mesenchymal stromal cells (MSC) are promising because of their anti-inflammatory properties and secretion of neurotrophins. The present study investigated the effect of canine adipose tissue MSC (Ad-MSC) transplantation in an experimental sciatic nerve crush injury. Wistar rats were divided into three groups: sham ( n = 8); Crush+PBS ( n = 8); Crush+MSC ( n = 8). Measurements of sciatic nerve functional index (SFI), muscle mass, and electromyography (EMG) were performed. Canine Ad-MSC showed mesodermal characteristics (CD34-, CD45-, CD44+, CD90+ and CD105+) and multipotentiality due to chondrogenic, adipogenic, and osteogenic differentiation. SFI during weeks 3 and 4 was significantly higher in the Crush+MSC group ( p < 0.001). During week 4, the EMG latency in the Crush+MSC groups had better near normality ( p < 0.05). The EMG amplitude showed results close to normality during week 4 in the Crush+MSC group ( p < 0.04). There were no statistical differences in muscle weight between the groups ( p > 0.05), but there was a tendency toward weight gain in the Crush+MSC groups. Better motor functional recovery after crush and perineural canine Ad-MSC transplantation was observed during week 2. This was maintained till week 4. In conclusion, the canine Ad-MSC transplantation showed early pro-regenerative effects between 2-4 weeks in the rat model of sciatic nerve crush injury.
Collapse
Affiliation(s)
- Diego Noé Rodríguez Sánchez
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Luiz Antonio de Lima Resende
- Department of Neurology and Psychiatry, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Giovana Boff Araujo Pinto
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ana Lívia de Carvalho Bovolato
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Fábio Sossai Possebon
- Department of Veterinary Hygiene and Public Health, College of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| | - Elenice Deffune
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
34
|
Mukhamedshina YO, Gracheva OA, Mukhutdinova DM, Chelyshev YA, Rizvanov AA. Mesenchymal stem cells and the neuronal microenvironment in the area of spinal cord injury. Neural Regen Res 2019; 14:227-237. [PMID: 30531002 PMCID: PMC6301181 DOI: 10.4103/1673-5374.244778] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell-based technologies are used as a therapeutic strategy in spinal cord injury (SCI). Mesenchymal stem cells (MSCs), which secrete various neurotrophic factors and cytokines, have immunomodulatory, anti-apoptotic and anti-inflammatory effects, modulate reactivity/phenotype of astrocytes and the microglia, thereby promoting neuroregeneration seem to be the most promising. The therapeutic effect of MSCs is due to a paracrine mechanism of their action, therefore the survival of MSCs and their secretory phenotype is of particular importance. Nevertheless, these data are not always reported in efficacy studies of MSC therapy in SCI. Here, we provide a review with summaries of preclinical trials data evaluating the efficacy of MSCs in animal models of SCI. Based on the data collected, we have tried (1) to establish the behavior of MSCs after transplantation in SCI with an evaluation of cell survival, migration potential, distribution in the area of injured and intact tissue and possible differentiation; (2) to determine the effects MSCs on neuronal microenvironment and correlate them with the efficacy of functional recovery in SCI; (3) to ascertain the conditions under which MSCs demonstrate their best survival and greatest efficacy.
Collapse
Affiliation(s)
- Yana O Mukhamedshina
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University; Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Olga A Gracheva
- Department of Therapy and Clinical Diagnostics with radiology Faculty of Veterinary Medicine, Bauman Kazan State Academy of Veterinary Medicine, Kazan, Russia
| | - Dina M Mukhutdinova
- Department of Therapy and Clinical Diagnostics with radiology Faculty of Veterinary Medicine, Bauman Kazan State Academy of Veterinary Medicine, Kazan, Russia
| | - Yurii A Chelyshev
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert A Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
35
|
Spejo AB, Teles CB, Zuccoli GDS, Oliveira ALRD. Synapse preservation and decreased glial reactions following ventral root crush (VRC) and treatment with 4‐hydroxy‐tempo (TEMPOL). J Neurosci Res 2018; 97:520-534. [DOI: 10.1002/jnr.24365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Aline Barroso Spejo
- Department of Structural and Functional Biology, Institute of Biology University of Campinas (UNICAMP) Campinas Brazil
| | - Caroline Brandão Teles
- Department of Structural and Functional Biology, Institute of Biology University of Campinas (UNICAMP) Campinas Brazil
| | - Giuliana da Silva Zuccoli
- Department of Structural and Functional Biology, Institute of Biology University of Campinas (UNICAMP) Campinas Brazil
| | | |
Collapse
|
36
|
Millán-Rivero JE, Nadal-Nicolás FM, García-Bernal D, Sobrado-Calvo P, Blanquer M, Moraleda JM, Vidal-Sanz M, Agudo-Barriuso M. Human Wharton's jelly mesenchymal stem cells protect axotomized rat retinal ganglion cells via secretion of anti-inflammatory and neurotrophic factors. Sci Rep 2018; 8:16299. [PMID: 30389962 PMCID: PMC6214908 DOI: 10.1038/s41598-018-34527-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation is emerging as an ideal tool to restore the wounded central nervous system (CNS). MSCs isolated from extra-embryonic tissues have some advantages compared to MSCs derived from adult ones, such as an improved proliferative capacity, life span, differentiation potential and immunomodulatory properties. In addition, they are more immunoprivileged, reducing the probability of being rejected by the recipient. Umbilical cords (UCs) are a good source of MSCs because they are abundant, safe, non-invasively harvested after birth and, importantly, they are not encumbered with ethical problems. Here we show that the intravitreal transplant of Wharton´s jelly mesenchymal stem cells isolated from three different human UCs (hWJMSCs) delays axotomy-induced retinal ganglion cell (RGC) loss. In vivo, hWJMSCs secrete anti-inflammatory molecules and trophic factors, the latter alone may account for the elicited neuroprotection. Interestingly, this expression profile differs between naive and injured retinas, suggesting that the environment in which the hWJMSCs are modulates their secretome. Finally, even though the transplant itself is not toxic for RGCs, it is not innocuous as it triggers a transient but massive infiltration of Iba1+cells from the choroid to the retina that alters the retinal structure.
Collapse
Affiliation(s)
- Jose E Millán-Rivero
- Unidad de Terapia Celular y Trasplante Hematopoyético. Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Dpto Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - Francisco M Nadal-Nicolás
- Dpto Oftalmología, Universidad de Murcia, Murcia, Spain.,Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - David García-Bernal
- Unidad de Terapia Celular y Trasplante Hematopoyético. Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Dpto Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - Paloma Sobrado-Calvo
- Dpto Oftalmología, Universidad de Murcia, Murcia, Spain.,Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Miguel Blanquer
- Unidad de Terapia Celular y Trasplante Hematopoyético. Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Dpto Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - Jose M Moraleda
- Unidad de Terapia Celular y Trasplante Hematopoyético. Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Dpto Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Dpto Oftalmología, Universidad de Murcia, Murcia, Spain.,Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Marta Agudo-Barriuso
- Dpto Oftalmología, Universidad de Murcia, Murcia, Spain. .,Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.
| |
Collapse
|
37
|
Nito C, Sowa K, Nakajima M, Sakamoto Y, Suda S, Nishiyama Y, Nakamura-Takahashi A, Nitahara-Kasahara Y, Ueda M, Okada T, Kimura K. Transplantation of human dental pulp stem cells ameliorates brain damage following acute cerebral ischemia. Biomed Pharmacother 2018; 108:1005-1014. [PMID: 30372800 DOI: 10.1016/j.biopha.2018.09.084] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 09/15/2018] [Accepted: 09/15/2018] [Indexed: 02/06/2023] Open
Abstract
AIMS Numerous experimental studies have shown that cellular therapy, including human dental pulp stem cells (DPSCs), is an attractive strategy for ischemic brain injury. Herein, we examined the effects of intravenous DPSC administration after transient middle cerebral artery occlusion in rats. METHODS Male Sprague-Dawley rats received a transient 90 min middle cerebral artery occlusion. DPSCs (1 × 106 cells) or vehicle were administered via the femoral vein at 0 h or 3 h after ischemia-reperfusion. PKH26, a red fluorescent cell linker, was used to track the transplanted cells in the brain. Infarct volume, neurological deficits, and immunological analyses were performed at 24 h and 72 h after reperfusion. RESULTS PKH26-positive cells were observed more frequently in the ipsilateral than the contralateral hemisphere. DPSCs transplanted at 0 h after reperfusion significantly reduced infarct volume and reversed motor deficits at 24 h and 72 h recovery. DPSCs transplanted at 3 h after reperfusion also significantly reduced infarct volume and improved motor function compared with vehicle groups at 24 h and 72 h recovery. Further, DPSC transplantation significantly inhibited microglial activation and pro-inflammatory cytokine expression compared with controls at 72 h after reperfusion. Moreover, DPSCs attenuated neuronal degeneration in the cortical ischemic boundary area. CONCLUSIONS Systemic delivery of human DPSCs after reperfusion reduced ischemic damage and improved functional recovery in a rodent ischemia model, with a clinically relevant therapeutic window. The neuroprotective action of DPSCs may relate to the modulation of neuroinflammation during the acute phase of stroke.
Collapse
Affiliation(s)
- Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan.
| | - Kota Sowa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Masataka Nakajima
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Yuki Sakamoto
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Yasuhiro Nishiyama
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Aki Nakamura-Takahashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Pharmacology, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan; Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| |
Collapse
|
38
|
Spejo AB, Chiarotto GB, Ferreira ADF, Gomes DA, Ferreira RS, Barraviera B, Oliveira ALR. Neuroprotection and immunomodulation following intraspinal axotomy of motoneurons by treatment with adult mesenchymal stem cells. J Neuroinflammation 2018; 15:230. [PMID: 30107848 PMCID: PMC6092804 DOI: 10.1186/s12974-018-1268-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/02/2018] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Treatment of spinal cord injury is dependent on neuronal survival, appropriate synaptic circuit preservation, and inflammatory environment management. In this sense, mesenchymal stem cell (MSC) therapy is a promising tool that can reduce glial reaction and provide trophic factors to lesioned neurons. METHODS Lewis adult female rats were submitted to a unilateral ventral funiculus cut at the spinal levels L4, L5, and L6. The animals were divided into the following groups: IA (intramedullary axotomy), IA + DMEM (Dulbecco's modified Eagle's medium), IA + FS (fibrin sealant), IA + MSC (106 cells), and IA + FS + MSC (106 cells). Seven days after injury, qPCR (n = 5) was performed to assess gene expression of VEGF, BDNF, iNOS2, arginase-1, TNF-α, IL-1β, IL-6, IL-10, IL-4, IL-13, and TGF-β. The cellular infiltrate at the lesion site was analyzed by hematoxylin-eosin (HE) staining and immunohistochemistry (IH) for Iba1 (microglia and macrophage marker) and arginase-1. Fourteen days after injury, spinal alpha motor neurons (MNs), evidenced by Nissl staining (n = 5), were counted. For the analysis of astrogliosis in spinal lamina IX and synaptic detachment around lesioned motor neurons (GAP-43-positive cells), anti-GFAP and anti-synaptophysin immunohistochemistry (n = 5) was performed, respectively. Twenty-eight days after IA, the gait of the animals was evaluated by the walking track test (CatWalk; n = 7). RESULTS The site of injury displayed strong monocyte infiltration, containing arginase-1-expressing macrophages. The FS-treated group showed upregulation of iNOS2, arginase-1, proinflammatory cytokine (TNF-α and IL-1β), and antiinflammatory cytokine (IL-10, IL-4, and IL-13) expression. Thus, FS enhanced early macrophage recruitment and proinflammatory cytokine expression, which accelerated inflammation. Rats treated with MSCs displayed high BDNF-positive immunolabeling, suggesting local delivery of this neurotrophin to lesioned motoneurons. This BDNF expression may have contributed to the increased neuronal survival and synapse preservation and decreased astrogliosis observed 14 days after injury. At 28 days after lesion, gait recovery was significantly improved in MSC-treated animals compared to that in the other groups. CONCLUSIONS Overall, the present data demonstrate that MSC therapy is neuroprotective and, when associated with a FS, shifts the immune response to a proinflammatory profile.
Collapse
Affiliation(s)
- A. B. Spejo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP Brazil
| | - G. B. Chiarotto
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP Brazil
| | - A. D. F. Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil
| | - D. A. Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil
| | - R. S. Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP Brazil
| | - B. Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP Brazil
| | - A. L. R. Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP Brazil
| |
Collapse
|
39
|
Mukhamedshina YO, Akhmetzyanova ER, Kostennikov AA, Zakirova EY, Galieva LR, Garanina EE, Rogozin AA, Kiassov AP, Rizvanov AA. Adipose-Derived Mesenchymal Stem Cell Application Combined With Fibrin Matrix Promotes Structural and Functional Recovery Following Spinal Cord Injury in Rats. Front Pharmacol 2018; 9:343. [PMID: 29692732 PMCID: PMC5902567 DOI: 10.3389/fphar.2018.00343] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/26/2018] [Indexed: 01/05/2023] Open
Abstract
The use of stem and progenitor cells to restore damaged organs and tissues, in particular, the central nervous system, is currently considered a most promising therapy in regenerative medicine. At the same time, another approach aimed at stimulating regeneration with the use of stem cells encapsulated into a biopolymer matrix and capable of creating a specific microenvironment for the implanted cells similar to the natural extracellular matrix is under active development. Here, we study effects of the application of adipose-derived mesenchymal stem cells (AD-MSCs) combined with a fibrin matrix on post-traumatic reactions in the spinal cord in rats. The AD-MSC application is found to exert a positive impact on the functional and structural recovery after spinal cord injury (SCI) that has been confirmed by the results of behavioral/electrophysiological and morphometric studies demonstrating reduced area of abnormal cavities and enhanced tissue retention in the site of injury. Immunohistochemical and real-time PCR analyses provide evidence that AD-MSC application decreases the GFAP expression in the area of SCI that might indicate the reduction of astroglial activation. Our results also demonstrate that AD-MSC application contributes to marked upregulation of PDGFβR and HSPA1b mRNA expression and decrease of Iba1 expression at the site of the central canal. Thus, the application of AD-MSCs combined with fibrin matrix at the site of SCI during the subacute period can stimulate important mechanisms of nervous tissue regeneration and should be further developed for clinical applications.
Collapse
Affiliation(s)
- Yana O Mukhamedshina
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Elvira R Akhmetzyanova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Alexander A Kostennikov
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Elena Y Zakirova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Luisa R Galieva
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ekaterina E Garanina
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Alexander A Rogozin
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Neurology and Manual Therapy, Kazan State Medical Academy, Kazan, Russia
| | - Andrey P Kiassov
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
40
|
Zheng Z, Zhang L, Qu Y, Xiao G, Li S, Bao S, Lu QR, Mu D. Mesenchymal Stem Cells Protect Against Hypoxia-Ischemia Brain Damage by Enhancing Autophagy Through Brain Derived Neurotrophic Factor/Mammalin Target of Rapamycin Signaling Pathway. Stem Cells 2018; 36:1109-1121. [PMID: 29451335 DOI: 10.1002/stem.2808] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 12/17/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious disease for neonates. However, present therapeutic strategies are not effective enough for treating HIE. Previous study showed that mesenchymal stem cells (MSCs) can exert neuroprotective effects for brain damage, but its mechanism remains elusive. Using in vitro coculture of rat cortical primary neurons and MSCs in HI conditions, we demonstrated that MSCs help increase brain derived neurotrophic factor (BDNF) and autophagy markers (LC3II and Beclin1) in the cultures and decrease cells death (lactate dehydrogenase levels). We demonstrated a similar mechanism using an in vivo rat model of HI in combination with MSCs transplantation. Using a behavioral study, we further showed that MSCs transplantation into the rat brain after HI injury can attenuate behavioral deficits. Finally, we found that the increase in BDNF and autophagy related factors after HI injury combined with MSCs transplantation can be reversed by anti-BDNF treatment and strengthen the point that the protective effects of BDNF work through inhibition of the mammalin target of rapamycin (mTOR) pathway. Collectively, we proposed that coculture/transplantation of MSCs after HI injury leads to increased BDNF expression and a subsequent reduction in mTOR pathway activation that results in increased autophagy and neuroprotection. This finding gives a hint to explore new strategies for treating neonates with HIE. Stem Cells 2018;36:1109-1121.
Collapse
Affiliation(s)
- Zhen Zheng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China.,Department of Pediatrics, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Guoguang Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Shan Bao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Q Richard Lu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
41
|
Distribution and Survival of Transplanted Adipose-Derived Mesenchymal Stem Cells in the Spinal Cord Injury. BIONANOSCIENCE 2017. [DOI: 10.1007/s12668-017-0440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
42
|
Nistor M, Behringer W, Schmidt M, Schiffner R. A Systematic Review of Neuroprotective Strategies during Hypovolemia and Hemorrhagic Shock. Int J Mol Sci 2017; 18:E2247. [PMID: 29072635 PMCID: PMC5713217 DOI: 10.3390/ijms18112247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023] Open
Abstract
Severe trauma constitutes a major cause of death and disability, especially in younger patients. The cerebral autoregulatory capacity only protects the brain to a certain extent in states of hypovolemia; thereafter, neurological deficits and apoptosis occurs. We therefore set out to investigate neuroprotective strategies during haemorrhagic shock. This review was performed in accordance to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Before the start of the search, a review protocol was entered into the PROSPERO database. A systematic literature search of Pubmed, Web of Science and CENTRAL was performed in August 2017. Results were screened and evaluated by two researchers based on a previously prepared inclusion protocol. Risk of bias was determined by use of SYRCLE's risk of bias tool. The retrieved results were qualitatively analysed. Of 9093 results, 119 were assessed in full-text form, 16 of them ultimately adhered to the inclusion criteria and were qualitatively analyzed. We identified three subsets of results: (1) hypothermia; (2) fluid therapy and/or vasopressors; and (3) other neuroprotective strategies (piracetam, NHE1-inhibition, aprotinin, human mesenchymal stem cells, remote ischemic preconditioning and sevoflurane). Overall, risk of bias according to SYRCLE's tool was medium; generally, animal experimental models require more rigorous adherence to the reporting of bias-free study design (randomization, etc.). While the individual study results are promising, the retrieved neuroprotective strategies have to be evaluated within the current scientific context-by doing so, it becomes clear that specific promising neuroprotective strategies during states of haemorrhagic shock remain sparse. This important topic therefore requires more in-depth research.
Collapse
Affiliation(s)
- Marius Nistor
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany.
| | - Wilhelm Behringer
- Emergency Department, Jena University Hospital, 07747 Jena, Germany.
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, 07747 Jena, Germany.
| | - René Schiffner
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany.
- Orthopedic Department, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
43
|
Pu Y, Meng K, Gu C, Wang L, Zhang X. Thrombospondin-1 modified bone marrow mesenchymal stem cells (BMSCs) promote neurite outgrowth and functional recovery in rats with spinal cord injury. Oncotarget 2017; 8:96276-96289. [PMID: 29221205 PMCID: PMC5707099 DOI: 10.18632/oncotarget.22018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/23/2017] [Indexed: 01/02/2023] Open
Abstract
Stem cell therapies are currently gaining momentum in the treatment of spinal cord injury (SCI). However, unsatisfied intrinsic neurite growth capacity constitutes significant obstacles for injured spinal cord repair and ultimately results in neurological dysfunction. The present study assessed the efficacy of thrombospondin-1 (TSP-1), a neurite outgrowth-promoting molecule, modified bone marrow mesenchymal stem cells (BMSCs) on promoting neurite outgrowth in vitro and in vivo of Oxygen–Glucose Deprivation (OGD) treated motor neurons and SCI rat models. The present results demonstrated that the treatment of BMSCs+TSP-1 could promote the neurite length, neuronal survival, and functional recovery after SCI. Additionally, TSP-1 could activate transforming growth factor-β1 (TGF-β1) then induced the smad2 phosphorylation, and expedited the expression of GAP-43 to promote neurite outgrowth. The present study for the first time demonstrated that BMSCs+TSP-1 could promote neurite outgrowth and functional recovery after SCI partly through the TGF-β1/p-Samd2 pathway. The study provided a novel encouraging evidence for the potential treatment of BMSCs modification with TSP-1 in patients with SCI.
Collapse
Affiliation(s)
- Yujie Pu
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ke Meng
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chuanlong Gu
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaoming Zhang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
44
|
Dynamics of plasma membrane surface related to the release of extracellular vesicles by mesenchymal stem cells in culture. Sci Rep 2017; 7:6767. [PMID: 28754913 PMCID: PMC5533874 DOI: 10.1038/s41598-017-07265-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 06/28/2017] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (exosomes and shedding vesicles) released by mesenchymal stem cells (MSCs) are regarded as a storable, cell-free alternative with comparable therapeutic potential to their parent cells. Shedding vesicles originate as bulges on the cell surface but little is known about their turnover or how their formation can be stimulated. We have used atomic force microscopy (AFM) to follow the formation dynamics of bulges in living adipose tissue-derived MSCs. AFM images showed that, in general, MSCs present hundreds of nanosized protrusions on their surface with life spans of 10-20 min. Scanning electron microscopy confirmed those images and showed that bulges are also formed on filamentous processes. Extracellular vesicles deposited on the culture surface have comparable sizes to those of bulges showing up on the cell surface. The amount of protrusions on cells treated with progesterone or PDGF-BB, two treatments that stimulate the secretion of extracellular vesicles in MSCs, was evaluated by AFM. Measurements of the cross-area at 50 nm over the cell surface provided estimates of the amount of protrusions and showed that these values increased with the stimulating treatments. Our study suggests that shedding vesicles constitute a large population of the extracellular vesicle pool.
Collapse
|
45
|
Neuron-Specific Fluorescence Reporter-Based Live Cell Tracing for Transdifferentiation of Mesenchymal Stem Cells into Neurons by Chemical Compound. Stem Cells Int 2017; 2017:8452830. [PMID: 28808446 PMCID: PMC5541830 DOI: 10.1155/2017/8452830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Although transdifferentiation of mesenchymal stem cells (MSCs) into neurons increases the possibility of therapeutic use of MSCs for neurodevelopmental disorders, the use of MSCs has the limitation on differentiation efficiency to neuronal lineage and lack of an easy method to monitor the transdifferentiation. In this study, using time-lapse live cell imaging, we assessed the neuronal differentiation of MSCs induced by a small molecule “NHPDQC (N-hydroxy-2-oxo-3-(3-phenylprophyl)-1,2-dihydroquinoxaline-6-carboxamide, C18H17N3O3).” Plasmid vector containing red fluorescence reporter genes under the control of the tubulin α1 (Tα1) promoter (pTα1-DsRed2) traced the neuronal differentiation of MSCs. Two days after NHPDQC treatment, MSCs showed neuron-like phenotype with neurite outgrowth and high expression of neuron-specific markers in more than 95% cells. The fluorescence signals increased in the cytoplasm of pTα1-DsRed2-transfected MSCs after NHPDQC treatment. In vitro monitoring of MSCs along the time courses showed progressive increase of fluorescence till 30 h after treatment, corresponding with the increase in neurite length. We examined an efficient neuronal differentiation of MSCs by NHPDQC alone and monitored the temporal changes of neuronal differentiation by neuron-specific fluorescence reporter along time. This method would help further our understanding of the differentiation of MSCs to produce neurons by simple treatment of small molecule.
Collapse
|
46
|
Nakajima M, Nito C, Sowa K, Suda S, Nishiyama Y, Nakamura-Takahashi A, Nitahara-Kasahara Y, Imagawa K, Hirato T, Ueda M, Kimura K, Okada T. Mesenchymal Stem Cells Overexpressing Interleukin-10 Promote Neuroprotection in Experimental Acute Ischemic Stroke. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:102-111. [PMID: 28725658 PMCID: PMC5502709 DOI: 10.1016/j.omtm.2017.06.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-10 is a contributing factor to neuroprotection of mesenchymal stem cell (MSC) transplantation after ischemic stroke. Our aim was to increase therapeutic effects by combining MSCs and ex vivo IL-10 gene transfer with an adeno-associated virus (AAV) vector using a rat transient middle cerebral artery occlusion (MCAO) model. Sprague-Dawley rats underwent 90 min MCAO followed by intravenous administration of MSCs alone or IL-10 gene-transferred MSCs (MSC/IL-10) at 0 or 3 hr after ischemia reperfusion. Infarct lesions, neurological deficits, and immunological analyses were performed within 7 days after MCAO. 0-hr transplantation of MSCs alone and MSC/IL-10 significantly reduced infarct volumes and improved motor function. Conversely, 3-hr transplantation of MSC/IL-10, but not MSCs alone, significantly reduced infarct volumes (p < 0.01) and improved motor function (p < 0.01) compared with vehicle groups at 72 hr and 7 days after MCAO. Immunological analysis showed that MSC/IL-10 transplantation significantly inhibits microglial activation and pro-inflammatory cytokine expression compared with MSCs alone. Moreover, overexpressing IL-10 suppressed neuronal degeneration and improved survival of engrafted MSCs in the ischemic hemisphere. These results suggest that overexpressing IL-10 enhances the neuroprotective effects of MSC transplantation by anti-inflammatory modulation and thereby supports neuronal survival during the acute ischemic phase.
Collapse
Affiliation(s)
- Masataka Nakajima
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Kota Sowa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Yasuhiro Nishiyama
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Aki Nakamura-Takahashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | | | - Tohru Hirato
- JCR Pharmaceuticals Co., Ltd., Hyogo, 659-0021, Japan
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
47
|
Abstract
Spinal cord injury (SCI) represents one of the most complicated and heterogeneous pathological processes of central nervous system (CNS) impairments, which is still beyond functional regeneration. Transplantation of mesenchymal stem cells (MSCs) has been shown to promote the repair of the injured spinal cord tissues in animal models, and therefore, there is much interest in the clinical use of these cells. However, many questions which are essential to improve the therapy effects remain unanswered. For instance, the functional roles and related molecular regulatory mechanisms of MSCs in vivo are not yet completely determined. It is important for transplanted cells to migrate into the injured tissue, to survive and undergo neural differentiation, or to play neural protection roles by various mechanisms after SCI. In this review, we will focus on some of the recent knowledge about the biological behavior and function of MSCs in SCI. Meanwhile, we highlight the function of biomaterials to direct the behavior of MSCs based on our series of work on silk fibroin biomaterials and attempt to emphasize combinational strategies such as tissue engineering for functional improvement of SCI.
Collapse
|
48
|
Jiang LH, Mousawi F, Yang X, Roger S. ATP-induced Ca 2+-signalling mechanisms in the regulation of mesenchymal stem cell migration. Cell Mol Life Sci 2017; 74:3697-3710. [PMID: 28534085 PMCID: PMC5597679 DOI: 10.1007/s00018-017-2545-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/03/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
The ability of cells to migrate to the destined tissues or lesions is crucial for physiological processes from tissue morphogenesis, homeostasis and immune responses, and also for stem cell-based regenerative medicines. Cytosolic Ca2+ is a primary second messenger in the control and regulation of a wide range of cell functions including cell migration. Extracellular ATP, together with the cognate receptors on the cell surface, ligand-gated ion channel P2X receptors and a subset of G-protein-coupled P2Y receptors, represents common autocrine and/or paracrine Ca2+ signalling mechanisms. The P2X receptor ion channels mediate extracellular Ca2+ influx, whereas stimulation of the P2Y receptors triggers intracellular Ca2+ release from the endoplasmic reticulum (ER), and activation of both type of receptors thus can elevate the cytosolic Ca2+ concentration ([Ca2+]c), albeit with different kinetics and capacity. Reduction in the ER Ca2+ level following the P2Y receptor activation can further induce store-operated Ca2+ entry as a distinct Ca2+ influx pathway that contributes in ATP-induced increase in the [Ca2+]c. Mesenchymal stem cells (MSC) are a group of multipotent stem cells that grow from adult tissues and hold promising applications in tissue engineering and cell-based therapies treating a great and diverse number of diseases. There is increasing evidence to show constitutive or evoked ATP release from stem cells themselves or mature cells in the close vicinity. In this review, we discuss the mechanisms for ATP release and clearance, the receptors and ion channels participating in ATP-induced Ca2+ signalling and the roles of such signalling mechanisms in mediating ATP-induced regulation of MSC migration.
Collapse
Affiliation(s)
- Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK. .,Sino-UK Joint Laboratory of Brain Function and Injury, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China. .,Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, 37032, Tours, France.
| | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Xuebin Yang
- Department of Oral Biology, University of Leeds, WTBB, St James University Hospital, Leeds, LS97TF, UK
| | - Sėbastien Roger
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, 37032, Tours, France
| |
Collapse
|
49
|
Ropper AE, Thakor DK, Han I, Yu D, Zeng X, Anderson JE, Aljuboori Z, Kim SW, Wang H, Sidman RL, Zafonte RD, Teng YD. Defining recovery neurobiology of injured spinal cord by synthetic matrix-assisted hMSC implantation. Proc Natl Acad Sci U S A 2017; 114:E820-E829. [PMID: 28096400 PMCID: PMC5293074 DOI: 10.1073/pnas.1616340114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal stem cells (MSCs) isolated from adult tissues offer tangible potential for regenerative medicine, given their feasibility for autologous transplantation. MSC research shows encouraging results in experimental stroke, amyotrophic lateral sclerosis, and neurotrauma models. However, further translational progress has been hampered by poor MSC graft survival, jeopardizing cellular and molecular bases for neural repair in vivo. We have devised an adult human bone marrow MSC (hMSC) delivery formula by investigating molecular events involving hMSCs incorporated in a uniquely designed poly(lactic-co-glycolic) acid scaffold, a clinically safe polymer, following inflammatory exposures in a dorsal root ganglion organotypic coculture system. Also, in rat T9-T10 hemisection spinal cord injury (SCI), we demonstrated that the tailored scaffolding maintained hMSC stemness, engraftment, and led to robust motosensory improvement, neuropathic pain and tissue damage mitigation, and myelin preservation. The scaffolded nontransdifferentiated hMSCs exerted multimodal effects of neurotrophism, angiogenesis, neurogenesis, antiautoimmunity, and antiinflammation. Hindlimb locomotion was restored by reestablished integrity of submidbrain circuits of serotonergic reticulospinal innervation at lumbar levels, the propriospinal projection network, neuromuscular junction, and central pattern generator, providing a platform for investigating molecular events underlying the repair impact of nondifferentiated hMSCs. Our approach enabled investigation of recovery neurobiology components for injured adult mammalian spinal cord that are different from those involved in normal neural function. The uncovered neural circuits and their molecular and cellular targets offer a biological underpinning for development of clinical rehabilitation therapies to treat disabilities and complications of SCI.
Collapse
Affiliation(s)
- Alexander E Ropper
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Devang K Thakor
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - InBo Han
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Dou Yu
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Xiang Zeng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Jamie E Anderson
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Zaid Aljuboori
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Soo-Woo Kim
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
| | - Hongjun Wang
- Biomedical Engineering, Chemistry, and Biological Sciences, Stevens Institute of Technology, Hoboken, NJ 07030
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School/Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Ross D Zafonte
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
| | - Yang D Teng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130;
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| |
Collapse
|
50
|
Abstract
Stem cells, especially neural stem cells (NSCs), are a very attractive cell source for potential reconstruction of injured spinal cord though either neuroprotection, neural regeneration, remyelination, replacement of lost neural cells, or reconnection of disrupted axons. The later have great potential since recent studies demonstrate long-distance growth and connectivity of axons derived from transplanted NSCs after spinal cord injury (SCI). In addition, transplanted NSCs constitute a permissive environment for host axonal regeneration and serve as new targets for host axonal connection. This reciprocal connection between grafted neurons and host neurons constitutes a neuronal relay formation that could restore functional connectivity after SCI.
Collapse
|