1
|
D'Souza RS, Her YF, Hussain N, Karri J, Schatman ME, Calodney AK, Lam C, Buchheit T, Boettcher BJ, Chang Chien GC, Pritzlaff SG, Centeno C, Shapiro SA, Klasova J, Grider JS, Hubbard R, Ege E, Johnson S, Epstein MH, Kubrova E, Ramadan ME, Moreira AM, Vardhan S, Eshraghi Y, Javed S, Abdullah NM, Christo PJ, Diwan S, Hassett LC, Sayed D, Deer TR. Evidence-Based Clinical Practice Guidelines on Regenerative Medicine Treatment for Chronic Pain: A Consensus Report from a Multispecialty Working Group. J Pain Res 2024; 17:2951-3001. [PMID: 39282657 PMCID: PMC11402349 DOI: 10.2147/jpr.s480559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Injectable biologics have not only been described and developed to treat dermal wounds, cardiovascular disease, and cancer, but have also been reported to treat chronic pain conditions. Despite emerging evidence supporting regenerative medicine therapy for pain, many aspects remain controversial. Methods The American Society of Pain and Neuroscience (ASPN) identified the educational need for an evidence-based guideline on regenerative medicine therapy for chronic pain. The executive board nominated experts spanning multiple specialties including anesthesiology, physical medicine and rehabilitation, and sports medicine based on expertise, publications, research, and clinical practice. A steering committee selected preliminary questions, which were reviewed and refined. Evidence was appraised using the United States Preventive Services Task Force (USPSTF) criteria for evidence level and degree of recommendation. Using a modified Delphi approach, consensus points were distributed to all collaborators and each collaborator voted on each point. If collaborators provided a decision of "disagree" or "abstain", they were invited to provide a rationale in a non-blinded fashion to the committee chair, who incorporated the respective comments and distributed revised versions to the committee until consensus was achieved. Results Sixteen questions were selected for guideline development. Questions that were addressed included type of injectable biologics and mechanism, evidence in treating chronic pain indications (eg, tendinopathy, muscular pathology, osteoarthritis, intervertebral disc disease, neuropathic pain), role in surgical augmentation, dosing, comparative efficacy between injectable biologics, peri-procedural practices to optimize therapeutic response and quality of injectate, federal regulations, and complications with mitigating strategies. Conclusion In well-selected individuals with certain chronic pain indications, use of injectable biologics may provide superior analgesia, functionality, and/or quality of life compared to conventional medical management or placebo. Future high-quality randomized clinical trials are warranted with implementation of minimum reporting standards, standardization of preparation protocols, investigation of dose-response associations, and comparative analysis between different injectable biologics.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yeng F Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Jay Karri
- Departments of Orthopedic Surgery and Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael E Schatman
- Department of Anesthesiology, Perioperative Care, & Pain Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Christopher Lam
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Thomas Buchheit
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Brennan J Boettcher
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Scott G Pritzlaff
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Sacramento, CA, USA
| | | | - Shane A Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jay S Grider
- Department of Anesthesiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ryan Hubbard
- Department of Sports Medicine, Anderson Orthopedic Clinic, Arlington, VA, USA
| | - Eliana Ege
- Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Shelby Johnson
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Max H Epstein
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Mohamed Ehab Ramadan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alexandra Michelle Moreira
- Department of Physical Medicine & Rehabilitation, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | - Swarnima Vardhan
- Department of Internal Medicine, Yale New Haven Health - Bridgeport Hospital, Bridgeport, CT, USA
| | - Yashar Eshraghi
- Department of Anesthesiology & Critical Care Medicine, Ochsner Health System, New Orleans, LA, USA
| | - Saba Javed
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Newaj M Abdullah
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Paul J Christo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sudhir Diwan
- Department of Pain Medicine, Advanced Spine on Park Avenue, New York City, NY, USA
| | | | - Dawood Sayed
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy R Deer
- Department of Anesthesiology and Pain Medicine, West Virginia University School of Medicine, Charleston, WV, USA
| |
Collapse
|
2
|
Zou XF, Zhang BZ, Qian WW, Cheng FM. Bone marrow mesenchymal stem cells in treatment of peripheral nerve injury. World J Stem Cells 2024; 16:799-810. [PMID: 39219723 PMCID: PMC11362854 DOI: 10.4252/wjsc.v16.i8.799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
Peripheral nerve injury (PNI) is a common neurological disorder and complete functional recovery is difficult to achieve. In recent years, bone marrow mesenchymal stem cells (BMSCs) have emerged as ideal seed cells for PNI treatment due to their strong differentiation potential and autologous transplantation ability. This review aims to summarize the molecular mechanisms by which BMSCs mediate nerve repair in PNI. The key mechanisms discussed include the differentiation of BMSCs into multiple types of nerve cells to promote repair of nerve injury. BMSCs also create a microenvironment suitable for neuronal survival and regeneration through the secretion of neurotrophic factors, extracellular matrix molecules, and adhesion molecules. Additionally, BMSCs release pro-angiogenic factors to promote the formation of new blood vessels. They modulate cytokine expression and regulate macrophage polarization, leading to immunomodulation. Furthermore, BMSCs synthesize and release proteins related to myelin sheath formation and axonal regeneration, thereby promoting neuronal repair and regeneration. Moreover, this review explores methods of applying BMSCs in PNI treatment, including direct cell transplantation into the injured neural tissue, implantation of BMSCs into nerve conduits providing support, and the application of genetically modified BMSCs, among others. These findings confirm the potential of BMSCs in treating PNI. However, with the development of this field, it is crucial to address issues related to BMSC therapy, including establishing standards for extracting, identifying, and cultivating BMSCs, as well as selecting application methods for BMSCs in PNI such as direct transplantation, tissue engineering, and genetic engineering. Addressing these issues will help translate current preclinical research results into clinical practice, providing new and effective treatment strategies for patients with PNI.
Collapse
Affiliation(s)
- Xiong-Fei Zou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Bao-Zhong Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China.
| | - Wen-Wei Qian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Florence Mei Cheng
- College of Nursing, The Ohio State University, Ohio, OH 43210, United States
| |
Collapse
|
3
|
Zhang J, Wu P, Wen Q. Optimization strategies for mesenchymal stem cell-based analgesia therapy: a promising therapy for pain management. Stem Cell Res Ther 2024; 15:211. [PMID: 39020426 PMCID: PMC11256674 DOI: 10.1186/s13287-024-03828-8] [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: 04/27/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
Pain is a very common and complex medical problem that has a serious impact on individuals' physical and mental health as well as society. Non-steroidal anti-inflammatory drugs and opioids are currently the main drugs used for pain management, but they are not effective in controlling all types of pain, and their long-term use can cause adverse effects that significantly impair patients' quality of life. Mesenchymal stem cells (MSCs) have shown great potential in pain treatment. However, limitations such as the low proliferation rate of MSCs in vitro and low survival rate in vivo restrict their analgesic efficacy and clinical translation. In recent years, researchers have explored various innovative approaches to improve the therapeutic effectiveness of MSCs in pain treatment. This article reviews the latest research progress of MSCs in pain treatment, with a focus on methods to enhance the analgesic efficacy of MSCs, including engineering strategies to optimize the in vitro culture environment of MSCs and to improve the in vivo delivery efficiency of MSCs. We also discuss the unresolved issues to be explored in future MSCs and pain research and the challenges faced by the clinical translation of MSC therapy, aiming to promote the optimization and clinical translation of MSC-based analgesia therapy.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Ping Wu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
4
|
Gao Q, Gao Z, Su M, Huang Y, Zhang C, Li C, Zhan H, Liu B, Zhou X. Umbilical Cord Mesenchymal Stem Cells Overexpressing Heme Oxygenase-1 Promotes Symptoms Recovery in Cystitis Rats by Alleviating Neuroinflammation. Stem Cells Int 2023; 2023:8887091. [PMID: 38020203 PMCID: PMC10663085 DOI: 10.1155/2023/8887091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) seriously reduces the patient's quality of life, yet current therapies only provide partial relief. In the spinal dorsal horn (SDH), neuroinflammation plays a pivotal role in the development of IC. Injection of human umbilical cord mesenchymal stem cells (hUMSCs) to reduce inflammation is an effective strategy, and heme oxygenase-1 (HO-1) exhibits anti-nociceptive effect in neuroinflammatory pain. This study aimed to test the therapeutic effects of hUMSCs overexpressing HO-1 on cyclophosphamide-induced cystitis rat model. Cystitis rats were transplanted with altered cells and then assessed for 3 weeks. A series of behavioral measurements would be trial including suprapubic mechanical allodynia, depressive-like behaviors, micturition frequency, and short-term memory function. Additionally, western blot, immunofluorescence staining, and ELISA kit test for anti-inflammation effect. HUMSCs were capable of being transduced to overexpress HO-1. Injection of hUMSCs overexpressing HO-1 was more effective than hUMSCs alone in alleviating behavioral symptoms in rats. Furthermore, hUMSCs overexpressing HO-1 inhibited the activation of glial and TLR4/p65/NLRP3 pathway, decreased the levels of pro-inflammatory cytokines in the SDH region. Surprisingly, it markedly increased anti-inflammatory cytokine IL-10, reduced MDA content, and protected GSH concentrations in local environment. Our results suggest that injecting hUMSCs overexpressing HO-1 intrathecally can significantly promote functional outcomes in cystitis rats by reducing neuroinflammation, at least, partly through downregulating TLR4/p65/NLRP3 signaling pathway in the SDH region. This cell therapy affords a new strategy for IC/BPS treatment.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Zhentao Gao
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Minzhi Su
- Department of Rehabilitation, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Yong Huang
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Chi Zhang
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Cuiping Li
- Department of Biotherapy Center, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Hailun Zhan
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Bolong Liu
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| |
Collapse
|
5
|
Yerofeyeva AV, Pinchuk SV, Rjabceva SN, Molchanova AY. The role of cannabinoid CB1 receptors in the antinociceptive and reparative actions of mesenchymal stem cells in rats with peripheral neuropathic pain. IBRAIN 2023; 9:245-257. [PMID: 37786759 PMCID: PMC10527798 DOI: 10.1002/ibra.12129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 10/04/2023]
Abstract
Mesenchymal stem cells (MSCs) can produce antinociceptive and reparative effects. Presumably, the MSCs-induced antinociception may be partly due to the involvement of the endocannabinoid system. The study aimed to evaluate the antinociceptive and reparative effects of adipose-derived MSCs (ADMSCs) upon pharmacological modulation of cannabinoid CB1 receptor in peripheral tissues or on ADMSCs' membranes in a rat model of peripheral neuropathy. ADMSCs were injected into the area of rat sciatic nerve injury (i) with no additional treatments, (ii) at the tissue CB1 receptor activation by endogenous agonist anandamide (AEA) or blockade with a selective AM251 antagonist; and (iii) preincubated with AEA or AM251. The evaluation of CB1 receptor activity involved analyzing nociceptive responses, gait parameters, and histology. Transplantation of ADMSCs upon activation of CB1 receptors, both on AMSCs' membranes or in the area of nerve injury, accelerated the analgesia and recovery of dynamic gait parameters, abolished static gait disturbances, and promoted the fastest nerve regeneration. Only blockade of CB1 receptors on ADMSCs shortened ADMSCs-induced analgesia and decreased the number of preserved nerve fibers. CB1 receptors on ADMSCs significantly contribute to their pain-relieving and tissue-repairing capabilities by stimulating the growth factors secretion and suppressing the release of pro-inflammatory cytokines. Peripheral CB1 receptors do not significantly influence ADMSC-induced antinociception.
Collapse
Affiliation(s)
| | - Sergey V. Pinchuk
- Institute of Biophysics and Cell EngineeringNational Academy of Sciences of BelarusMinskBelarus
| | | | - Alla Y. Molchanova
- Institute of PhysiologyNational Academy of Sciences of BelarusMinskBelarus
| |
Collapse
|
6
|
Zhang SB, Zhao GH, Lv TR, Gong CY, Shi YQ, Nan W, Zhang HH. Bibliometric and visual analysis of microglia-related neuropathic pain from 2000 to 2021. Front Mol Neurosci 2023; 16:1142852. [PMID: 37273906 PMCID: PMC10233022 DOI: 10.3389/fnmol.2023.1142852] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/28/2023] [Indexed: 06/06/2023] Open
Abstract
Background Microglia has gradually gained researchers' attention in the past few decades and has shown its promising prospect in treating neuropathic pain. Our study was performed to comprehensively evaluate microglia-related neuropathic pain via a bibliometric approach. Methods We retrospectively reviewed publications focusing on microglia-related neuropathic pain from 2000 to 2021 in WoSCC. VOS viewer software and CiteSpace software were used for statistical analyses. Results A total of 2,609 articles were finally included. A steady increase in the number of relevant publications was observed in the past two decades. China is the most productive country, while the United States shares the most-cited and highest H-index country. The University of London, Kyushu University, and the University of California are the top 3 institutions with the highest number of publications. Molecular pain and Pain are the most productive and co-cited journals, respectively. Inoue K (Kyushu University) is the most-contributed researcher and Ji RR (Duke University) ranks 1st in both average citations per article and H-index. Keywords analyses revealed that pro-inflammatory cytokines shared the highest burst strength. Sex differences, neuroinflammation, and oxidative stress are the emerging keywords in recent years. Conclusion In the field of microglia-related neuropathic pain, China is the largest producer and the United States is the most influential country. The signaling communication between microglia and neurons has continued to be vital in this field. Sexual dimorphism, neuroinflammation, and stem-cell therapies might be emerging trends that should be closely monitored.
Collapse
Affiliation(s)
- Shun-Bai Zhang
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Guang-Hai Zhao
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Tian-Run Lv
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Chao-Yang Gong
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Yong-Qiang Shi
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Wei Nan
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, Lanzhou, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, China
| |
Collapse
|
7
|
Yu JI, Cho YH, Seo TB, Kim YP. Effect of combined intervention of exercise and autologous bone marrow stromal cell transplantation on neurotrophic factors and pain-related cascades over time after sciatic nerve injury. J Exerc Rehabil 2023; 19:19-26. [PMID: 36910683 PMCID: PMC9993005 DOI: 10.12965/jer.2244006.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/11/2023] [Indexed: 02/25/2023] Open
Abstract
The purpose of this study was to determine whether combined inter-vention of treadmill exercise and bone marrow stromal cell (BMSC) transplantation would affect the expression of neurotrophic factors in the sciatic nerve injury (SNI) and neuropathic pain-related cascades in ipsilateral lumbar 4-5 dorsal root ganglion (DRG) during the early or late stage of sciatic nerve regeneration. The rats were randomly divided into the normal control group (CONT, n=6), sedentary group (SS, n=24), exercise group (SE, n=24), BMSC transplantation group (SB, n=24), BMSC transplantation+exercise group (SBE, n=24) 1, 2, 3, and 5 weeks after SNI. Single dose of 5×106 harvested BMSC was injected into the injury area sing by a 30 gauge needle. Treadmill exercise was performed at a speed of 8 m/min for 30 min once a day. Tropomyosin-receptor kinase B, brain-derived neurotrophic factor and ciliary neurotrophic fac-tor were significantly upregulated in the SE and SBE groups at 1- and 2-week postinjury than those in the CONT and SS groups, and SB and SBE groups continuously kept up proinflammatory cytokines until the late stage of regeneration. Nuclear factor kappa-light-chain-enhancer of activated B cells, interleukin and tumor necrosis factor alpha in ipsi-lateral DRG were progressively decreased by exercise alone application and/or BMSC transplantation at early and late stage of regeneration. Present results provide reliable information that combined intervention of treadmill exercise and BMSC transplantation might be one of the effective treatment strategies for recovering sciatic nerve injury-induced neuropathic pain over time.
Collapse
Affiliation(s)
- Joo-In Yu
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| | - Yeong-Hyun Cho
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| | - Tae-Beom Seo
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| | - Young-Pyo Kim
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| |
Collapse
|
8
|
Yin Q, Zou T, Sun S, Yang D. Cell therapy for neuropathic pain. Front Mol Neurosci 2023; 16:1119223. [PMID: 36923653 PMCID: PMC10008860 DOI: 10.3389/fnmol.2023.1119223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/07/2023] [Indexed: 03/02/2023] Open
Abstract
Neuropathic pain (NP) is caused by a lesion or a condition that affects the somatosensory system. Pathophysiologically, NP can be ascribed to peripheral and central sensitization, implicating a wide range of molecular pathways. Current pharmacological and non-pharmacological approaches are not very efficacious, with over half of NP patients failing to attain adequate pain relief. So far, pharmacological and surgical treatments have focused primarily on symptomatic relief by modulating pain transduction and transmission, without treating the underlying pathophysiology. Currently, researchers are trying to use cell therapy as a therapeutic alternative for the treatment of NP. In fact, mounting pre-clinical and clinical studies showed that the cell transplantation-based therapy for NP yielded some encouraging results. In this review, we summarized the use of cell grafts for the treatment of NP caused by nerve injury, synthesized the latest advances and adverse effects, discussed the possible mechanisms to inform pain physicians and neurologists who are endeavoring to develop cell transplant-based therapies for NP and put them into clinical practice.
Collapse
Affiliation(s)
- QingHua Yin
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - TianHao Zou
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - ShuJun Sun
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Yang
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Wang H, Wang F, Wang Y, Li X, Di C, Liang C, Mu Y, Zhou J. Study on the Mechanism of BMSCs in Regulating NF-κB Signal Pathway by Targeting miR-449a to Improve the Inflammatory Response to Peripheral Nerve Injury. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2022; 22:546-561. [PMID: 36458392 PMCID: PMC9716300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To evaluate the mechanism of Bone Marrow Mesenchymal Stem Cells (BMSCs) in regulating NF-κB signal pathway by targeting miR-449a. METHODS Stem cells were transfected by over-expressing and inhibiting miR-449a to detect the levels and viability of miR-449a in stem cells after transfection. Stem cells and neurons were co-cultured in vitro to evaluate the in vitro mechanism of stem cells over-expressing miR-449a on neurons. RESULTS After the addition of neurons, the neuronal activity of miR-449a over-expression group increased significantly, the expression of NF-κB signal pathway proteins (IκBα, p50, and p65) decreased, and the inflammatory cytokines (TNF-α and IL-1β) decreased significantly (P<0.05). In vivo experiments in rats also showed that rats were unresponsive, did not chirp or elude after being stimulated. After stem cell therapy, the weight and response of rats gradually returned to normal levels. miR-449a expression significantly increased in the stem cell + miR-449a over-expression group, expression of NF-κB signal pathway proteins (IκBα, p50, and p65) decreased, inflammatory cytokines (TNF-α and IL-1β) significantly decreased, and cell activity significantly increased (P<0.05). CONCLUSIONS BMSCs can modulate NF-κB signaling pathway by targeting miR-449a, so as to reduce the inflammatory response to peripheral nerve injury and repair nerve injury.
Collapse
Affiliation(s)
- Hongjiao Wang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| | - Fangyuan Wang
- Department of General Surgery, Qiqihar First Hospital, China
| | - Yuejing Wang
- Department of Histology and Embryology, Qiqihar Medical College, China
| | - Xiaonan Li
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| | - Cihan Di
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| | - Chunming Liang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| | - Yuyuan Mu
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| | - Jiexin Zhou
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, China
| |
Collapse
|
10
|
Kan H, Fan L, Gui X, Li X, Yang S, Huang Y, Chen L, Shen W. Stem Cell Therapy for Neuropathic Pain: A Bibliometric and Visual Analysis. J Pain Res 2022; 15:1797-1811. [PMID: 35769691 PMCID: PMC9236174 DOI: 10.2147/jpr.s365524] [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: 03/22/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Neuropathic pain is intractable and current treatment modalities are ineffective to cure this intractable pain, which has become a global problem. In recent years, there have been an increasing number of studies on stem cell therapy for neuropathic pain that have shown enormous potential. Using a visual analysis approach of the existing literature on stem cell therapy for neuropathic pain, we hope to understand the current research status and hot issues in this field and to provide valuable predictions for future research in this field. Methods We used Citespace software to visually analyze 291 articles and reviews indexed by the Web of Science Core Collection Database exploring stem cell-based treatment of neuropathic pain from 1995 to 2021. The Gunnmap online world map evaluated the number of countries and regional articles separately. Microsoft Excel 2016 was used to generate a graph of trends in annual publications. Results Visualization analysis revealed that the number of publications has increased yearly. The top three countries in terms of number of articles published are United States, China, and Japan. Analysis of highly co-cited articles revealed that the contents of these articles primarily involved the expression of IL-1β, IL-10, NPY, TRPA1, p-p38, p-ERK1/2, TGF-β, PKCδ, CaMKIIɑ, P2X4, P2X7 and TNF-ɑ. Keywords and citation burst analysis demonstrated that activation, regeneration, chemotherapy, and expression are likely the research hotspots and future directions of stem cell research in neuropathic pain. Conclusion Stem cell therapy may be a potential means of future treatment of neuropathic pain. The study of the mechanisms underlying stem cell therapy for neuropathic pain is still a focus of future research.
Collapse
Affiliation(s)
- Houming Kan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu, People's Republic of China
| | - Lijun Fan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu, People's Republic of China
| | - Xiaodie Gui
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Xiaoqiang Li
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Sen Yang
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yuting Huang
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Liping Chen
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Wen Shen
- Department of Pain, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| |
Collapse
|
11
|
Zheng D, Bhuvan T, Payne NL, Heng TSP. Secondary Lymphoid Organs in Mesenchymal Stromal Cell Therapy: More Than Just a Filter. Front Immunol 2022; 13:892443. [PMID: 35784291 PMCID: PMC9243307 DOI: 10.3389/fimmu.2022.892443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic potential in inflammatory models of human disease. However, clinical translation has fallen short of expectations, with many trials failing to meet primary endpoints. Failure to fully understand their mechanisms of action is a key factor contributing to the lack of successful commercialisation. Indeed, it remains unclear how the long-ranging immunomodulatory effects of MSCs can be attributed to their secretome, when MSCs undergo apoptosis in the lung shortly after intravenous infusion. Their apoptotic fate suggests that efficacy is not based solely on their viable properties, but also on the immune response to dying MSCs. The secondary lymphoid organs (SLOs) orchestrate immune responses and play a key role in immune regulation. In this review, we will discuss how apoptotic cells can modify immune responses and highlight the importance of MSC-immune cell interactions in SLOs for therapeutic outcomes.
Collapse
Affiliation(s)
- Di Zheng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tejasvini Bhuvan
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Natalie L. Payne
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Tracy S. P. Heng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, Australia
- *Correspondence: Tracy S. P. Heng,
| |
Collapse
|
12
|
Li X, Guan Y, Li C, Zhang T, Meng F, Zhang J, Li J, Chen S, Wang Q, Wang Y, Peng J, Tang J. Immunomodulatory effects of mesenchymal stem cells in peripheral nerve injury. Stem Cell Res Ther 2022; 13:18. [PMID: 35033187 PMCID: PMC8760713 DOI: 10.1186/s13287-021-02690-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/18/2021] [Indexed: 12/14/2022] Open
Abstract
Various immune cells and cytokines are present in the aftermath of peripheral nerve injuries (PNI), and coordination of the local inflammatory response is of great significance for the recovery of PNI. Mesenchymal stem cells (MSCs) exhibit immunosuppressive and anti-inflammatory abilities which can accelerate tissue regeneration and attenuate inflammation, but the role of MSCs in the regulation of the local inflammatory microenvironment after PNI has not been widely studied. Here, we summarize the known interactions between MSCs, immune cells, and inflammatory cytokines following PNI with a focus on the immunosuppressive role of MSCs. We also discuss the immunomodulatory potential of MSC-derived extracellular vesicles as a new cell-free treatment for PNI.
Collapse
Affiliation(s)
- Xiangling Li
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,The School of Medicine, Jinzhou Medical University, Jinzhou, 121099, People's Republic of China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Fanqi Meng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,Department of Spine Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Jian Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Junyang Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.,The School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China
| | - Qi Wang
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.,The School of Medicine, Jinzhou Medical University, Jinzhou, 121099, People's Republic of China
| | - Yi Wang
- Department of Stomatology, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People's Republic of China.
| | - Jinshu Tang
- The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| |
Collapse
|
13
|
Asgharzade S, Talaei A, Farkhondeh T, Forouzanfar F. A Review on Stem Cell Therapy for Neuropathic Pain. Curr Stem Cell Res Ther 2021; 15:349-361. [PMID: 32056531 DOI: 10.2174/1574888x15666200214112908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/16/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
Neuropathic pain is a complex, chronic pain state that is heterogeneous in nature and caused by the consequence of a lesion or disease affecting the somatosensory system. Current medications give a long-lasting pain relief only in a limited percentage of patients also associated with numerous side effects. Stem cell transplantation is one of the attractive therapeutic platforms for the treatment of a variety of diseases, such as neuropathic pain. Here, the authors review the therapeutic effects of stem cell transplantation of different origin and species in different models of neuropathic pain disorders. Stem cell transplantation could alleviate the neuropathic pain; indeed, stem cells are the source of cells, which differentiate into a variety of cell types and lead trophic factors to migrate to the lesion site opposing the effects of damage. In conclusion, this review suggests that stem cell therapy can be a novel approach for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Andisheh Talaei
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Wang Q, He H, Xie S, Wei Q, He C. Mesenchymal Stem Cells Transplantation for Neuropathic Pain Induced By Peripheral Nerve Injury in Animal Models: A Systematic Review. Stem Cells Dev 2020; 29:1420-1428. [PMID: 32962522 DOI: 10.1089/scd.2020.0131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain is defined as a lesion or disease of the somatosensory system, currently remaining a challenging condition to treat. Mesenchymal stem cells (MSCs) transplantation is emerging as a promising strategy to alleviate the neuropathic pain conditions induced by peripheral nerve injury. The aim of this systematic review was to assess the efficacy and safety of MSCs transplantation in neuropathic pain induced by peripheral nerve injury in controlled animal studies, and thus to yield evidence-based decision making. Following the PRISMA guidelines, PubMed, Cochrane Central Library, Embase, and CINAHL were searched for preclinical controlled animal studies from the inception to April 16, 2020. Seventeen studies are included in this review. Substantial heterogeneity is observed regarding the animal's species, models of neuropathic pain, regimen of MSCs transplantation, and outcome of measures across the included studies. Both mechanical allodynia and thermal hyperalgesia could be significantly attenuated by transplanted MSCs. The MSCs-elicited analgesic effect is independent of the type of MSCs, time of administration, and route of delivery, and is efficiently enhanced by genetic transfection with fibroblast growth factor, proenkephalin, and glial cell line-derived neurotrophic factor. The migration of MSCs after intrathecal or intravenous injection has been shown to be directed toward the surface of dorsal spinal cord or dorsal root ganglions on the ipsilateral side of injury. No adverse effects have been reported. The accumulating evidence demonstrates the therapeutic effect of MSCs-based cell therapy on prevention and alleviation of the neuropathic pain induced by peripheral nerve injury in rat or mouse models. The robust preclinical studies are deserved to optimize the regimen of MSCs transplantation and to promote the translation of the MSCs-based therapy into clinical studies.
Collapse
Affiliation(s)
- Qian Wang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Hongchen He
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Shuhang Xie
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Quan Wei
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Chengqi He
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Xu C, Diao YF, Wang J, Liang J, Xu HH, Zhao ML, Zheng B, Luan Z, Wang JJ, Yang XP, Wei MG, Duan JH, Wang KQ, Chen C, Chen F, Ming D, Zhang S, Sun HT, Li XH. Intravenously Infusing the Secretome of Adipose-Derived Mesenchymal Stem Cells Ameliorates Neuroinflammation and Neurological Functioning After Traumatic Brain Injury. Stem Cells Dev 2020; 29:222-234. [PMID: 31830866 DOI: 10.1089/scd.2019.0173] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Chao Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Yun-Feng Diao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
- Department of Neurosurgery, Shanxi Dayi Hospital, Taiyuan, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Ming-Liang Zhao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zuo Luan
- Department of Pediatrics, Pediatric Surgery of Navy General Hospital, Beijing, China
| | - Jing-Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xi-Ping Yang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Meng-Guang Wei
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing-Hao Duan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Ke-Qiang Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Feng Chen
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Sai Zhang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Hong-Tao Sun
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| |
Collapse
|
16
|
Yoo SH, Lee SH, Lee S, Park JH, Lee S, Jin H, Park HJ. The effect of human mesenchymal stem cell injection on pain behavior in chronic post-ischemia pain mice. Korean J Pain 2020; 33:23-29. [PMID: 31888314 PMCID: PMC6944374 DOI: 10.3344/kjp.2020.33.1.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/30/2022] Open
Abstract
Background Neuropathic pain (NP) is considered a clinically incurable condition despite various treatment options due to its diverse causes and complicated disease mechanisms. Since the early 2000s, multipotent human mesenchymal stem cells (hMSCs) have been used in the treatment of NP in animal models. However, the effects of hMSC injections have not been studied in chronic post-ischemia pain (CPIP) mice models. Here, we investigated whether intrathecal (IT) and intrapaw (IP) injections of hMSCs can reduce mechanical allodynia in CPIP model mice. Methods Seventeen CPIP C57/BL6 mice were selected and randomized into four groups: IT sham (n = 4), IT stem (n = 5), IP sham (n = 4), and IP stem (n = 4). Mice in the IT sham and IT stem groups received an injection of 5 μL saline and 2 × 104 hMSCs, respectively, while mice in the IP sham and IP stem groups received an injection of 5 μL saline and 2 × 105 hMSCs, respectively. Mechanical allodynia was assessed using von Frey filaments from pre-injection to 30 days post-injection. Glial fibrillary acidic protein (GFAP) expression in the spinal cord and dorsal root ganglia were also evaluated. Results IT and IP injections of hMSCs improved mechanical allodynia. GFAP expression was decreased on day 25 post-injection compared with the sham group. Injections of hMSCs improved allodynia and GFAP expression was decreased compared with the sham group. Conclusions These results suggested that hMSCs may be also another treatment modality in NP model by ischemia-reperfusion.
Collapse
Affiliation(s)
- Sie Hyeon Yoo
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Hyun Lee
- Department of Anesthesiology and Pain Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seunghwan Lee
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Hong Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seunghyeon Lee
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Heecheol Jin
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Hue Jung Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
17
|
Teng Y, Zhang Y, Yue S, Chen H, Qu Y, Wei H, Jia X. Intrathecal injection of bone marrow stromal cells attenuates neuropathic pain via inhibition of P2X 4R in spinal cord microglia. J Neuroinflammation 2019; 16:271. [PMID: 31847848 PMCID: PMC6918679 DOI: 10.1186/s12974-019-1631-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/05/2019] [Indexed: 01/23/2023] Open
Abstract
Background Neuropathic pain is one of the most debilitating of all chronic pain syndromes. Intrathecal (i.t.) bone marrow stromal cell (BMSC) injections have a favorable safety profile; however, results have been inconsistent, and complete understanding of how BMSCs affect neuropathic pain remains elusive. Methods We evaluated the analgesic effect of BMSCs on neuropathic pain in a chronic compression of the dorsal root ganglion (CCD) model. We analyzed the effect of BMSCs on microglia reactivity and expression of purinergic receptor P2X4 (P2X4R). Furthermore, we assessed the effect of BMSCs on the expression of transient receptor potential vanilloid 4 (TRPV4), a key molecule in the pathogenesis of neuropathic pain, in dorsal root ganglion (DRG) neurons. Results I.t. BMSC transiently but significantly ameliorated neuropathic pain behavior (37.6% reduction for 2 days). We found no evidence of BMSC infiltration into the spinal cord parenchyma or DRGs, and we also demonstrated that intrathecal injection of BMSC-lysates provides similar relief. These findings suggest that the analgesic effects of i.t. BMSC were largely due to the release of BMSC-derived factors into the intrathecal space. Mechanistically, we found that while i.t. BMSCs did not change TRPV4 expression in DRG neurons, there was a significant reduction of P2X4R expression in the spinal cord microglia. BMSC-lysate also reduced P2X4R expression in activated microglia in vitro. Coadministration of additional pharmacological interventions targeting P2X4R confirmed that modulation of P2X4R might be a key mechanism for the analgesic effects of i.t. BMSC. Conclusion Altogether, our results suggest that i.t. BMSC is an effective and safe treatment of neuropathic pain and provides novel evidence that BMSC’s analgesic effects are largely mediated by the release of BMSC-derived factors resulting in microglial P2X4R downregulation.
Collapse
Affiliation(s)
- Yongbo Teng
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan, China
| | - Yang Zhang
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan, China.,Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Shouwei Yue
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan, China.
| | - Huanwen Chen
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yujuan Qu
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan, China
| | - Hui Wei
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan, China
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Ren J, Liu N, Sun N, Zhang K, Yu L. Mesenchymal Stem Cells and their Exosomes: Promising Therapeutics for Chronic Pain. Curr Stem Cell Res Ther 2019; 14:644-653. [PMID: 31512998 DOI: 10.2174/1574888x14666190912162504] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/27/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
Abstract
Chronic pain is a common condition that seriously affects the quality of human life with
variable etiology and complicated symptoms; people who suffer from chronic pain may experience
anxiety, depression, insomnia, and other harmful emotions. Currently, chronic pain treatments are nonsteroidal
anti-inflammatory drugs and opioids; these drugs are demonstrated to be insufficient and
cause severe side effects. Therefore, research into new therapeutic strategies for chronic pain is a top
priority. In recent years, stem cell transplantation has been demonstrated to be a potent alternative for
the treatment of chronic pain. Mesenchymal stem cells (MSCs), a type of pluripotent stem cell, exhibit
multi-directional differentiation, promotion of stem cell implantation, and immune regulation; they
have also been shown to exert analgesic effects in several chronic pain models. Exosomes produced by
MSCs have been demonstrated to relieve painful symptoms with fewer side effects. In this review, we
summarize the therapeutic use of MSCs in various chronic pain studies. We also discuss ways to enhance
the treatment effect of MSCs. We predict in the future, cell-free therapies for chronic pain will
develop from exosomes secreted by MSCs.
Collapse
Affiliation(s)
- Jinxuan Ren
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Kehan Zhang
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lina Yu
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Wu S, Ni S, Jiang X, Kuss MA, Wang HJ, Duan B. Guiding Mesenchymal Stem Cells into Myelinating Schwann Cell-Like Phenotypes by Using Electrospun Core-Sheath Nanoyarns. ACS Biomater Sci Eng 2019; 5:5284-5294. [PMID: 33455233 DOI: 10.1021/acsbiomaterials.9b00748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nerve guidance conduit (NGC)-infilling substrates have been reported to facilitate the regeneration of injured peripheral nerves (PNs), especially for large nerve gaps. In this study, longitudinally oriented electrospun core-sheath nanoyarns (csNYs), consisting of a polylactic acid microfiber core and an electrospun nanofiber sheath, were fabricated for potential PN tissue engineering applications. Our novel csNY displayed a well-aligned nanofibrous surface topography, resembling the ultrastructure of axons and fascicles of a native PN system, and it also provided a mechanically stable structure. The biological results showed that the csNY significantly enhanced the attachment, growth, and proliferation of human adipose derived mesenchymal stem cells (hADMSC) and also promoted the migration, proliferation, and phenotype maintenance of rabbit Schwann cells (rSCs). Our csNY notably increased the differentiation capability of hADMSC into SC-like cells (hADMSC-SC), in comparison with a 2D tissue culture polystyrene plate. More importantly, when combined with the appropriate induction medium, our csNY promoted hADMSC-SC to express high levels of myelination-associated markers. Overall, this study demonstrates that our csNYs have great potential to serve as not only ideal in vitro culture models for understanding SC-axon interaction and SC myelination but also as promising NGC-infilling substrates for PN regeneration applications.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles & Clothing; Collaborative Innovation Center of Marine Biomass Fibers, Qingdao University, Qingdao 266071, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | | | | | | | - Bin Duan
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
20
|
Pretreatment with AM1241 Enhances the Analgesic Effect of Intrathecally Administrated Mesenchymal Stem Cells. Stem Cells Int 2019; 2019:7025473. [PMID: 31611918 PMCID: PMC6755285 DOI: 10.1155/2019/7025473] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells have cannabinoid (CB) receptors type 1 and type 2 and can alleviate a variety of neuropathic pains, including chronic constriction injury (CCI). A selective CB2 receptor agonist is AM1241. In the present study, it was found that mice with CCI displayed a longer duration of mechanical and thermal analgesia when intrathecally (i.t.) injected with AM1241-treated mesenchymal stem cells, compared to those injected with untreated mesenchymal stem cells or AM1241 alone. Moreover, CCI-induced upregulation of the phosphorylated extracellular signal-regulated kinase (ERK) 1/2 (p-ERK1/2) was inhibited following i.t. injection of AM1241-treated mesenchymal stem cells and this inhibition was noticeably higher compared to injection with untreated mesenchymal stem cells. The expression of transforming growth factor-β1 (TGF-β1) was also analyzed in the dorsal root ganglion (DRGs) and spinal cord of CCI mice. In untreated CCI mice, expression of TGF-β1 was increased, whereas pretreatment with AM1241-treated mesenchymal stem cells regulated the expression of TGF-β1 on 10 days and 19 days after surgery. In addition, i.t. injection of exogenous TGF-β1 slightly alleviated neuropathic pain whilst neutralization of TGF-β1 potently blocked the effect of AM1241-treated mesenchymal stem cells on thermal hyperalgesia and mechanical allodynia of CCI mice. In an in vitro experiment, AM1241 could enhance the release of TGF-β1 in the supernatant of BMSCs after lipopolysaccharide (LPS) simulation. Taken together, the findings of the current study show that i.t. administration of AM1241-treated mesenchymal stem cells has a positive effect on analgesia and that TGF-β1 and p-ERK1/2 may be the molecular signaling pathway involved in this process.
Collapse
|
21
|
Price TJ, Gold MS. From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain. PAIN MEDICINE 2019; 19:1525-1549. [PMID: 29077871 DOI: 10.1093/pm/pnx108] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective Persistent pain causes untold misery worldwide and is a leading cause of disability. Despite its astonishing prevalence, pain is undertreated, at least in part because existing therapeutics are ineffective or cause intolerable side effects. In this review, we cover new findings about the neurobiology of pain and argue that all but the most transient forms of pain needed to avoid tissue damage should be approached as a disease where a cure can be the goal of all treatment plans, even if attaining this goal is not yet always possible. Design We reviewed the literature to highlight recent advances in the area of the neurobiology of pain. Results We discuss barriers that are currently hindering the achievement of this goal, as well as the development of new therapeutic strategies. We also discuss innovations in the field that are creating new opportunities to treat and even reverse persistent pain, some of which are in late-phase clinical trials. Conclusion We conclude that the confluence of new basic science discoveries and development of new technologies are creating a path toward pain therapeutics that should offer significant hope of a cure for patients and practitioners alike. Classification of Evidence. Our review points to new areas of inquiry for the pain field to advance the goal of developing new therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, Bedi S, Toledano-Furman NE, Triolo F, Kamhieh-Milz J, Moll G, Cox CS. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front Immunol 2019; 10:1645. [PMID: 31417542 PMCID: PMC6685059 DOI: 10.3389/fimmu.2019.01645] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.
Collapse
Affiliation(s)
- Henry Caplan
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mitchell George
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pamela Wenzel
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Supinder Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Naama E. Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
23
|
Abstract
Nerve injury-induced neuropathic pain is difficult to treat. In this study, we used exosomes derived from human umbilical cord mesenchymal stem cell (UCMSC) as a cell-free therapy for nerve injury-induced pain in rats. Isolated UCMSC exosomes range in size from 30 to 160 nm and contain CD63, HSP60, and CD81 exosome markers. After L5/6 spinal nerve ligation surgery, single intrathecal injection of exosomes reversed nerve ligation-induced mechanical and thermal hypersensitivities of right hindpaw of rats at initial and well-developed pain stages. Moreover, continuous intrathecal infusion of exosomes achieved excellent preventive and reversal effects for nerve ligation-induced pain. In immunofluorescent study, lots of Exo-green-labelled exosomes could be found majorly in the ipsilateral L5 spinal dorsal horn, dorsal root ganglion, and peripheral axons, suggesting the homing ability of UCMSC exosomes. They also appeared in the central terminals or cell bodies of IB4, CGRP, and NF200 sensory neurons. In addition, exosome treatment suppressed nerve ligation-induced upregulation of c-Fos, CNPase, GFAP, and Iba1. All these data suggest that the analgesic effects of exosomes may involve their actions on neuron and glial cells. Exosomes also inhibited the level of TNF-α and IL-1β, while enhanced the level of IL-10, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in the ipsilateral L5/6 dorsal root ganglion of nerve-ligated rats, indicating anti-inflammatory and proneurotrophic abilities. Protein analysis revealed the content of vascular endothelial growth factor C, angiopoietin-2, and fibroblast growth factor-2 in the exosomes. In summary, intrathecal infusion of exosomes from UCMSCs may be considered as a novel therapeutic approach for nerve injury-induced pain.
Collapse
|
24
|
Altun A, Çokluk C, Yarar E, Kuruoğlu E, Aydın K, Genç E, Gunes S, Yıldız L, Marangoz AH. Histopathological evaluation of mesenchymal stem cells in the healing of anastomosed carotid arteries. AMERICAN JOURNAL OF STEM CELLS 2019; 8:19-27. [PMID: 31139494 PMCID: PMC6526360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/20/2019] [Indexed: 06/09/2023]
Abstract
The objective of this study was to evaluate the influence of mesenchymal stem cells on the healing of experimental carotid artery anastomoses histopathologically. Twenty-four female Sprague-Dawley rats were used in this study. After random separation of the subjects into two groups, in both groups carotid arteries were transected and anastomosed in end-to-end fashion. Anastomoses were locally treated with 1 ml 0.09% NaCl, and 1 ml mesenchymal stem cell suspension (1×106 cells) in control and trial groups, respectively. Anastomoses were wrapped with an 8 mm sheet of surgicel and soaked with BioGlue in order to sequestrate the stem cells. After patencies were confirmed via Doppler USG, surgical site was closed with 2/0 silk sutures. Histopathological evaluation was carried out after 4 weeks. In respect to endothelial continuity, vessel patency (along with presence or absence of restenosis), integrities of internal and external elastic laminae, muscularis and adventitia; no statistically significant differences were present between the trial and control groups. In Trial and Control Groups, luminal thrombus was present in 8 (66.6%) and 3 (25%) of the 12 subjects, respectively. The difference was statistically significant (P < 0.05). Recanalization was present in 6 subjects in trial group; 1 subjects in Control Group, respectively. Our results suggest that local administration of mesenchyme stem cell does not have a positive influence on success of an anastomosis.
Collapse
Affiliation(s)
- Adnan Altun
- Neurosurgery, Medicana Private HospitalSamsun, Turkey
| | - Cengiz Çokluk
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Ercan Yarar
- Department of Neurosurgery, Bafra State HospitalSinop, Turkey
| | - Enis Kuruoğlu
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Keramettin Aydın
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Eyüp Genç
- Department of Neurosurgery, Turhal State HospitalTokat, Turkey
| | - Sezgin Gunes
- Department of Medical Biology, Ondokuz Mayis UniversitySamsun, Turkey
| | - Levent Yıldız
- Department of Pathology, Ondokuz Mayis UniversitySamsun, Turkey
| | | |
Collapse
|
25
|
Review of the Current Knowledge on the Role of Stem Cell Transplantation in Neurorehabilitation. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3290894. [PMID: 30931325 PMCID: PMC6413404 DOI: 10.1155/2019/3290894] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/05/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
The management involving stem cell (SC) therapy along with physiotherapy offers tremendous chance for patients after spinal cord injury (SCI), traumatic brain injury (TBI), stroke, etc. However, there are still only a limited number of reports assessing the impact of stem cells (SCs) on the rehabilitation process and/or the results of the simultaneous use of SC and rehabilitation. Additionally, since there is still not enough convincing evidence about the effect of SCT on humans, e.g., in stroke, there have been no studies conducted concerning rehabilitation program formation and expected outcomes. It has been shown that bone marrow-derived mesenchymal stem cell (BMSCs) transplantation in rats combined with hyperbaric oxygen therapy (HBO) can promote the functional recovery of hind limbs after SCI. An anti-inflammatory effect has been shown. One case study showed that, after the simultaneous use of SCT and rehabilitation, an SCI patient progressed from ASIA Grade A to ASIA Grade C. Such promising data in the case of complete tetraplegia could be a breakthrough in the treatment of neurologic disorders in humans. Although SCT appears as a promising method for the treatment of neurological conditions, e.g., complete tetraplegia, much work should be done towards the development of rehabilitation protocols.
Collapse
|
26
|
Yilmaz A, Topcu A, Erdogan C, Sahin B, Abban G, Coskun E, Ozkul A. The Effect of Vascular Graft and Human Umbilical Cord Blood-Derived CD34+ Stem Cell on Peripheral Nerve Healing. Open Access Maced J Med Sci 2018; 6:1946-1952. [PMID: 30559841 PMCID: PMC6290437 DOI: 10.3889/oamjms.2018.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 09/10/2018] [Accepted: 10/13/2018] [Indexed: 11/08/2022] Open
Abstract
AIM: There are many trials concerning peripheral nerve damage causes and treatment options. Unfortunately, nerve damage is still a major problem regarding health, social and economic issues. On this study, we used vascular graft and human cord blood derived stem cells to find an alternative treatment solution to this problem. MATERIAL AND METHODS: We used 21 female Wistar rats on our study. They were anesthetized with ketamine and we studied right hind limbs. On Group 1, we did a full layer cut on the right sciatic nerve. On Group 2, we did a full layer cut on the right sciatic nerve, and we covered synthetic vascular graft on cut area. On Group 3, we did a full layer cut on right sciatic nerve, and we covered the area with stem cell applied vascular graft. RESULTS: At the end of postoperative 8. weeks, we performed EMG on the rats. When we compared healthy and degenerated areas as a result of EMG, we found significant amplitude differences between the groups on healthy areas whereas there was no significant difference on degenerated areas between the groups. Then we re-opened the operated area again to reveal the sciatic nerve cut area, and we performed electron microscope evaluation. On the stem cell group, we observed that both the axon and the myelin sheet prevented degeneration. CONCLUSION: This study is a first on using synthetic vascular graft and cord blood derived CD34+ cells in peripheral nerve degeneration. On the tissues that were examined with electron microscope, we observed that CD34+ cells prevented both axonal and myelin sheath degeneration. Nerve tissue showed similar results to the control group, and the damage was minimal.
Collapse
Affiliation(s)
- Ali Yilmaz
- Adnan Menderes University, Neurosurgery Department, Aydın, Turkey
| | - Abdullah Topcu
- Adnan Menderes University, Neurosurgery Department, Aydın, Turkey
| | - Cagdas Erdogan
- Pamukkale University, Neurology Department, Denizli, Turkey
| | - Barbaros Sahin
- University Medicine Faculty, Animal Laboratory, Denizli, Turkey
| | - Gulcin Abban
- Pamukkale University Medicine Faculty, Histology and Embryology Department, Denizli, Turkey
| | - Erdal Coskun
- Pamukkale University, Medicine Faculty, Neurosurgery Department, Denizli, Turkey
| | - Ayca Ozkul
- Adnan Menderes University, Neurosurgery Department, Aydın, Turkey
| |
Collapse
|
27
|
Abdelrahman SA, Samak MA, Shalaby SM. Fluoxetine pretreatment enhances neurogenic, angiogenic and immunomodulatory effects of MSCs on experimentally induced diabetic neuropathy. Cell Tissue Res 2018; 374:83-97. [PMID: 29687216 DOI: 10.1007/s00441-018-2838-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/22/2018] [Indexed: 02/07/2023]
Abstract
Being one of the most debilitating complications among diabetic patients, diabetic polyneuropathy (DPN) is a paramount point of continuous research. Stem cell therapies have shown promising results. However, limited cell survival and paracrine activities hinder its transfer from bench to bedside. We designed this study to evaluate fluoxetine-pretreatment technique of mesenchymal stem cells (MSCs) as an approach to enhance their paracrine and immunomodulatory properties in DPN. Effects of fluoxetine treatment of MSCs were tested in vitro. Forty-two adult Wistar male albino rats were utilized, further subdivided into control, diabetic, MSC-treated and fluoxetine-pretreated MSC groups. Sciatic nerve sections were prepared for light and electron microscope examination and immunohistochemical detection of neurofilament (NF) protein. Also, we assessed in vitro survival and paracrine properties of fluoxetine-pretreated MSCs. Real time PCR of BDNF, VEGF, IL-1β, and IL-10 expression in tissue homogenate was performed. Our results showed restoration of normal neuronal histomorphology and ultrastructure, moreover, immunohistochemical expression of anti-neurofilament protein was significantly elevated in MSC-treated groups compared to the diabetic one. Fluoxetine enhanced the MSC survival and their paracrine properties of MSCs in vitro. Furthermore, the fluoxetine-pretreated MSC group revealed a significant elevation of mRNA expression of BDNF (neurotrophic factor) and VEGF (angiogenic factor), denoting ameliorated MSC paracrine properties. Similarly, improved immunomodulatory functions were evident by a significant reduction of interleukin-1β mRNA expression (pro-inflammatory) and a reciprocal significant increase of interleukin-10 (anti-inflammatory). We concluded that fluoxetine-pretreatment of MSCs boosts their survival, paracrine, and immunomodulatory traits and directly influenced neuronal histomorphology. Hence, it presents a promising intervention of diabetic polyneuropathy. Graphical Abstract.
Collapse
Affiliation(s)
- Shaimaa A Abdelrahman
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Asharquia, Zagazig, 44519, Egypt
| | - Mai A Samak
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Asharquia, Zagazig, 44519, Egypt.
| | - Sally M Shalaby
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
28
|
Sun Y, Zhang D, Li H, Long R, Sun Q. Intrathecal administration of human bone marrow mesenchymal stem cells genetically modified with human proenkephalin gene decrease nociceptive pain in neuropathic rats. Mol Pain 2018; 13:1744806917701445. [PMID: 28326940 PMCID: PMC5391071 DOI: 10.1177/1744806917701445] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background Mesenchymal stem cell (MSC) has been one of the potential tools in neuropathic pain therapy; however, the augmented efficacy may be expected when they are modified with human proenkephalin (hPPE) gene. In the current study, the antinociceptive effect of human bone marrow stem cells (hBMSCs) engineered with hPPE gene (hPPE-hBMSCs) on sciatic nerve chronic constriction injury (CCI)-induced neuropathic pain in rats was investigated. Methods Primary-cultured hBMSCs were passaged and modified with hPPE, and the cell suspensions (6 × 106) were then intrathecally injected into a rat model of CCI. Paw mechanical withdrawal threshold and paw withdrawal thermal latency were measured before and after CCI surgery. The effects of hPPE gene transfer on hBMSCs bioactivity were analyzed in vitro and in vivo. Results No changes were observed in the surface phenotypes and differentiation of hBMSCs after gene transfer. The hPPE-hBMSC group showed improved paw mechanical withdrawal threshold and paw thermal withdrawal latency values on the ipsilateral side of rats with CCI from day 9 post-surgery, and the analgesic effect was reversed by naloxone. Leucine-enkephalin (L-EK) secretion was augmented in the hPPE-engineered hBMSC group. Conclusions The intrathecal administration of BMSCs modified with hPPE gene can effectively relieve pain caused by chronic constriction injury in rats and might be a potentially therapeutic tool for neuropathic pain in humans.
Collapse
|
29
|
Zou W, Xu W, Song Z, Zhong T, Weng Y, Huang C, Li M, Zhang C, Zhan X, Guo Q. Proteomic Identification of an Upregulated Isoform of Annexin A3 in the Spinal Cords of Rats in a Neuropathic Pain Model. Front Neurosci 2017; 11:484. [PMID: 28928629 PMCID: PMC5591859 DOI: 10.3389/fnins.2017.00484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/15/2017] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain (NP) is induced by nerve damage or a disturbance in the peripheral or central nervous systems. Nerve damage causes the activation of sensitizing mechanisms in the peripheral and central nervous systems, which induces transcriptional and post-transcriptional alterations in sensory nerves. However, the underlying mechanisms of NP remain elusive. In the study, Two-dimensional gel electrophoresis (2DGE)-based comparative proteomics identified 38 differential gel spots, and 15 differentially expressed proteins (DEPs) between the sham and the chronic constriction injury (CCI)-induced neuropathic pain rats. Of them, Annexin A3 (ANXA3) was significantly increased after CCI with Western blot analysis and immunofluorescence imaging. A lentivirus delivering ANXA3 shRNA (LV-shANXA3) was administered intrathecally to determine the analgesic effects of ANXA3 on allodynia and hyperalgesia in a CCI-induced neuropathic pain model in rats. Further study showed that LV-shANXA3 reversed the upregulation of ANXA3, alleviated CCI-induced mechanical allodynia and thermal hyperalgesia. The study indicated that ANXA3 may play an important role in neuropathic pain.
Collapse
Affiliation(s)
- Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Wei Xu
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Zongbin Song
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Tao Zhong
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yingqi Weng
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Changsheng Huang
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Maoyu Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South UniversityChangsha, China
| | - Chuanlei Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South UniversityChangsha, China.,Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South UniversityChangsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
30
|
Fischer G, Wang F, Xiang H, Bai X, Yu H, Hogan QH. Inhibition of neuropathic hyperalgesia by intrathecal bone marrow stromal cells is associated with alteration of multiple soluble factors in cerebrospinal fluid. Exp Brain Res 2017; 235:2627-2638. [PMID: 28573310 PMCID: PMC6688185 DOI: 10.1007/s00221-017-5000-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/24/2017] [Indexed: 01/09/2023]
Abstract
Injury-induced neuropathic pain remains a serious clinical problem. Recent studies indicate that bone marrow stromal cells (BMSCs) effectively attenuate chronic neuropathic pain in animal models. Here, we examined the therapeutic effect of intrathecal administration of BMSCs isolated from young (1-month-old) rats on pain hypersensitivity induced by tibial nerve injury. Cerebrospinal fluid (CSF) was collected and analyzed to examine the effect of BMSC administration on the expression of 67 soluble factors in CSF. A sustained remission in injury-induced mechanical hyperalgesia was observed in BMSC-treated rats but not in control animals. Engrafted BMSCs were observed in spinal cords and dorsal root ganglia at 5 weeks after cell injection. Injury significantly decreased the levels of six soluble factors in CSF: intercellular adhesion molecule 1 (ICAM-1), interleukin-1β (IL-1β), IL-10, hepatocyte growth factor (HGF), Nope protein, and neurogenic locus notch homolog protein 1 (Notch-1). Intrathecal BMSCs significantly attenuated the injury-induced reduction of ICAM-1, IL-1β, HGF, IL-10, and Nope. This study adds to evidence supporting the use of intrathecal BMSCs in pain control and shows that this effect is accompanied by the reversal of injury-induced reduction of multiple CSF soluble factors. Our findings suggest that these soluble factors may be potential targets for treating chronic pain.
Collapse
Affiliation(s)
- Gregory Fischer
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, People's Republic of China
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| |
Collapse
|
31
|
Zorzopulos J, Opal SM, Hernando-Insúa A, Rodriguez JM, Elías F, Fló J, López RA, Chasseing NA, Lux-Lantos VA, Coronel MF, Franco R, Montaner AD, Horn DL. Immunomodulatory oligonucleotide IMT504: Effects on mesenchymal stem cells as a first-in-class immunoprotective/immunoregenerative therapy. World J Stem Cells 2017; 9:45-67. [PMID: 28396715 PMCID: PMC5368622 DOI: 10.4252/wjsc.v9.i3.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/12/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023] Open
Abstract
The immune responses of humans and animals to insults (i.e., infections, traumas, tumoral transformation and radiation) are based on an intricate network of cells and chemical messengers. Abnormally high inflammation immediately after insult or abnormally prolonged pro-inflammatory stimuli bringing about chronic inflammation can lead to life-threatening or severely debilitating diseases. Mesenchymal stem cell (MSC) transplant has proved to be an effective therapy in preclinical studies which evaluated a vast diversity of inflammatory conditions. MSCs lead to resolution of inflammation, preparation for regeneration and actual regeneration, and then ultimate return to normal baseline or homeostasis. However, in clinical trials of transplanted MSCs, the expectations of great medical benefit have not yet been fulfilled. As a practical alternative to MSC transplant, a synthetic drug with the capacity to boost endogenous MSC expansion and/or activation may also be effective. Regarding this, IMT504, the prototype of a major class of immunomodulatory oligonucleotides, induces in vivo expansion of MSCs, resulting in a marked improvement in preclinical models of neuropathic pain, osteoporosis, diabetes and sepsis. IMT504 is easily manufactured and has an excellent preclinical safety record. In the small number of patients studied thus far, IMT504 has been well-tolerated, even at very high dosage. Further clinical investigation is necessary to demonstrate the utility of IMT504 for resolution of inflammation and regeneration in a broad array of human diseases that would likely benefit from an immunoprotective/immunoregenerative therapy.
Collapse
|
32
|
Li J, Deng G, Wang H, Yang M, Yang R, Li X, Zhang X, Yuan H. Interleukin-1β pre-treated bone marrow stromal cells alleviate neuropathic pain through CCL7-mediated inhibition of microglial activation in the spinal cord. Sci Rep 2017; 7:42260. [PMID: 28195183 PMCID: PMC5307320 DOI: 10.1038/srep42260] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
Although neuropathic pain is one of the most intractable diseases, recent studies indicate that systemic or local injection of bone marrow stromal cells (BMSCs) decreases pro-inflammatory cytokines release and alleviates neuropathic pain. However, it is still not clear whether pre-treated BMSCs have a strong anti-inflammatory and/or analgesia effect. Using the spinal nerve ligation model of neuropathic pain, IL-1β pre-treated BMSCs (IL-1β-BMSCs) were injected into rats followed by SNL in order to determine possible effects. Results indicated that IL-1β-BMSCs were more efficacious in both amelioration of neuropathic pain and inhibition of microglia activation. Specifically, microglia inhibition was found to be mediated by chemokine C-C motif ligand 7 (CCL7) but not CCL2. Results also showed that IL-1β-BMSCs had a stronger inhibitory effect on astrocyte activation as well as CCL7 release, which was found to be mediated by IL-10 not transforming growth factor-β1. In addition, we also found directional migration of IL-1β-BMSCs was mediated by inceased C-X-C motif chemokine ligand (CXCL) 13 expression following SNL. In conclusion, our results indicated IL-1β-BMSCs could inhibit microglia activation and neuropathic pain by decreasing CCL7 level in spinal cord.
Collapse
Affiliation(s)
- Jian Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Haowei Wang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Mei Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Rui Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Xiangnan Li
- Department of Anesthesiology, the Third People's Hospital of Yancheng, Yancheng, 224001, China
| | - Xiaoping Zhang
- Department of Interventional &Vascular Surgery, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
33
|
Xie J, Xiao D, Xu Y, Zhao J, Jiang L, Hu X, Zhang Y, Yu L. Up-regulation of immunomodulatory effects of mouse bone-marrow derived mesenchymal stem cells by tetrahydrocannabinol pre-treatment involving cannabinoid receptor CB2. Oncotarget 2016; 7:6436-47. [PMID: 26824325 PMCID: PMC4872725 DOI: 10.18632/oncotarget.7042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/19/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic pain is commonly and closely correlated with inflammation. Both cannabinoid signaling and mesenchymal stem cells (MSCs) have been demonstrated to reduce inflammatory pain. Although cannabinoid signaling is essential for mesenchymal stem cell survival and differentiation, little is known about its role in modulatory effect of MSCs on inflammation and pain sensitivity. Here we showed that mouse bone-marrow derived MSCs (BM-MSCs) expressed both cannabinoid receptor type 1 and 2 (CB1 and CB2). CB2 expression level in BM-MSCs increased with their maturation. In addition, we found that tetrahydrocannabinol (THC) activated CB2 receptor and ERK signaling, consequently enhancing the modulation of MSCs on inflammation-associated cytokine release from lipopolysaccharides-stimulated microglia. Consistent with in vitro data, THC pretreatment enhanced the immunomodulatory effects of BM-MSC on thermal hyperalgesia and mechanical allodynia in chronic constriction injury model, by decreasing the release of pro-inflammation cytokines. Our study revealed the crucial role of THC in promoting the immunomodulatory effects of MSCs and proposed a new strategy to alleviate pain based on stem cells therapy.
Collapse
Affiliation(s)
- Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Dongju Xiao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, People's Republic of China
| | - Yun Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, People's Republic of China
| | - Jinning Zhao
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Li Jiang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xuming Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, People's Republic of China
| | - Yaping Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, People's Republic of China
| | - Lina Yu
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
34
|
Kano F, Matsubara K, Ueda M, Hibi H, Yamamoto A. Secreted Ectodomain of Sialic Acid-Binding Ig-Like Lectin-9 and Monocyte Chemoattractant Protein-1 Synergistically Regenerate Transected Rat Peripheral Nerves by Altering Macrophage Polarity. Stem Cells 2016; 35:641-653. [PMID: 27862629 DOI: 10.1002/stem.2534] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/30/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022]
Abstract
Peripheral nerves (PNs) exhibit remarkable self-repairing reparative activity after a simple crush or cut injury. However, the neuronal transection involving a nerve gap overwhelms their repairing activity and causes persistent paralysis. Here, we show that an implantation of the serum-free conditioned medium from stem cells from human exfoliated deciduous teeth (SHED-CM) immersed in a collagen sponge into the nerve gap formed by rat facial nerves transection restored the neurological function. In contrast, SHED-CM specifically depleted of a set of anti-inflammatory M2 macrophage inducers, monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9) lost the ability to restore neurological function in this model. Notably, the combination of MCP-1 and sSiglec-9 induced the polarization of M2 macrophages in vitro, resulting in the expression of multiple trophic factors that enhanced proliferation, migration, and differentiation of Schwann cells, blood vessel formation, and nerve fiber extension. Furthermore, the implantation of a collagen graft containing MCP-1/sSiglec-9 into the nerve gap induced anti-inflammatory M2 macrophage polarization, generated a Schwann-cell bridge instead of fibrotic scar, induced axonal regrowth, and restored nerve function. The specific elimination of M2 macrophages by Mannosylated-Clodrosome suppressed the MCP-1/sSiglec-9-mediated neurological recovery. Taken together, our data suggest that MCP-1/sSiglec-9 regenerates PNs by inducing tissue-repairing M2 macrophages and may provide therapeutic benefits for severe peripheral nerve injuries. Stem Cells 2017;35:641-653.
Collapse
Affiliation(s)
- Fumiya Kano
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Kohki Matsubara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
35
|
Chen C, Chen F, Yao C, Shu S, Feng J, Hu X, Hai Q, Yao S, Chen X. Intrathecal Injection of Human Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorates Neuropathic Pain in Rats. Neurochem Res 2016; 41:3250-3260. [PMID: 27655256 DOI: 10.1007/s11064-016-2051-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/23/2016] [Accepted: 08/27/2016] [Indexed: 02/05/2023]
Abstract
Neuropathic pain (NP) is a clinically incurable disease with miscellaneous causes, complicated mechanisms and available therapies show poor curative effect. Some recent studies have indicated that neuroinflammation plays a vital role in the occurrence and promotion of NP and anti-inflammatory therapy has the potential to relieve the pain. During the past decades, mesenchymal stem cells (MSCs) with properties of multipotentiality, low immunogenicity and anti-inflammatory activity have showed excellent therapeutic effects in cell therapy from animal models to clinical application, thus aroused great attention. However there are no reports about the effect of intrathecal human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) on NP which is induced by peripheral nerve injury. Therefore, in this study, intrathecally transplanted HUC-MSCs were utilized to examine the effect on neuropathic pain induced by a rat model with spinal nerve ligation (SNL), so as to explore the possible mechanism of those effects. As shown in the results, the HUC-MSCs transplantation obviously ameliorated SNL-induced mechanical allodynia and thermal hyperalgesia, which was related to the inhibiting process of neuroinflammation, including the suppression of activated astrocytes and microglia, as well as the significant reduction of pro-inflammatory cytokines Interleukin-1β (IL-1β) and Interleukin -17A (IL-17A) and the up-regulation of anti-inflammatory cytokine Interleukin -10 (IL-10). Therefore, through the effect on glial cells, pro-inflammatory and anti-inflammatory cytokine, the targeting intrathecal HUC-MSCs may offer a novel treatment strategy for NP.
Collapse
Affiliation(s)
- Chunxiu Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengfeng Chen
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengye Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaofang Shu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Feng
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesia, The First Affiliated Hospital of University of South China, Henyang, China
| | - Xiaoling Hu
- Department of Anesthesia, The First Affiliated Hospital of University of South China, Henyang, China
| | - Quan Hai
- Sichuan Province Regenerative Medicine Engineering Technology Research Center, Chengdu, China
| | - Shanglong Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Hofer HR, Tuan RS. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther 2016; 7:131. [PMID: 27612948 PMCID: PMC5016979 DOI: 10.1186/s13287-016-0394-0] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stem cells (MSCs) represent a subject of intense experimental and biomedical interest. Recently, trophic activities of MSCs have become the topic of a number of revealing studies that span both basic and clinical fields. In this review, we focus on recent investigations that have elucidated trophic mechanisms and shed light on MSC clinical efficacy relevant to musculoskeletal applications. Innate differences due to MSC sourcing may play a role in the clinical utility of isolated MSCs. Pain management, osteochondral, nerve, or blood vessel support by MSCs derived from both autologous and allogeneic sources have been examined. Recent mechanistic insights into the trophic activities of these cells point to ultimate regulation by nitric oxide, nuclear factor-kB, and indoleamine, among other signaling pathways. Classic growth factors and cytokines-such as VEGF, CNTF, GDNF, TGF-β, interleukins (IL-1β, IL-6, and IL-8), and C-C ligands (CCL-2, CCL-5, and CCL-23)-serve as paracrine control molecules secreted or packaged into extracellular vesicles, or exosomes, by MSCs. Recent studies have also implicated signaling by microRNAs contained in MSC-derived exosomes. The response of target cells is further regulated by their microenvironment, involving the extracellular matrix, which may be modified by MSC-produced matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs. Trophic activities of MSCs, either resident or introduced exogenously, are thus intricately controlled, and may be further fine-tuned via implant material modifications. MSCs are actively being investigated for the repair and regeneration of both osteochondral and other musculoskeletal tissues, such as tendon/ligament and meniscus. Future rational and effective MSC-based musculoskeletal therapies will benefit from better mechanistic understanding of MSC trophic activities, for example using analytical "-omics" profiling approaches.
Collapse
Affiliation(s)
- Heidi R Hofer
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
37
|
Hua Z, Liu L, Shen J, Cheng K, Liu A, Yang J, Wang L, Qu T, Yang H, Li Y, Wu H, Narouze J, Yin Y, Cheng J. Mesenchymal Stem Cells Reversed Morphine Tolerance and Opioid-induced Hyperalgesia. Sci Rep 2016; 6:32096. [PMID: 27554341 PMCID: PMC4995471 DOI: 10.1038/srep32096] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/02/2016] [Indexed: 01/27/2023] Open
Abstract
More than 240 million opioid prescriptions are dispensed annually to treat pain in the US. The use of opioids is commonly associated with opioid tolerance (OT) and opioid-induced hyperalgesia (OIH), which limit efficacy and compromise safety. The dearth of effective way to prevent or treat OT and OIH is a major medical challenge. We hypothesized that mesenchymal stem cells (MSCs) attenuate OT and OIH in rats and mice based on the understanding that MSCs possess remarkable anti-inflammatory properties and that both OT and chronic pain are associated with neuroinflammation in the spinal cord. We found that the development of OT and OIH was effectively prevented by either intravenous or intrathecal MSC transplantation (MSC-TP), which was performed before morphine treatment. Remarkably, established OT and OIH were significantly reversed by either intravenous or intrathecal MSCs when cells were transplanted after repeated morphine injections. The animals did not show any abnormality in vital organs or functions. Immunohistochemistry revealed that the treatments significantly reduced activation level of microglia and astrocytes in the spinal cord. We have thus demonstrated that MSC-TP promises to be a potentially safe and effective way to prevent and reverse two of the major problems of opioid therapy.
Collapse
Affiliation(s)
- Zhen Hua
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA.,Department of Anesthesiology, Beijing Hospital, No. 1 Dahua Road, Beijing 100730, China
| | - LiPing Liu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jun Shen
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Kathleen Cheng
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Aijun Liu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jing Yang
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Lina Wang
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Tingyu Qu
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - HongNa Yang
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yan Li
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Haiyan Wu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - John Narouze
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Yan Yin
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jianguo Cheng
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| |
Collapse
|
38
|
Chiang CY, Liu SA, Sheu ML, Chen FC, Chen CJ, Su HL, Pan HC. Feasibility of Human Amniotic Fluid Derived Stem Cells in Alleviation of Neuropathic Pain in Chronic Constrictive Injury Nerve Model. PLoS One 2016; 11:e0159482. [PMID: 27441756 PMCID: PMC4956194 DOI: 10.1371/journal.pone.0159482] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 07/05/2016] [Indexed: 01/01/2023] Open
Abstract
Purpose The neurobehavior of neuropathic pain by chronic constriction injury (CCI) of sciatic nerve is very similar to that in humans, and it is accompanied by a profound local inflammation response. In this study, we assess the potentiality of human amniotic fluid derived mesenchymal stem cells (hAFMSCs) for alleviating the neuropathic pain in a chronic constriction nerve injury model. Methods and Methods This neuropathic pain animal model was conducted by four 3–0 chromic gut ligatures loosely ligated around the left sciatic nerve in Sprague—Dawley rats. The intravenous administration of hAFMSCs with 5x105 cells was conducted for three consecutive days. Results The expression IL-1β, TNF-α and synaptophysin in dorsal root ganglion cell culture was remarkably attenuated when co-cultured with hAFMSCs. The significant decrease of PGP 9.5 in the skin after CCI was restored by administration of hAFMSCs. Remarkably increased expression of CD 68 and TNF-α and decreased S-100 and neurofilament expression in injured nerve were rescued by hAFMSCs administration. Increases in synaptophysin and TNF-α over the dorsal root ganglion were attenuated by hAFMSCs. Significant expression of TNF-α and OX-42 over the dorsal spinal cord was substantially attenuated by hAFMSCs. The increased amplitude of sensory evoked potential as well as expression of synaptophysin and TNF-α expression was alleviated by hAFMSCs. Human AFMSCs significantly improved the threshold of mechanical allodynia and thermal hyperalgesia as well as various parameters of CatWalk XT gait analysis. Conclusion Human AFMSCs administration could alleviate the neuropathic pain demonstrated in histomorphological alteration and neurobehavior possibly through the modulation of the inflammatory response.
Collapse
Affiliation(s)
- Chien-Yi Chiang
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Shih-An Liu
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Fu-Chou Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hong-Lin Su
- Institute of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Hung-Chuan Pan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
39
|
Yousefifard M, Nasirinezhad F, Shardi Manaheji H, Janzadeh A, Hosseini M, Keshavarz M. Human bone marrow-derived and umbilical cord-derived mesenchymal stem cells for alleviating neuropathic pain in a spinal cord injury model. Stem Cell Res Ther 2016; 7:36. [PMID: 26957122 PMCID: PMC4784350 DOI: 10.1186/s13287-016-0295-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/03/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Stem cell therapy can be used for alleviating the neuropathic pain induced by spinal cord injuries (SCIs). However, survival and differentiation of stem cells following their transplantation vary depending on the host and intrinsic factors of the cell. Therefore, the present study aimed to determine the effect of stem cells derived from bone marrow (BM-MSC) and umbilical cord (UC-MSC) on neuropathic pain relief. METHODS A compression model was used to induce SCI in a rat model. A week after SCI, about 1 million cells were transplanted into the spinal cord. Behavioral tests, including motor function recovery, mechanical allodynia, cold allodynia, mechanical hyperalgesia, and thermal hyperalgesia, were carried out every week for 8 weeks after SCI induction. A single unit recording and histological evaluation were then performed. RESULTS We show that BM-MSC and UC-MSC transplantations led to improving functional recovery, allodynia, and hyperalgesia. No difference was seen between the two cell groups regarding motor recovery and alleviating the allodynia and hyperalgesia. These cells survived in the tissue at least 8 weeks and prevented cavity formation due to SCI. However, survival rate of UC-MSC was significantly higher than BM-MSC. Electrophysiological evaluations showed that transplantation of UC-MSC brings about better results than BM-MSCs in wind up of wide dynamic range neurons. CONCLUSIONS The results of the present study show that BM-MSC and UC-MSC transplantations alleviated the symptoms of neuropathic pain and resulted in subsequent motor recovery after SCI. However, survival rate and electrophysiological findings of UC-MSC were significantly better than BM-MSC.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farinaz Nasirinezhad
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Department of Physiology, Iran University of Medical Sciences, Tehran, Iran.
| | - Homa Shardi Manaheji
- Department of Physiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Atousa Janzadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran. .,Pediatric Chronic Kidney Disease Research Center, Childrens Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mansoor Keshavarz
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Kiyomoto M, Shinoda M, Honda K, Nakaya Y, Dezawa K, Katagiri A, Kamakura S, Inoue T, Iwata K. p38 phosphorylation in medullary microglia mediates ectopic orofacial inflammatory pain in rats. Mol Pain 2015; 11:48. [PMID: 26260484 PMCID: PMC4531532 DOI: 10.1186/s12990-015-0053-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/03/2015] [Indexed: 12/22/2022] Open
Abstract
Background Orofacial inflammatory pain is likely to accompany referred pain in uninflamed orofacial structures. The ectopic pain precludes precise diagnosis and makes treatment problematic, because the underlying mechanism is not well understood. Using the established ectopic orofacial pain model induced by complete Freund’s adjuvant (CFA) injection into trapezius muscle, we analyzed the possible role of p38 phosphorylation in activated microglia in ectopic orofacial pain. Results Mechanical allodynia in the lateral facial skin was induced following trapezius muscle inflammation, which accompanied microglial activation with p38 phosphorylation and hyperexcitability of wide dynamic range (WDR) neurons in the trigeminal spinal subnucleus caudalis (Vc). Intra-cisterna successive administration of a p38 mitogen-activated protein kinase selective inhibitor, SB203580, suppressed microglial activation and its phosphorylation of p38. Moreover, SB203580 administration completely suppressed mechanical allodynia in the lateral facial skin and enhanced WDR neuronal excitability in Vc. Microglial interleukin-1β over-expression in Vc was induced by trapezius muscle inflammation, which was significantly suppressed by SB203580 administration. Conclusions These findings indicate that microglia, activated via p38 phosphorylation, play a pivotal role in WDR neuronal hyperexcitability, which accounts for the mechanical hypersensitivity in the lateral facial skin associated with trapezius muscle inflammation.
Collapse
Affiliation(s)
- Masaaki Kiyomoto
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Kuniya Honda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Yuka Nakaya
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Ko Dezawa
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Ayano Katagiri
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Satoshi Kamakura
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Tomio Inoue
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| |
Collapse
|
41
|
Geng CK, Cao HH, Ying X, Yu HL. Effect of mesenchymal stem cells transplantation combining with hyperbaric oxygen therapy on rehabilitation of rat spinal cord injury. ASIAN PAC J TROP MED 2015. [PMID: 26194832 DOI: 10.1016/j.apjtm.2015.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To investigate the effect of BMSCs transplantation plus hyperbaric oxygen (HBO) on repair of rat SCI. METHODS Seventy five male rats were divided randomly into five groups: sham, vehicle, BMSCs transplantation group, combination group, 15 rats in each group. Every week after the SCI onset, all animals were evaluated for behavior outcome by Basso-Beattle-Bresnahan (BBB) score and inclined plane test. Axon recovery was examined with focal spinal cord tissue by electron microscope at 6 weeks after the SCI onset. HE staining and BrdU staining were performed to examine the BMSCs and lesion post injury. Somatosensory evoked potential (SEP) testing was performed to detect the recovery of neural conduction. RESULTS Results from the behavior tests from combination group were significant higher than rats which received only transplantation or HBO treatment. Results from histopathology showed favorable recovery from combination group than other treatment groups. The number of BrdU(+) in combination group were measureable more than transplantation group (P < 0.05). The greatest decrease in TNF-α, IL-1β, IL-6, IFN-α determined by Elisa assay in combination group were evident too. CONCLUSIONS BMSCs transplantation can promote the functional recovery of rat hind limbs after SCI, and its combination with HBO has a synergistic effect.
Collapse
Affiliation(s)
- Cheng-Kui Geng
- Department of Orthopedics, Yan'an Hospital of Kunming City, the Affiliated Hospital of Kunming Medical University, Kunming 650051, China; Department of Minimally Invasive Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Hong-Hua Cao
- Department of Hematology, Tumor Hospital of Yunnan Province & The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Xiong Ying
- Department of Orthopedics, Yan'an Hospital of Kunming City, the Affiliated Hospital of Kunming Medical University, Kunming 650051, China
| | - Hua-Lin Yu
- Department of Minimally Invasive Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
42
|
Yarar E, Kuruoglu E, Kocabıcak E, Altun A, Genc E, Ozyurek H, Kefeli M, Marangoz AH, Aydın K, Cokluk C. Electrophysiological and histopathological effects of mesenchymal stem cells in treatment of experimental rat model of sciatic nerve injury. Int J Clin Exp Med 2015; 8:8776-84. [PMID: 26309529 PMCID: PMC4537965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/03/2015] [Indexed: 06/04/2023]
Abstract
AIM The aim of this study was to evaluate electrophysiological and histopathological effects of mesenchymal stem cells in treatment of sciatic nerve injury. MATERIAL AND METHODS Thirty-two female Spraque-Dawley rat were used in this study. Eight rats were used as a reference group in electrophysiological analysis for evaluation of non-injured nerve recordings (Control Group). Twenty-four rats were used for experimental evaluation. Twelve rats were anastomosed without treatment with mesenchymal stem cells (Sham Group) and twelve other rats were anastomosed and treated with mesenchymal stem cells (Stem Cell Group). Surgicel and bioglue were used in anastomosed line in both Groups. Eight weeks after the surgery, electrophysiological evaluation of rats was performed and, then, rats were decapitated under anesthesia and specimens including sciatic nerves and anastomosed line were taken for histopathological evaluation. Electromyography and nerve conduction velocity testing and histopathological scoring including rate of Wallerian degeneration, and neuroma and scar formation were evaluated for both Groups. RESULTS There were no statistically significant differences between Sham and Stem Cell Groups with respect to histopathological evaluation. However, nerve conduction velocity showed significant difference between groups (P = 0.001). Nerve conduction velocity was significantly improved in Stem Cell Group when compared to Sham Group. CONCLUSION In this study, based on nerve conduction velocity data, it was concluded that treatment with mesenchymal stem cells during end-to-end anastomosis improves functional regeneration.
Collapse
Affiliation(s)
- Ercan Yarar
- Department of Neurosurgery, Atatürk State HospitalSinop, Turkey
| | - Enis Kuruoglu
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Ersoy Kocabıcak
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Adnan Altun
- Department of Neurosurgery, Medicana Private HospitalSamsun, Turkey
| | - Eyup Genc
- Department of Neurosurgery, Turhal State HospitalTokat, Turkey
| | - Hamit Ozyurek
- Department of Pediatric Neurology, Ondokuz Mayis UniversitySamsun, Turkey
| | - Mehmet Kefeli
- Department of Pathology, Ondokuz Mayis UniversitySamsun, Turkey
| | | | - Keramettin Aydın
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| | - Cengiz Cokluk
- Department of Neurosurgery, Ondokuz Mayis UniversitySamsun, Turkey
| |
Collapse
|
43
|
Hosseini M, Yousefifard M, Aziznejad H, Nasirinezhad F. The Effect of Bone Marrow-Derived Mesenchymal Stem Cell Transplantation on Allodynia and Hyperalgesia in Neuropathic Animals: A Systematic Review with Meta-Analysis. Biol Blood Marrow Transplant 2015; 21:1537-44. [PMID: 25985918 DOI: 10.1016/j.bbmt.2015.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 05/07/2015] [Indexed: 01/02/2023]
Abstract
Stem cell transplantation has been considered a possible therapeutic method for neuropathic pain. However, no quantitative data synthesis of stem cell therapy for neuropathic pain exists. Therefore, the present systematic review and meta-analysis assessed the efficacy of bone marrow mesenchymal stem cell (BMMSC) transplantation on alleviating pain symptoms in animal models of neuropathic pain. In the present meta-analysis, controlled animal studies assessing the effect of administrating BMMSC on neuropathic pain were included through an extensive literature search of online databases. After collecting data, effect sizes were computed and the standardized mean difference (SMD) with 95% confidence interval (CI) was entered in all analyses. Random-effects models were used for data analysis. Sensitivity and subgroup analyses were performed to investigate expected or measured heterogeneity. Finally, 14 study were included. The analyses showed that BMMSC transplantation lead to significant improvement on allodynia (SMD = 2.06; 95% CI, 1.09 to 3.03; I(2) = 99.7%; P < .001). The type of neuropathy (P = .036), time between injury and intervention (P = .02), and the number of transplanted cells (P = .023) influence the improvement of allodynia after BMMSC transplantation. BMMSC transplantation has no effect on hyperalgesia (SMD = .3; 95% CI, -1.09 to 1.68; I(2) = 100%; P < .001) unless it occurs during the first 4 days after injury (P = .02). The present systematic review with meta-analysis suggests that BMMSC transplantation improves allodynia but does not have any significant effect on hyperalgesia unless it is given during the first 4 days after injury.
Collapse
Affiliation(s)
- Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heidar Aziznejad
- The Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farinaz Nasirinezhad
- Physiology Research Center, Department of Physiology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Yu H, Fischer G, Ebert AD, Wu HE, Bai X, Hogan QH. Analgesia for neuropathic pain by dorsal root ganglion transplantation of genetically engineered mesenchymal stem cells: initial results. Mol Pain 2015; 11:5. [PMID: 25888914 PMCID: PMC4331376 DOI: 10.1186/s12990-015-0002-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/26/2015] [Indexed: 02/07/2023] Open
Abstract
Background Cell-based therapy may hold promise for treatment of chronic pain. Mesenchymal stem cells (MSCs) are readily available and robust, and their secretion of therapeutic peptides can be enhanced by genetically engineering. We explored the analgesic potential of transplanting bone marrow-derived MSCs that have been transduced with lentivectors. To optimize efficacy and safety, primary sensory neurons were targeted by MSC injection into the dorsal root ganglia (DRGs). Results MSCs were transduced using lentivectors to express enhanced green fluorescent protein (EGFP) or to co-express the analgesic peptide glial cell line-derived neurotrophic factor (GDNF) and EGFP by a viral 2A bicistronic transgene cassette. Engineered MSCs were injected into the 4th lumbar (L4) and L5 DRGs of adult allogeneic rats to evaluate survival in the DRGs. MSCs were detected by immunofluorescence staining up to 2–3 weeks after injection, distributed in the extracellular matrix space without disrupting satellite glial cell apposition to sensory neurons, suggesting well-tolerated integration of engrafted MSCs into DRG tissue. To examine their potential for inhibiting development of neuropathic pain, MSCs were injected into the L4 and L5 DRGs ipsilateral to a spinal nerve ligation injury. Animals injected with GDNF-engineered MSCs showed moderate but significant reduction in mechanical allodynia and hyperalgesia compared to controls implanted with MSCs expressing EGFP alone. We also observed diminished long-term survival of allografted MSCs at 3 weeks, and the development of a highly-proliferating population of MSCs in 12% of DRGs after transplantation. Conclusions These data indicate that genetically modified MSCs secreting analgesic peptides could potentially be developed as a novel DRG-targeted cell therapy for treating neuropathic pain. However, further work is needed to address the challenges of MSC survival and excess proliferation, possibly with trials of autologous MSCs, evaluation of clonally selected populations of MSCs, and investigation of regulation of MSC proliferation.
Collapse
Affiliation(s)
- Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Gregory Fischer
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA. .,Zablocki Veterans Affairs Medical Center, 5000 W National Ave, Milwaukee, WI, 53295, USA.
| |
Collapse
|