1
|
Wu S, Chu X, Lv G, Gao J, Huang Y, Li H, Jiang X, Liu Y, Zhang J, Fang X, Yao Z, Bu W. Mesenchymal Stem Cells With Polydopamine-Coated NaGdF 4 Nanoparticles with Ca 2+ Chelation Ability for Ischemic Stroke Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416020. [PMID: 39887461 DOI: 10.1002/adma.202416020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/21/2024] [Indexed: 02/01/2025]
Abstract
Mesenchymal stem cells (MSCs) transplantation is a promising therapeutic strategy for ischemic stroke. However, the survival of transplanted MSCs is often compromised by the excessive levels of reactive oxygen species (ROS) and calcium ions (Ca2+) in the ischemic microenvironment following blood flow occlusion. In this study, a protective strategy is developed using functional nanomaterials to escort and shield MSCs. Specifically, NaGdF4@PDA-ALD nanoparticles (NPANs) are synthesized, featuring a NaGdF4 core coated with polydopamine (PDA) for ROS scavenging and further modified with alendronate sodium (ALD) for Ca2+ chelation. The internalization of NPANs by MSCs protected them from oxidative damage and calcium overload, thereby promoting their viability and functionality. Furthermore, NaGdF4 generated T1 signal enhancement, enabling in vivo tracking of MSCs via magnetic resonance imaging. The NPANs-treated MSCs demonstrated improved survival and migration to the ischemic region, promoting blood flow restoration and angiogenesis. These findings confirm the feasibility of employing functional nanoparticles to augment MSCs-based therapies, offering a promising strategy to improve their therapeutic efficacy in ischemic stroke treatment.
Collapse
Affiliation(s)
- Shiman Wu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xu Chu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| | - Guanglei Lv
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| | - Jiahao Gao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxin Huang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| | - Jiawen Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiangming Fang
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Jiangsu, 214023, P. R. China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Wenbo Bu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
2
|
Athiel Y, Cariot L, Jouannic JM, Maillet C, Mauffré V, Adam C, Huet H, Larghero J, Nasone J, Guilbaud L. Safety and efficacy of human umbilical cord-derived mesenchymal stromal cells in fetal ovine myelomeningocele repair. Stem Cell Res Ther 2024; 15:444. [PMID: 39568021 PMCID: PMC11580231 DOI: 10.1186/s13287-024-03991-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND The aim of this study was to assess the safety and efficacy of human umbilical cord mesenchymal stromal cells (hUC-MSCs) patch used as an adjuvant therapy in fetal myelomeningocele (MMC) surgery in the ovine model. METHODS hUC-MSCs were isolated from human umbilical cords (UC) using the explant method, cultured and characterized. hUC-MSCs were then embedded in a fibrin patch. MMC were surgically created at 75 days of gestation and repaired at 89 days of gestation in sheep fetuses. Two groups were compared: the hUC-MSCs group in which MMC was repaired using a cellular patch and the control group, in which MMC was repaired using an acellular patch. Safety was evaluated by clinical ewes' monitoring during gestation, and clinical and histological examinations of lambs after birth. Efficacy was assessed by clinical neurological evaluation at 2 and 24 h of life using the sheep locomotor rating scale and by histological analyses. RESULTS Among the 17 operated lambs, nine were born alive: six in the hUC-MSCs group and three in the control group. Overall fetal loss was 47% (8/17) without differences between the two groups. No fever was reported in ewes. No tumors were detected in clinical and histological examinations in the lambs. At 24 h of life, mean Sheep Locomotor Rating score was higher in the hUC-MSCs group than in the control group: 15.0 versus 2.0 (p = 0.07). Histological analyses showed a higher large neurons density in the hUC-MSCs group in comparison with the control group: 9.9 versus 6.3/mm2 of gray matter (p = 0.04). Lambs in the hUC-MSCs group had lower fibrosis around the spinal cord and at the level of the MMC scar: 70.9 versus 253.7 μm (p = 0.10) and 691.3 versus 1684.4 μm (p = 0,18), respectively. CONCLUSIONS Ovine fetal repair of MMC using human UC-MSCs seems to be an effective and safe procedure.
Collapse
Affiliation(s)
- Yoann Athiel
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Laura Cariot
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Jean-Marie Jouannic
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France
| | - Corentin Maillet
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Vincent Mauffré
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Clovis Adam
- Service d'anatomopathologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Hélène Huet
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jérôme Larghero
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Justine Nasone
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Lucie Guilbaud
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France.
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France.
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France.
| |
Collapse
|
3
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [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: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
4
|
Yu YQ, Wang H. Imbalance of Th1 and Th2 Cytokines and Stem Cell Therapy in Pathological Pain. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:88-101. [PMID: 36573059 DOI: 10.2174/1871527322666221226145828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/05/2022] [Accepted: 11/12/2022] [Indexed: 12/28/2022]
Abstract
The pathophysiological importance of T helper 1 (Th1) and Th2 cell cytokines in pathological pain has been highly debated in recent decades. However, the analgesic strategy targeting individual cytokines still has a long way to go for clinical application. In this review, we focus on the contributions of Th1 cytokines (TNF-α, IFN-γ, and IL-2) and Th2 cytokines (IL-4, IL-5, IL-10, and IL-13) in rodent pain models and human pain-related diseases. A large number of studies have shown that Th1 and Th2 cytokines have opposing effects on pain modulation. The imbalance of Th1 and Th2 cytokines might determine the final effect of pain generation or inhibition. However, increasing evidence indicates that targeting the individual cytokine is not sufficient for the treatment of pathological pain. It is practical to suggest a promising therapeutic strategy against the combined effects of Th1 and Th2 cytokines. We summarize the current advances in stem cell therapy for pain-related diseases. Preclinical and clinical studies show that stem cells inhibit proinflammatory cytokines and release enormous Th2 cytokines that exhibit a strong analgesic effect. Therefore, a shift of the imbalance of Th1 and Th2 cytokines induced by stem cells will provide a novel therapeutic strategy against intractable pain. It is extremely important to reveal the cellular and molecular mechanisms of stem cell-mediated analgesia. The efficiency and safety of stem cell therapy should be carefully evaluated in animal models and patients with pathological pain.
Collapse
Affiliation(s)
- Yao-Qing Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Huan Wang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| |
Collapse
|
5
|
Hu H, Long H, Ren Z, Liu T, Xu J, Xiao F. Partially brain effects of injection of human umbilical cord mesenchymal stem cells at injury sites in a mouse model of thoracic spinal cord contusion. Front Mol Neurosci 2023; 16:1179175. [PMID: 37342099 PMCID: PMC10278944 DOI: 10.3389/fnmol.2023.1179175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/05/2023] [Indexed: 06/22/2023] Open
Abstract
Purpose The pain caused by spinal cord injury (SCI) poses a major burden on patients, and pain management is becoming a focus of treatment. Few reports have described changes in the brain after SCI. Particularly, the exact mechanism through which brain regions affect post-injury pain remains unclear. In this study, we aimed to determine the potential therapeutic mechanisms of pain. A mouse model of spinal cord contusion was established, and molecular expression in the anterior cingulate cortex (ACC) and periaqueductal gray (PAG) in the brain and animal behavior was observed after local injection of human umbilical cord mesenchymal stem cells (HU-MSCs) at the site of SCI. Method Sixty-three female C57BL/6J mice were divided into four groups: a sham operation group (n = 15); a spinal injury group (SCI, n = 16); an SCI + HU-MSCs group (n = 16) and an SCI + PBS group (n = 16), in which the SCI site was injected with HU-MSCs/phosphate buffer. The BMS score was determined, and the von Frey test and Hargreaves test were used to assess behavior every week after surgery. Mice were sacrificed in the fourth week after operation, and samples were collected. The expression of CGRP, Substance P, C-Fos and KCC2 in the ACC and PAG were observed with immunohistochemistry. Chromic cyanine staining was used to observe transverse sections of the injured spinal cord. Result In the ACC and PAG after SCI, the expression of CGRP, SP and C-Fos increased, and the expression of KCC2 decreased, whereas after HU-MSC injection, the expression of CGRP, SP and C-Fos decreased, and the expression of KCC2 increased. The SCI + HU-MSC group showed better exercise ability from 2 to 4 weeks after surgery than the SCI/SCI + PBS groups (P < 0.001). Local injection of HU-MSCs significantly improved the mechanical hyperalgesia caused by SCI in the fourth week after surgery (P < 0.0001), and sensation was significantly recovered 2 weeks after surgery (P < 0.0001); no improvement in thermal hypersensitivity was observed (P > 0.05). The HU-MSC group retained more white matter than the SCI/SCI + PBS groups (P < 0.0001). Conclusion Local transplantation of HU-MSCs at the site of SCI partially relieves the neuropathic pain and promotes recovery of motor function. These findings suggest a feasible direction for the future treatment of SCI.
Collapse
Affiliation(s)
- Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Houqing Long
- Department of Spine Surgery, Orthopaedic, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University/The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Zhenxiao Ren
- Department of Spine Surgery, Orthopaedic, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University/The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology/Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tianhua Liu
- Department of Oncology, Guangzhou Modern Hospital, Guangzhou, Guangdong, China
| | - Jinghui Xu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology/Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
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
|
7
|
Soares MBP, Gonçalves RGJ, Vasques JF, da Silva-Junior AJ, Gubert F, Santos GC, de Santana TA, Almeida Sampaio GL, Silva DN, Dominici M, Mendez-Otero R. Current Status of Mesenchymal Stem/Stromal Cells for Treatment of Neurological Diseases. Front Mol Neurosci 2022; 15:883378. [PMID: 35782379 PMCID: PMC9244712 DOI: 10.3389/fnmol.2022.883378] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders include a wide spectrum of clinical conditions affecting the central and peripheral nervous systems. For these conditions, which affect hundreds of millions of people worldwide, generally limited or no treatments are available, and cell-based therapies have been intensively investigated in preclinical and clinical studies. Among the available cell types, mesenchymal stem/stromal cells (MSCs) have been widely studied but as yet no cell-based treatment exists for neurological disease. We review current knowledge of the therapeutic potential of MSC-based therapies for neurological diseases, as well as possible mechanisms of action that may be explored to hasten the development of new and effective treatments. We also discuss the challenges for culture conditions, quality control, and the development of potency tests, aiming to generate more efficient cell therapy products for neurological disorders.
Collapse
Affiliation(s)
- Milena B. P. Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Renata G. J. Gonçalves
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J. da Silva-Junior
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Gubert
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Girlaine Café Santos
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Thaís Alves de Santana
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Gabriela Louise Almeida Sampaio
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | | | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Rosalia Mendez-Otero
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Effects of mesenchymal stem cell transplantation on spinal cord injury patients. Cell Tissue Res 2022; 389:373-384. [PMID: 35697943 DOI: 10.1007/s00441-022-03648-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/02/2022] [Indexed: 11/02/2022]
Abstract
Spinal cord injury (SCI) is a traumatic injury with sensory and motor deficits that more than 1 million patients worldwide suffer from disability due to it. Many pharmacological therapies help reduce SCI-related injury and protect CNS from more damage but no current therapy could improve the axonal repair. In this regard, stem cell therapy is considered a regenerative method for SCI patient treatment. The neurotrophic and immunomodulatory factor secretion, differentiation, neuroprotecting, and remyelinating properties have made mesenchymal stem cells (MSCs) principally useful in this field. There are studies on the role of MSCs in patients suffering from SCI. However, low number of SCI patients and the lack of control groups in these studies, the cell transplantation appropriate methods, including cell source, dose, route of delivery, and transplantation timing, are various in trials. This study reviews the beneficial effects of MSC transplantation in SCI clinical studies with a special focus on the MSC properties and limitations of MSC transplantation.
Collapse
|
9
|
Yang Y, Tang Y, Qin H, Xu J. Efficacy of transcutaneous electrical nerve stimulation in people with pain after spinal cord injury: a meta-analysis. Spinal Cord 2022; 60:375-381. [PMID: 35277650 PMCID: PMC9106573 DOI: 10.1038/s41393-022-00776-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022]
Abstract
Study Design Meta-analysis. Objectives This study aimed to evaluate the effect of transcutaneous electrical nerve stimulation in people with pain after spinal cord injury by meta-analysis. Methods Reviewed PubMed, Embase, Cochrane library, as well as China National Knowledge Infrastructure (CNKI), Wanfang, and Vip databases to search the randomized controlled trials of pain after spinal cord injury through transcutaneous electrical nerve stimulation from the beginning of the library to March 2021, and analyze the literature with RevMan 5.3 software and the bias in the literature with STATA 12.0 software. Results There are six randomized controlled trials in the study with 165 cases. 83 cases in the test group were given transcutaneous electrical nerve stimulation, and 82 cases in the control group used sham stimulation or other treatments. Meta-analysis results showed the experimental group’s visual analog scale (MD = −1.52, 95%CI, −2.44 to −0.60, P = 0.001) and short-form McGill pain questionnaire scores (MD = −0.70, 95% CI, −1.03 to −0.25, P = 0.002) were lower than those of the control group. Conclusions Transcutaneous electrical nerve stimulation has some clinical therapeutic effects on persons with pain after spinal cord injury, but due to the lack of literature, the sample size is not large, and clinical trials need to be further improved later.
Collapse
Affiliation(s)
- Ye Yang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yun Tang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Huiqing Qin
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianwen Xu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
10
|
Pang QM, Chen SY, Xu QJ, Fu SP, Yang YC, Zou WH, Zhang M, Liu J, Wan WH, Peng JC, Zhang T. Neuroinflammation and Scarring After Spinal Cord Injury: Therapeutic Roles of MSCs on Inflammation and Glial Scar. Front Immunol 2021; 12:751021. [PMID: 34925326 PMCID: PMC8674561 DOI: 10.3389/fimmu.2021.751021] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Transected axons are unable to regenerate after spinal cord injury (SCI). Glial scar is thought to be responsible for this failure. Regulating the formation of glial scar post-SCI may contribute to axonal regrow. Over the past few decades, studies have found that the interaction between immune cells at the damaged site results in a robust and persistent inflammatory response. Current therapy strategies focus primarily on the inhibition of subacute and chronic neuroinflammation after the acute inflammatory response was executed. Growing evidences have documented that mesenchymal stem cells (MSCs) engraftment can be served as a promising cell therapy for SCI. Numerous studies have shown that MSCs transplantation can inhibit the excessive glial scar formation as well as inflammatory response, thereby facilitating the anatomical and functional recovery. Here, we will review the effects of inflammatory response and glial scar formation in spinal cord injury and repair. The role of MSCs in regulating neuroinflammation and glial scar formation after SCI will be reviewed as well.
Collapse
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qi-Jing Xu
- Department of Human Anatomy, Zunyi Medical University, Zunyi, China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi-Chun Yang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wang-Hui Zou
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Juan Liu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia-Chen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
11
|
The Conditioned Medium of Lactobacillus rhamnoides GG Regulates Microglia/Macrophage Polarization and Improves Functional Recovery after Spinal Cord Injury in Rats. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3376496. [PMID: 34337004 PMCID: PMC8289592 DOI: 10.1155/2021/3376496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/25/2021] [Indexed: 11/18/2022]
Abstract
Lactobacillus rhamnoides, a human intestinal colonizer, can act through various pathways to induce microglia/macrophages to produce cytokines and to polarize microglia/macrophages to different phenotypes to reduce the inflammatory response. In this article, we evaluated the treatment potential of the Lactobacillus rhamnoides GG conditioned medium (LGG-CM) in rat model with SCI (acute spinal cord injury), including functional, neurophysiological, and histological outcomes and the underlying neuroprotective mechanisms. In our experiment, LGG-CM (30 mg/kg) was injected directly into the injury site in rats immediately after SCI. Measured by the BBB scale (Basso, Beattie, and Bresnahan locomotor rating scale) and inclined plane test, rats in the LGG-CM-treated group showed better locomotor scores. Moreover, compared to the vehicle treatment group, LGG-CM increased the mRNA level of the M2 marker (CD206), and decreased that of the M1 marker (iNOS). Western blot assays showed that LGG-CM-treated SCI rats had a higher grayscale ratio of p65 and a lower ratio of p-IκBα/IκBα. Our study shows that local injection of LGG-CM after acute SCI can inhibit inflammatory responses and improve motor function recovery. These effects may be related with the inhibition to the NF-κB (The nuclear factor-kappa B) signal pathway which leads to M2 microglia/macrophage polarization.
Collapse
|
12
|
hiPSC-derived NSCs effectively promote the functional recovery of acute spinal cord injury in mice. Stem Cell Res Ther 2021; 12:172. [PMID: 33706803 PMCID: PMC7953804 DOI: 10.1186/s13287-021-02217-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 12/26/2022] Open
Abstract
Background Spinal cord injury (SCI) is a common disease that results in motor and sensory disorders and even lifelong paralysis. The transplantation of stem cells, such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), or subsequently generated stem/progenitor cells, is predicted to be a promising treatment for SCI. In this study, we aimed to investigate effect of human iPSC-derived neural stem cells (hiPSC-NSCs) and umbilical cord-derived MSCs (huMSCs) in a mouse model of acute SCI. Methods Acute SCI mice model were established and were randomly treated as phosphate-buffered saline (PBS) (control group), repaired with 1 × 105 hiPSC-NSCs (NSC group), and 1 × 105 huMSCs (MSC group), respectively, in a total of 54 mice (n = 18 each). Hind limb motor function was evaluated in open-field tests using the Basso Mouse Scale (BMS) at days post-operation (dpo) 1, 3, 5, and 7 after spinal cord injury, and weekly thereafter. Spinal cord and serum samples were harvested at dpo 7, 14, and 21. Haematoxylin-eosin (H&E) staining and Masson staining were used to evaluate the morphological changes and fibrosis area. The differentiation of the transplanted cells in vivo was evaluated with immunohistochemical staining. Results The hiPSC-NSC-treated group presented a significantly smaller glial fibrillary acidic protein (GFAP) positive area than MSC-treated mice at all time points. Additionally, MSC-transplanted mice had a similar GFAP+ area to mice receiving PBS. At dpo 14, the immunostained hiPSC-NSCs were positive for SRY-related high-mobility-group (HMG)-box protein-2 (SOX2). Furthermore, the transplanted hiPSC-NSCs differentiated into GFAP-positive astrocytes and beta-III tubulin-positive neurons, whereas the transplanted huMSCs differentiated into GFAP-positive astrocytes. In addition, hiPSC-NSC transplantation reduced fibrosis formation and the inflammation level. Compared with the control or huMSC transplanted group, the group with transplantation of hiPSC-NSCs exhibited significantly improved behaviours, particularly limb coordination. Conclusions HiPSC-NSCs promote functional recovery in mice with acute SCI by replacing missing neurons and attenuating fibrosis, glial scar formation, and inflammation. Graphical abstract ![]()
Collapse
|
13
|
Ultrashort Wave Combined with Human Umbilical Cord Mesenchymal Stem Cell (HUC-MSC) Transplantation Inhibits NLRP3 Inflammasome and Improves Spinal Cord Injury via MK2/TTP Signalling Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3021750. [PMID: 33376718 PMCID: PMC7738785 DOI: 10.1155/2020/3021750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/09/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022]
Abstract
Objective To investigate the curative effects of HUC-MSCs combined with USW on spinal cord injury (SCI) and the effects on inflammatory microenvironment and to explore the regulatory mechanisms of MK2/TTP signalling pathway and NLRP3 inflammasome. Methods The SCI rat model was established using the modified Allen method; rats were administered with USW, HUC-MSCs, and combination therapy of USW and HUC-MSCs; the therapeutic efficacies in each group of rats were monitored and represented in BBB score. SCI levels were observed using HE staining and IF. The microglia polarisation state and released contents of inflammatory factors were detected. IF and Western Blotting were performed on to detect the expression levels of MK2/TTP signalling pathway and NLRP3 inflammasome-related proteins. Furthermore, the regulatory mechanisms of MK2/TTP pathway and NLRP3 were explored by performing on the in vitro study. Results Combination therapy of USW and HUC-MSCs was found of significant efficacy on improving motor functions of SCI rats, and it was further proved that this combination therapy can reduce spinal cord injury in SCI rats, of which USW plays a more important role. While transplantation of HUC-MSCs can promote microglial cells developing to SCI repair, and M2 microglial cells were taking advantages gradually. The combination therapy can inhibit the expression of MK2; downregulate NLRP3 inflammasome; suppress the expression levels of pro-caspase-1, pro-IL-1β, and pro-IL-18; and simultaneously suppress the release of IL-1β and IL-18, thereby reducing spinal cord neurons apoptosis. It was found that the steady state of microglial polarisation maintained by combined treatment of USW and HUC-MSCs was destroyed with the upregulation of MK2 expression in cells, of which, M1 type microglial cell was dominant and the increased contents of inflammatory factors were detected. However, overexpressed MK2 relieved the inhibition of NLRP3 expression by TTP. Conclusions Combination therapy of USW and HUC-MSCs can downregulate NLRP3 expression, relieve inflammatory responses, improve immune microenvironment, and rescue spinal cord injury via suppressing phosphorylation level of MK2.
Collapse
|
14
|
Xie Q, Liu R, Jiang J, Peng J, Yang C, Zhang W, Wang S, Song J. What is the impact of human umbilical cord mesenchymal stem cell transplantation on clinical treatment? Stem Cell Res Ther 2020; 11:519. [PMID: 33261658 PMCID: PMC7705855 DOI: 10.1186/s13287-020-02011-z] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background Human umbilical cord mesenchymal stem cells (HUC-MSCs) present in the umbilical cord tissue are self-renewing and multipotent. They can renew themselves continuously and, under certain conditions, differentiate into one or more cell types constituting human tissues and organs. HUC-MSCs differentiate, among others, into osteoblasts, chondrocytes, and adipocytes and have the ability to secrete cytokines. The possibility of noninvasive harvesting and low immunogenicity of HUC-MSCs give them a unique advantage in clinical applications. In recent years, HUC-MSCs have been widely used in clinical practice, and some progress has been made in their use for therapeutic purposes. Main body This article describes two aspects of the clinical therapeutic effects of HUC-MSCs. On the one hand, it explains the benefits and mechanisms of HUC-MSC treatment in various diseases. On the other hand, it summarizes the results of basic research on HUC-MSCs related to clinical applications. The first part of this review highlights several functions of HUC-MSCs that are critical for their therapeutic properties: differentiation into terminal cells, immune regulation, paracrine effects, anti-inflammatory effects, anti-fibrotic effects, and regulating non-coding RNA. These characteristics of HUC-MSCs are discussed in the context of diabetes and its complications, liver disease, systemic lupus erythematosus, arthritis, brain injury and cerebrovascular diseases, heart diseases, spinal cord injury, respiratory diseases, viral infections, and other diseases. The second part emphasizes the need to establish an HUC-MSC cell bank, discusses tumorigenicity of HUC-MSCs and the characteristics of different in vitro generations of these cells in the treatment of diseases, and provides technical and theoretical support for the clinical applications of HUC-MSCs. Conclusion HUC-MSCs can treat a variety of diseases clinically and have achieved good therapeutic effects, and the development of HUC-MSC assistive technology has laid the foundation for its clinical application.
Collapse
Affiliation(s)
- Qixin Xie
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Rui Liu
- Department of Medical Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Jia Jiang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Jing Peng
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Chunyan Yang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Wen Zhang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Sheng Wang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Jing Song
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China.
| |
Collapse
|