1
|
Reis ALG, Maximino JR, Lage LADPC, Gomes HR, Pereira J, Brofman PRS, Senegaglia AC, Rebelatto CLK, Daga DR, Paiva WS, Chadi G. Proteomic analysis of cerebrospinal fluid of amyotrophic lateral sclerosis patients in the presence of autologous bone marrow derived mesenchymal stem cells. Stem Cell Res Ther 2024; 15:301. [PMID: 39278909 PMCID: PMC11403799 DOI: 10.1186/s13287-024-03820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/27/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressive motoneuron degenerative disorder. There are still no drugs capable of slowing disease evolution or improving life quality of ALS patients. Thus, autologous stem cell therapy has emerged as an alternative treatment regime to be investigated in clinical ALS. METHOD Using Proteomics and Protein-Protein Interaction Network analyses combined with bioinformatics, the possible cellular mechanisms and molecular targets related to mesenchymal stem cells (MSCs, 1 × 106 cells/kg, intrathecally in the lumbar region of the spine) were investigated in cerebrospinal fluid (CSF) of ALS patients who received intrathecal infusions of autologous bone marrow-derived MSCs thirty days after cell therapy. Data are available via ProteomeXchange with identifier PXD053129. RESULTS Proteomics revealed 220 deregulated proteins in CSF of ALS subjects treated with MSCs compared to CSF collected from the same patients prior to MSCs infusion. Bioinformatics enriched analyses highlighted events of Extracellular matrix and Cell adhesion molecules as well as related key targets APOA1, APOE, APP, C4A, C5, FGA, FGB, FGG and PLG in the CSF of cell treated ALS subjects. CONCLUSIONS Extracellular matrix and cell adhesion molecules as well as their related highlighted components have emerged as key targets of autologous MSCs in CSF of ALS patients. TRIAL REGISTRATION Clinicaltrial.gov identifier NCT0291768. Registered 28 September 2016.
Collapse
Affiliation(s)
- Ana Luiza Guimarães Reis
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Jessica Ruivo Maximino
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | | | - Hélio Rodrigues Gomes
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Juliana Pereira
- LIM-31, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine and Life Sciences, PontifÃcia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine and Life Sciences, PontifÃcia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine and Life Sciences, PontifÃcia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine and Life Sciences, PontifÃcia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Wellingson Silva Paiva
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Gerson Chadi
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
| |
Collapse
|
2
|
Zhiguo F, Ji W, Shenyuan C, Guoyou Z, Chen K, Hui Q, Wenrong X, Zhai X. A swift expanding trend of extracellular vesicles in spinal cord injury research: a bibliometric analysis. J Nanobiotechnology 2023; 21:289. [PMID: 37612689 PMCID: PMC10463993 DOI: 10.1186/s12951-023-02051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/04/2023] [Indexed: 08/25/2023] Open
Abstract
Extracellular vesicles (EVs) in the field of spinal cord injury (SCI) have garnered significant attention for their potential applications in diagnosis and therapy. However, no bibliometric assessment has been conducted to evaluate the scientific progress in this area. A search of articles in Web of Science (WoS) from January 1, 1991, to May 1, 2023, yielded 359 papers that were analyzed using various online analysis tools. These articles have been cited 10,842 times with 30.2 times per paper. The number of publications experienced explosive growth starting in 2015. China and the United States led this research initiative. Keywords were divided into 3 clusters, including "Pathophysiology of SCI", "Bioactive components of EVs", and "Therapeutic effects of EVs in SCI". By integrating the average appearing year (AAY) of keywords in VoSviewer with the time zone map of the Citation Explosion in CiteSpace, the focal point of research has undergone a transformative shift. The emphasis has moved away from pathophysiological factors such as "axon", "vesicle", and "glial cell" to more mechanistic and applied domains such as "activation", "pathways", "hydrogels" and "therapy". In conclusions, institutions are expected to allocate more resources towards EVs-loaded hydrogel therapy and the utilization of innovative materials for injury mitigation.
Collapse
Affiliation(s)
- Fan Zhiguo
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Wu Ji
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Chen Shenyuan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhang Guoyou
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Kai Chen
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China.
| | - Qian Hui
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Xu Wenrong
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Xiao Zhai
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
3
|
Monsour M, Garbuzova-Davis S, Borlongan CV. Patching Up the Permeability: The Role of Stem Cells in Lessening Neurovascular Damage in Amyotrophic Lateral Sclerosis. Stem Cells Transl Med 2022; 11:1196-1209. [PMID: 36181767 PMCID: PMC9801306 DOI: 10.1093/stcltm/szac072] [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: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating disease with poor prognosis. The pathophysiology of ALS is commonly debated, with theories involving inflammation, glutamate excitotoxity, oxidative stress, mitochondria malfunction, neurofilament accumulation, inadequate nutrients or growth factors, and changes in glial support predominating. These underlying pathological mechanisms, however, act together to weaken the blood brain barrier and blood spinal cord barrier, collectively considered as the blood central nervous system barrier (BCNSB). Altering the impermeability of the BCNSB impairs the neurovascular unit, or interdependent relationship between the brain and advances the concept that ALS is has a significant neurovascular component contributing to its degenerative presentation. This unique categorization of ALS opens a variety of treatment options targeting the reestablishment of BCNSB integrity. This review will critically assess the evidence implicating the significant neurovascular components of ALS pathophysiology, while also offering an in-depth discussion regarding the use of stem cells to repair these pathological changes within the neurovascular unit.
Collapse
Affiliation(s)
- Molly Monsour
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cesario V Borlongan
- Corresponding author: Cesar V. Borlongan, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Boulevard, Tampa, FL 33612, USA.
| |
Collapse
|
4
|
Nam JY, Lee TY, Kim K, Chun S, Kim MS, Shin JH, Sung JJ, Kim BJ, Kim BJ, Oh KW, Kim KS, Kim SH. Efficacy and safety of Lenzumestrocel (Neuronata-R® inj.) in patients with amyotrophic lateral sclerosis (ALSUMMIT study): study protocol for a multicentre, randomized, double-blind, parallel-group, sham procedure-controlled, phase III trial. Trials 2022; 23:415. [PMID: 35585556 PMCID: PMC9115933 DOI: 10.1186/s13063-022-06327-4] [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: 11/21/2021] [Accepted: 04/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A single cycle (two repeated treatments) with intrathecal autologous bone marrow-derived mesenchymal stem cells (BM-MSCs, 26-day interval) showed safety and provided therapeutic benefit lasting 6 months in patients with ALS but did not demonstrate long-term efficacy. This phase III clinical trial (ALSUMMIT) protocol was developed to evaluate the long-term efficacy and safety of the combined protocol of single-cycle intrathecal therapy and three additional booster injections of BM-MSC (Lenzumestrocel) treatment in patients with ALS. METHODS ALSUMMIT is a multicentre, randomized, double-blind, parallel-group, sham procedure-controlled, phase III trial for ALS. The 115 subjects will be randomized (1:2:2) into three groups: (1) study Group 1 (single-cycle, two repeated injections with 26-day interval), (2) study Group 2 (single-cycle + three additional booster injections at 4, 7, and 10 months), and (3) the control group. Participants who have an intermediate rate of disease progression will be included in this trial to reduce clinical heterogeneity. The primary endpoint will be evaluated by combined assessment of function and survival (CAFS), also known as joint rank scores (JRS), at 6 months (study Group 1 vs. control) and 12 months (study Group 2 vs. control) after the first Lenzumestrocel or placebo administration. Safety assessment will be performed throughout the study period. Additionally, after the 56-week main study, a long-term follow-up observational study will be conducted to evaluate the long-term efficacy and safety up to 36 months. DISCUSSION Lenzumestrocel is the orphan cell therapy product for ALS conditionally approved by the South Korea Ministry of Food and Drug Safety (MFDS). This ALSUMMIT protocol was developed for the adoption of enrichment enrolment, add-on design, and consideration of ethical issues for the placebo group. TRIAL REGISTRATION ClinicalTrials.gov NCT04745299 . Registered on Feb 9, 2021. Clinical Research Information Service (CRIS) KCT0005954 . Registered on Mar 4, 2021.
Collapse
Affiliation(s)
- Jae-Yong Nam
- Central Research Center, Corestem Inc, Seoul, South Korea
| | - Tae Yong Lee
- Central Research Center, Corestem Inc, Seoul, South Korea.,College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Kwijoo Kim
- Central Research Center, Corestem Inc, Seoul, South Korea
| | - Sehwan Chun
- Central Research Center, Corestem Inc, Seoul, South Korea
| | - Min Sung Kim
- Central Research Center, Corestem Inc, Seoul, South Korea.,College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Jin-Hong Shin
- Department of Neurology, Pusan National University, Yangsan, South Korea
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University, Seoul, South Korea
| | - Byoung Joon Kim
- Department of Neurology, Samsung Medical Center, Seoul, South Korea
| | - Byung-Jo Kim
- Department of Neurology, Korea University Anam Hospital, Seoul, South Korea
| | - Ki-Wook Oh
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Kyung Suk Kim
- Central Research Center, Corestem Inc, Seoul, South Korea.
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea.
| |
Collapse
|
5
|
Liu B, Li M, Zhang L, Chen Z, Lu P. Motor neuron replacement therapy for amyotrophic lateral sclerosis. Neural Regen Res 2022; 17:1633-1639. [PMID: 35017408 PMCID: PMC8820706 DOI: 10.4103/1673-5374.332123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Amyotrophic lateral sclerosis is a motor neuron degenerative disease that is also known as Lou Gehrig's disease in the United States, Charcot's disease in France, and motor neuron disease in the UK. The loss of motor neurons causes muscle wasting, paralysis, and eventually death, which is commonly related to respiratory failure, within 3-5 years after onset of the disease. Although there are a limited number of drugs approved for amyotrophic lateral sclerosis, they have had little success at treating the associated symptoms, and they cannot reverse the course of motor neuron degeneration. Thus, there is still a lack of effective treatment for this debilitating neurodegenerative disorder. Stem cell therapy for amyotrophic lateral sclerosis is a very attractive strategy for both basic and clinical researchers, particularly as transplanted stem cells and stem cell-derived neural progenitor/precursor cells can protect endogenous motor neurons and directly replace the lost or dying motor neurons. Stem cell therapies may also be able to re-establish the motor control of voluntary muscles. Here, we review the recent progress in the use of neural stem cells and neural progenitor cells for the treatment of amyotrophic lateral sclerosis. We focus on MN progenitor cells derived from fetal central nervous system tissue, embryonic stem cells, and induced pluripotent stem cells. In our recent studies, we found that transplanted human induced pluripotent stem cell-derived motor neuron progenitors survive well, differentiate into motor neurons, and extend axons into the host white matter, not only in the rostrocaudal direction, but also along motor axon tracts towards the ventral roots in the immunodeficient rat spinal cord. Furthermore, the significant motor axonal extension after neural progenitor cell transplantation in amyotrophic lateral sclerosis models demonstrates that motor neuron replacement therapy could be a promising therapeutic strategy for amyotrophic lateral sclerosis, particularly as a variety of stem cell derivatives, including induced pluripotent stem cells, are being considered for clinical trials for various diseases.
Collapse
Affiliation(s)
- Bochao Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education; Center of Neural Injury and Repair; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Mo Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education; Center of Neural Injury and Repair; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Lingyan Zhang
- iXCells Biotechnologies USA, Inc., San Diego, CA, USA; Amogene Biotech, Xiamen, Fujian Province, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education; Center of Neural Injury and Repair; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Paul Lu
- Veterans Administration San Diego Healthcare System, San Diego; Department of Neurosciences, University of California - San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Zhou Q, Yuan M, Qiu W, Cao W, Xu R. Preclinical studies of mesenchymal stem cells transplantation in amyotrophic lateral sclerosis: a systemic review and metaanalysis. Neurol Sci 2021; 42:3637-3646. [PMID: 33433755 DOI: 10.1007/s10072-020-05036-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 12/31/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To assess the quality of preclinical evidence for mesenchymal stromal cell (MSCs) therapy of amyotrophic lateral sclerosis (ALS), decide the effect size of MSCs treatment, and identify clinical parameters that associate with differences in MSCs effects. METHODS A literature search identified studies of MSCs in animal models of ALS. Four main indicators (age of onset, disease progression deceleration, survival time, hazard ratio reduction) obtained through specific neurobehavioral assessment, and 14 relative clinical parameters were extracted for metaanalysis and systematic review. Subgroup analysis and metaregression were performed to explore sources of heterogeneity. RESULTS A total of 25 studies and 41 independent treated arms were used for systematic review and metaanalysis. After adjusted by sensitivity analysis, the mean effect sizes were significantly improved by 0.28 for the age of onset, 0.25 for the disease progression deceleration, 0.54 for the survival time, and 0.48 for hazard ratio reduction. With further analysis, we demonstrated that both the clinical parameter of animal gender and immunosuppressive drug of cyclosporin A (CSA) had a close correlation with disease progression deceleration effect size. CONCLUSIONS These results showed that MSCs transplantation was beneficial for neurobehavioral improvement in the treatment of ALS animal model and recommended that all potential reparative roles of MSCs postdelivery, should be carefully considered and fused to maximize the effectiveness of MSCs therapy in ALS.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, No. 152, Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Min Yuan
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, No. 152, Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Weiwen Qiu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, No. 152, Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Wenfeng Cao
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, No. 152, Aiguo Road, Nanchang, 330006, Jiangxi, China.
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, No. 152, Aiguo Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Wang J, Xu X, Dong W. Correlation Analysis Between Local Cerebral Blood Flow and Severity of Vascular Cognitive Dysfunction. JOURNAL OF MEDICAL IMAGING AND HEALTH INFORMATICS 2021. [DOI: 10.1166/jmihi.2021.3529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Research methods: This paper analyses the correlation between cerebral blood flow perfusion caused by cerebral vascular stenosis and the reduction of patients with cognitive dysfunction and white matter damage. A total of 118 patients with reduced cerebral blood flow perfusion
due to cerebrovascular stenosis were selected to be included in the disease group, and 118 patients with no cerebrovascular stenosis and no neurological disease were included in the control group. The cerebral blood flow perfusion index and cognitive function index were compared between the
two groups of patients. The correlation between each index and the degree of brain white matter damage was analysed. Results: The scores of brain white matter damage in patients with disease group were higher than those in control group, and cCBV, cCBF, TTP, MTT, MoCA, MMSE, ADL, and
WMS were lower than those in control group, and the difference was statistically significant (P < 0.05). cCBV, cCBF, TTP, MTT, and white matter damage scores were highly correlated with MoCA, MMSE, ADL, and WMS (P < 0.05). There is a clear correlation between cerebral vascular
perfusion, cognitive dysfunction, and white matter damage in patients with cerebrovascular stenosis. The more severe the perfusion of cerebral blood flow, the more severe the cognitive dysfunction and the white matter damage.
Collapse
Affiliation(s)
- Jinling Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xin Xu
- Department of Neurology, Daqing Oilfield General Hospital, Zhongkang Street, Daqing, Heilongjiang, 163001, China
| | - Wanhui Dong
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| |
Collapse
|
8
|
Ohashi N, Terashima T, Katagi M, Nakae Y, Okano J, Suzuki Y, Kojima H. GLT1 gene delivery based on bone marrow-derived cells ameliorates motor function and survival in a mouse model of ALS. Sci Rep 2021; 11:12803. [PMID: 34140581 PMCID: PMC8211665 DOI: 10.1038/s41598-021-92285-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an intractable neurodegenerative disease. CD68-positive bone marrow (BM)-derived cells (BMDCs) accumulate in the pathological lesion in the SOD1(G93A) ALS mouse model after BM transplantation (BMT). Therefore, we investigated whether BMDCs can be applied as gene carriers for cell-based gene therapy by employing the accumulation of BMDCs. In ALS mice, YFP reporter signals were observed in 12-14% of white blood cells (WBCs) and in the spinal cord via transplantation of BM after lentiviral vector (LV) infection. After confirmation of gene transduction by LV with the CD68 promoter in 4-7% of WBCs and in the spinal cord of ALS mice, BM cells were infected with LVs expressing glutamate transporter (GLT) 1 that protects neurons from glutamate toxicity, driven by the CD68 promoter, which were transplanted into ALS mice. The treated mice showed improvement of motor behaviors and prolonged survival. Additionally, interleukin (IL)-1β was significantly suppressed, and IL-4, arginase 1, and FIZZ were significantly increased in the mice. These results suggested that GLT1 expression by BMDCs improved the spinal cord environment. Therefore, our gene therapy strategy may be applied to treat neurodegenerative diseases such as ALS in which BMDCs accumulate in the pathological lesion by BMT.
Collapse
Affiliation(s)
- Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Yuki Nakae
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Junko Okano
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Yoshihisa Suzuki
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
9
|
Garbuzova-Davis S, Shell R, Mustafa H, Hailu S, Willing AE, Sanberg PR, Borlongan CV. Advancing Stem Cell Therapy for Repair of Damaged Lung Microvasculature in Amyotrophic Lateral Sclerosis. Cell Transplant 2021; 29:963689720913494. [PMID: 32207340 PMCID: PMC7444221 DOI: 10.1177/0963689720913494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease of motor neuron
degeneration in the brain and spinal cord. Progressive paralysis of
the diaphragm and other respiratory muscles leading to respiratory
dysfunction and failure is the most common cause of death in ALS
patients. Respiratory impairment has also been shown in animal models
of ALS. Vascular pathology is another recently recognized hallmark of
ALS pathogenesis. Central nervous system (CNS) capillary damage is a
shared disease element in ALS rodent models and ALS patients.
Microvascular impairment outside of the CNS, such as in the lungs, may
occur in ALS, triggering lung damage and affecting breathing function.
Stem cell therapy is a promising treatment for ALS. However, this
therapeutic strategy has primarily targeted rescue of degenerated
motor neurons. We showed functional benefits from intravenous delivery
of human bone marrow (hBM) stem cells on restoration of capillary
integrity in the CNS of an superoxide dismutase 1 (SOD1) mouse model
of ALS. Due to the widespread distribution of transplanted cells via
this route, administered cells may enter the lungs and effectively
restore microvasculature in this respiratory organ. Here, we provided
preliminary evidence of the potential role of microvasculature
dysfunction in prompting lung damage and treatment approaches for
repair of respiratory function in ALS. Our initial studies showed
proof-of-principle that microvascular damage in ALS mice results in
lung petechiae at the late stage of disease and that systemic
transplantation of mainly hBM-derived endothelial progenitor cells
shows potential to promote lung restoration via re-established
vascular integrity. Our new understanding of previously underexplored
lung competence in this disease may facilitate therapy targeting
restoration of respiratory function in ALS.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Robert Shell
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hilmi Mustafa
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Surafuale Hailu
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alison E Willing
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Paul R Sanberg
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cesario V Borlongan
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
10
|
Terashima T, Kobashi S, Watanabe Y, Nakanishi M, Honda N, Katagi M, Ohashi N, Kojima H. Enhancing the Therapeutic Efficacy of Bone Marrow-Derived Mononuclear Cells with Growth Factor-Expressing Mesenchymal Stem Cells for ALS in Mice. iScience 2020; 23:101764. [PMID: 33251493 PMCID: PMC7677706 DOI: 10.1016/j.isci.2020.101764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/16/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
Several treatments have been attempted in amyotrophic lateral sclerosis (ALS) animal models and patients. Recently, transplantation of bone marrow-derived mononuclear cells (MNCs) was investigated as a regenerative therapy for ALS, but satisfactory treatments remain to be established. To develop an effective treatment, we focused on mesenchymal stem cells (MSCs) expressing hepatocyte growth factor, glial cell line-derived neurotrophic factor, and insulin-like growth factor using human artificial chromosome vector (HAC-MSCs). Here, we demonstrated the transplantation of MNCs with HAC-MSCs in ALS mice. As per our results, the progression of motor dysfunction was significantly delayed, and their survival was prolonged dramatically. Additional analysis revealed preservation of motor neurons, suppression of gliosis, engraftment of numerous MNCs, and elevated chemotaxis-related cytokines in the spinal cord of treated mice. Therefore, growth factor-expressing MSCs enhance the therapeutic effects of bone marrow-derived MNCs for ALS and have a high potential as a novel cell therapy for patients with ALS. MNCs with growth factor-expressing MSCs is an effective cell therapy for ALS mice The MSCs enhance therapeutic effects by migration of MNCs into ALS mice spinal cord This cell therapy suppresses neuronal loss and gliosis in ALS mice spinal cord This cell therapy induces several cytokines expression in ALS mice spinal cord
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Shuhei Kobashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Mami Nakanishi
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Naoto Honda
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
11
|
Namba F. Mesenchymal stem cells for the prevention of bronchopulmonary dysplasia. Pediatr Int 2019; 61:945-950. [PMID: 31487104 DOI: 10.1111/ped.14001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/06/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in preterm infants who have been treated with supplemental oxygen and mechanical ventilation. Despite major advances in perinatal and neonatal medicine, limited progress has been made in reducing BPD rates. The use of mesenchymal stem cells (MSC) is a promising and innovative therapy for several diseases because they are easy to extract and they have low immunogenicity, anti-inflammatory properties, and regenerative ability. According to several pre-clinical studies that have used BPD animal models, one mechanism of action for MSC in BPD is mainly due to the paracrine effects of MSC-derived humoral factors, such as interleukin (IL)-6, IL-8, vascular endothelial growth factor, collagen, and elastin, rather than the multilineage and regenerative capacities of MSC. Cell-free preparations derived from MSC, including conditioned media and exosomes, remain a pre-clinical technology despite their great clinical potential. A first-in-human clinical trial of MSC treatment for BPD was performed as a phase I dose-escalation trial using umbilical cord blood-derived MSC. That trial demonstrated the short- and long-term safety and feasibility of MSC, given that significantly reduced inflammatory marker expression was observed in tracheal aspirates. As of recently, several clinical trials of MSC use for BPD are ongoing or are planned in some countries to investigate the efficacy of MSC in the prevention or treatment of BPD in premature infants. Many clinicians are currently awaiting the results from these trials so that MSC can be used clinically for human BPD.
Collapse
Affiliation(s)
- Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| |
Collapse
|
12
|
Human intracerebroventricular (ICV) injection of autologous, non-engineered, adipose-derived stromal vascular fraction (ADSVF) for neurodegenerative disorders: results of a 3-year phase 1 study of 113 injections in 31 patients. Mol Biol Rep 2019; 46:5257-5272. [PMID: 31327120 DOI: 10.1007/s11033-019-04983-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
We have chosen to test the safety of human intracerebroventricular (ICV) brain injections of autologous non-genetically-modified adipose-derived stromal vascular fraction (ADSVF). In this IRB-approved trial, 24 patients received ICV ADSVF via an implanted reservoir between 5/22/14 and 5/22/17. Seven others were injected via their ventriculo-peritoneal shunts. Ten patients had Alzheimer's disease (AD), 6 had amyotrophic lateral sclerosis (ALS), 6 had progressive multiple sclerosis (MS-P), 6 had Parkinson's "Plus" (PD+), 1 had spinal cord injury, 1 had traumatic brain injury, and 1 had stroke. Median age was 74 (range 41-83). Injections were planned every 2-3 months. Thirty-one patients had 113 injections. Patients received SVF injection volumes of 3.5-20 cc (median:4 cc) containing 4.05 × 105 to 6.2 × 107 cells/cc, which contained an average of 8% hematopoietic and 7.5% adipose stem cells. Follow-up ranged from 0 to 36 months (median: 9.2 months). MRIs post injection(s) were unchanged, except for one AD patient whose hippocampal volume increased from < 5th percentile to 48th percentile (NeuroQuant® volumetric MRI). Of the 10 AD patients, 8 were stable or improved in tests of cognition. Two showed improvement in P-tau and ß-amyloid levels. Of the 6 MS-P patients all are stable or improved. Four of 6 ALS patients died of disease progression. Twelve of 111 injections (11%) led to 1-4 days of transient meningismus, and mild temperature elevation, which resolved with acetaminophen and/or dexamethasone. Two (1.8% of injections) required hospitalization for these symptoms. One patient (0.9% of injections) had his reservoir removed and later replaced for presumed infection. In this Phase 1 safety trial, ADSVF was safely injected into the human brain ventricular system in patients with no other treatment options. Secondary endpoints of clinical improvement or stability were particularly promising in the AD and MS-P groups. These results will be submitted for a Phase 2 FDA-approved trial.
Collapse
|