1
|
Soumya BS, Gamit N, Patil M, Shreenidhi VP, Dharmarajan A, Warrier S. Modeling amyotrophic lateral sclerosis with amniotic membrane-derived mesenchymal stem cells: A novel approach for disease modeling. Exp Cell Res 2025; 446:114449. [PMID: 39961464 DOI: 10.1016/j.yexcr.2025.114449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/16/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Advancement of therapeutics for neurodegenerative diseases like amyotrophic lateral sclerosis (ALS) has been predominantly hampered by the dearth of relevant disease models. Despite numerous animal models, significant challenges remain in correlating these with human disease complexities. In this study, the ALS model was created using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) which were differentiated into motor neurons (MN) with specific MN induction media and transiently transfected with mutated human SOD1 G93A plasmid to induce ALS-like condition. Characterization included gene expression analysis, immunocytochemistry, flow cytometry, and Western blot. Functional assays assessed the extent of degeneration and model efficiency. AM-MSCs demonstrated multipotency and were positive for MSC markers. Upon differentiation, the expression of MN markers like MNX1, Olig2, and ChAT were found to be elevated. SOD1 G93A overexpression, downregulated MN markers, upregulated NURR1 gene, reduced acetylcholine (ACh), reduced glutathione, and elevated oxidative stress markers. This robust in-vitro ALS model derived from AM-MSCs offers an alternative to animal models to provide an efficient and cost-effective platform to conduct rapid drug screening.
Collapse
Affiliation(s)
- B S Soumya
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - V P Shreenidhi
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Arun Dharmarajan
- School of Human Sciences, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, WA 6009, Australia; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India; Division of Regenerative Medicine and Cancer Stem Cells, Department of Biotechnology, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, 600116, India.
| |
Collapse
|
2
|
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, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia 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
|
3
|
Mazzini L, De Marchi F, Buzanska L, Follenzi A, Glover JC, Gelati M, Lombardi I, Maioli M, Mesa-Herrera F, Mitrečić D, Olgasi C, Pivoriūnas A, Sanchez-Pernaute R, Sgromo C, Zychowicz M, Vescovi A, Ferrari D. Current status and new avenues of stem cell-based preclinical and therapeutic approaches in amyotrophic lateral sclerosis. Expert Opin Biol Ther 2024; 24:933-954. [PMID: 39162129 DOI: 10.1080/14712598.2024.2392307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Cell therapy development represents a critical challenge in amyotrophic lateral sclerosis (ALS) research. Despite more than 20 years of basic and clinical research, no definitive safety and efficacy results of cell-based therapies for ALS have been published. AREAS COVERED This review summarizes advances using stem cells (SCs) in pre-clinical studies to promote clinical translation and in clinical trials to treat ALS. New technologies have been developed and new experimental in vitro and animal models are now available to facilitate pre-clinical research in this field and to determine the most promising approaches to pursue in patients. New clinical trial designs aimed at developing personalized SC-based treatment with biological endpoints are being defined. EXPERT OPINION Knowledge of the basic biology of ALS and on the use of SCs to study and potentially treat ALS continues to grow. However, a consensus has yet to emerge on how best to translate these results into therapeutic applications. The selection and follow-up of patients should be based on clinical, biological, and molecular criteria. Planning of SC-based clinical trials should be coordinated with patient profiling genetically and molecularly to achieve personalized treatment. Much work within basic and clinical research is still needed to successfully transition SC therapy in ALS.
Collapse
Affiliation(s)
- Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Antonia Follenzi
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
- Dipartimento Attività Integrate Ricerca Innovazione, Azienda Ospedaliero-Universitaria SS. Antonio e Biagio e C. Arrigo, Alessandria, Italy
| | - Joel Clinton Glover
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital; Laboratory of Neural Development and Optical Recording (NDEVOR), Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maurizio Gelati
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ivan Lombardi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Center for Developmental Biology and Reprogramming-CEDEBIOR, University of Sassari, Sassari, Italy
| | - Fatima Mesa-Herrera
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research and Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Cristina Olgasi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Rosario Sanchez-Pernaute
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
- Ikerbaske, Basque Foundation for Science, Bilbao, Spain
| | - Chiara Sgromo
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Angelo Vescovi
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
4
|
Bruno A, Milillo C, Anaclerio F, Buccolini C, Dell’Elice A, Angilletta I, Gatta M, Ballerini P, Antonucci I. Perinatal Tissue-Derived Stem Cells: An Emerging Therapeutic Strategy for Challenging Neurodegenerative Diseases. Int J Mol Sci 2024; 25:976. [PMID: 38256050 PMCID: PMC10815412 DOI: 10.3390/ijms25020976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Over the past 20 years, stem cell therapy has been considered a promising option for treating numerous disorders, in particular, neurodegenerative disorders. Stem cells exert neuroprotective and neurodegenerative benefits through different mechanisms, such as the secretion of neurotrophic factors, cell replacement, the activation of endogenous stem cells, and decreased neuroinflammation. Several sources of stem cells have been proposed for transplantation and the restoration of damaged tissue. Over recent decades, intensive research has focused on gestational stem cells considered a novel resource for cell transplantation therapy. The present review provides an update on the recent preclinical/clinical applications of gestational stem cells for the treatment of protein-misfolding diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). However, further studies should be encouraged to translate this promising therapeutic approach into the clinical setting.
Collapse
Affiliation(s)
- Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Cristina Milillo
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Federico Anaclerio
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlotta Buccolini
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Anastasia Dell’Elice
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ilaria Angilletta
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Gatta
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ivana Antonucci
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
5
|
Kim SG, George NP, Hwang JS, Park S, Kim MO, Lee SH, Lee G. Human Bone Marrow-Derived Mesenchymal Stem Cell Applications in Neurodegenerative Disease Treatment and Integrated Omics Analysis for Successful Stem Cell Therapy. Bioengineering (Basel) 2023; 10:bioengineering10050621. [PMID: 37237691 DOI: 10.3390/bioengineering10050621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Neurodegenerative diseases (NDDs), which are chronic and progressive diseases, are a growing health concern. Among the therapeutic methods, stem-cell-based therapy is an attractive approach to NDD treatment owing to stem cells' characteristics such as their angiogenic ability, anti-inflammatory, paracrine, and anti-apoptotic effects, and homing ability to the damaged brain region. Human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) are attractive NDD therapeutic agents owing to their widespread availability, easy attainability and in vitro manipulation and the lack of ethical issues. Ex vivo hBM-MSC expansion before transplantation is essential because of the low cell numbers in bone marrow aspirates. However, hBM-MSC quality decreases over time after detachment from culture dishes, and the ability of hBM-MSCs to differentiate after detachment from culture dishes remains poorly understood. Conventional analysis of hBM-MSCs characteristics before transplantation into the brain has several limitations. However, omics analyses provide more comprehensive molecular profiling of multifactorial biological systems. Omics and machine learning approaches can handle big data and provide more detailed characterization of hBM-MSCs. Here, we provide a brief review on the application of hBM-MSCs in the treatment of NDDs and an overview of integrated omics analysis of the quality and differentiation ability of hBM-MSCs detached from culture dishes for successful stem cell therapy.
Collapse
Affiliation(s)
- Seok Gi Kim
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Nimisha Pradeep George
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Ji Su Hwang
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Seokho Park
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Biomedical Science, Graduate School of Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Soo Hwan Lee
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Biomedical Science, Graduate School of Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
| |
Collapse
|
6
|
Khamaysa M, Pradat PF. Status of ALS Treatment, Insights into Therapeutic Challenges and Dilemmas. J Pers Med 2022; 12:1601. [PMID: 36294741 PMCID: PMC9605458 DOI: 10.3390/jpm12101601] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/14/2022] [Accepted: 09/23/2022] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an extremely heterogeneous disease of motor neurons that eventually leads to death. Despite impressive advances in understanding the genetic, molecular, and pathological mechanisms of the disease, the only drug approved to date by both the FDA and EMA is riluzole, with a modest effect on survival. In this opinion view paper, we will discuss how to address some challenges for drug development in ALS at the conceptual, technological, and methodological levels. In addition, socioeconomic and ethical issues related to the legitimate need of patients to benefit quickly from new treatments will also be addressed. In conclusion, this brief review takes a more optimistic view, given the recent approval of two new drugs in some countries and the development of targeted gene therapies.
Collapse
Affiliation(s)
- Mohammed Khamaysa
- Laboratoire d’Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75006 Paris, France
| | - Pierre-François Pradat
- Laboratoire d’Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75006 Paris, France
- Centre Référent SLA, Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, C-TRIC, Altnagelvin Hospital, Derry-Londonderry BT47 6SB, UK
| |
Collapse
|
7
|
Johnson SA, Fang T, De Marchi F, Neel D, Van Weehaeghe D, Berry JD, Paganoni S. Pharmacotherapy for Amyotrophic Lateral Sclerosis: A Review of Approved and Upcoming Agents. Drugs 2022; 82:1367-1388. [PMID: 36121612 DOI: 10.1007/s40265-022-01769-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2022] [Indexed: 11/03/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder involving loss of upper and lower motor neurons, with most cases ending in death within 3-5 years of onset. Several molecular and cellular pathways have been identified to cause ALS; however, treatments to stop or reverse disease progression are yet to be found. Riluzole, a neuroprotective agent offering only a modest survival benefit, has long been the sole disease-modifying therapy for ALS. Edaravone, which demonstrated statistically significant slowing of ALS disease progression, is gaining approval in an increasing number of countries since its first approval in 2015. Sodium phenylbutyrate and taurursodiol (PB-TURSO) was conditionally approved in Canada in 2022, having shown significant slowing of disease progression and prolonged survival. Most clinical trials have focused on testing small molecules affecting common cellular pathways in ALS: targeting glutamatergic, apoptotic, inflammatory, and oxidative stress mechanisms among others. More recently, clinical trials utilizing stem cell transplantation and other biologics have emerged. This rich and ever-growing pipeline of investigational products, along with innovative clinical trial designs, collaborative trial networks, and an engaged ALS community', provide renewed hope to finding a cure for ALS. This article reviews existing ALS therapies and the current clinical drug development pipeline.
Collapse
Affiliation(s)
- Stephen A Johnson
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA
| | - Ton Fang
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, Maggiore della Carità Hospital, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100, Novara, Italy
| | | | - Donatienne Van Weehaeghe
- Nuclear Medicine Subdivision, Department of Imaging and Pathology, University Hospital Leuven, Leuven, Belgium
| | - James D Berry
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA
| | - Sabrina Paganoni
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA.
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
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
|
9
|
Lin TJ, Cheng KC, Wu LY, Lai WY, Ling TY, Kuo YC, Huang YH. Potential of Cellular Therapy for ALS: Current Strategies and Future Prospects. Front Cell Dev Biol 2022; 10:851613. [PMID: 35372346 PMCID: PMC8966507 DOI: 10.3389/fcell.2022.851613] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive upper and lower motor neuron (MN) degeneration with unclear pathology. The worldwide prevalence of ALS is approximately 4.42 per 100,000 populations, and death occurs within 3-5 years after diagnosis. However, no effective therapeutic modality for ALS is currently available. In recent years, cellular therapy has shown considerable therapeutic potential because it exerts immunomodulatory effects and protects the MN circuit. However, the safety and efficacy of cellular therapy in ALS are still under debate. In this review, we summarize the current progress in cellular therapy for ALS. The underlying mechanism, current clinical trials, and the pros and cons of cellular therapy using different types of cell are discussed. In addition, clinical studies of mesenchymal stem cells (MSCs) in ALS are highlighted. The summarized findings of this review can facilitate the future clinical application of precision medicine using cellular therapy in ALS.
Collapse
Affiliation(s)
- Ting-Jung Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuang-Chao Cheng
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Luo-Yun Wu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Yu Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Parambi DGT, Alharbi KS, Kumar R, Harilal S, Batiha GES, Cruz-Martins N, Magdy O, Musa A, Panda DS, Mathew B. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:191-233. [PMID: 34655056 PMCID: PMC8518903 DOI: 10.1007/s12035-021-02555-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022]
Abstract
The etiology of many neurological diseases affecting the central nervous system (CNS) is unknown and still needs more effective and specific therapeutic approaches. Gene therapy has a promising future in treating neurodegenerative disorders by correcting the genetic defects or by therapeutic protein delivery and is now an attraction for neurologists to treat brain disorders, like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, spinocerebellar ataxia, epilepsy, Huntington's disease, stroke, and spinal cord injury. Gene therapy allows the transgene induction, with a unique expression in cells' substrate. This article mainly focuses on the delivering modes of genetic materials in the CNS, which includes viral and non-viral vectors and their application in gene therapy. Despite the many clinical trials conducted so far, data have shown disappointing outcomes. The efforts done to improve outcomes, efficacy, and safety in the identification of targets in various neurological disorders are also discussed here. Adapting gene therapy as a new therapeutic approach for treating neurological disorders seems to be promising, with early detection and delivery of therapy before the neuron is lost, helping a lot the development of new therapeutic options to translate to the clinic.
Collapse
Affiliation(s)
- Della Grace Thomas Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Khalid Saad Alharbi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Omnia Magdy
- Department of Clinical Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014 Kingdom of Saudi Arabia
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
| | - Arafa Musa
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371 Egypt
| | - Dibya Sundar Panda
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Al Jouf, Sakaka, 72341 Kingdom of Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
11
|
Liu E, Karpf L, Bohl D. Neuroinflammation in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia and the Interest of Induced Pluripotent Stem Cells to Study Immune Cells Interactions With Neurons. Front Mol Neurosci 2022; 14:767041. [PMID: 34970118 PMCID: PMC8712677 DOI: 10.3389/fnmol.2021.767041] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a shared hallmark between amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). For long, studies were conducted on tissues of post-mortem patients and neuroinflammation was thought to be only bystander result of the disease with the immune system reacting to dying neurons. In the last two decades, thanks to improving technologies, the identification of causal genes and the development of new tools and models, the involvement of inflammation has emerged as a potential driver of the diseases and evolved as a new area of intense research. In this review, we present the current knowledge about neuroinflammation in ALS, ALS-FTD, and FTD patients and animal models and we discuss reasons of failures linked to therapeutic trials with immunomodulator drugs. Then we present the induced pluripotent stem cell (iPSC) technology and its interest as a new tool to have a better immunopathological comprehension of both diseases in a human context. The iPSC technology giving the unique opportunity to study cells across differentiation and maturation times, brings the hope to shed light on the different mechanisms linking neurodegeneration and activation of the immune system. Protocols available to differentiate iPSC into different immune cell types are presented. Finally, we discuss the interest in studying monocultures of iPS-derived immune cells, co-cultures with neurons and 3D cultures with different cell types, as more integrated cellular approaches. The hope is that the future work with human iPS-derived cells helps not only to identify disease-specific defects in the different cell types but also to decipher the synergistic effects between neurons and immune cells. These new cellular tools could help to find new therapeutic approaches for all patients with ALS, ALS-FTD, and FTD.
Collapse
Affiliation(s)
- Elise Liu
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Léa Karpf
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Delphine Bohl
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
12
|
Kim HJ, Cho KR, Jang H, Lee NK, Jung YH, Kim JP, Lee JI, Chang JW, Park S, Kim ST, Moon SW, Seo SW, Choi SJ, Na DL. Intracerebroventricular injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer's disease dementia: a phase I clinical trial. ALZHEIMERS RESEARCH & THERAPY 2021; 13:154. [PMID: 34521461 PMCID: PMC8439008 DOI: 10.1186/s13195-021-00897-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/02/2021] [Indexed: 12/22/2022]
Abstract
Backgrounds Alzheimer’s disease is the most common cause of dementia, and currently, there is no disease-modifying treatment. Favorable functional outcomes and reduction of amyloid levels were observed following transplantation of mesenchymal stem cells (MSCs) in animal studies. Objectives We conducted a phase I clinical trial in nine patients with mild-to-moderate Alzheimer’s disease dementia to evaluate the safety and dose-limiting toxicity of three repeated intracerebroventricular injections of human umbilical cord blood–derived MSCs (hUCB-MSCs). Methods We recruited nine mild-to-moderate Alzheimer’s disease dementia patients from Samsung Medical Center, Seoul, Republic of Korea. Four weeks prior to MSC administration, the Ommaya reservoir was implanted into the right lateral ventricle of the patients. Three patients received a low dose (1.0 × 107 cells/2 mL), and six patients received a high dose (3.0 × 107 cells/2 mL) of hUCB-MSCs. Three repeated injections of MSCs were performed (4-week intervals) in all nine patients. These patients were followed up to 12 weeks after the first hUCB-MSC injection and an additional 36 months in the extended observation study. Results After hUCB-MSC injection, the most common adverse event was fever (n = 9) followed by headache (n = 7), nausea (n = 5), and vomiting (n = 4), which all subsided within 36 h. There were three serious adverse events in two participants that were considered to have arisen from the investigational product. Fever in a low dose participant and nausea with vomiting in another low dose participant each required extended hospitalization by a day. There were no dose-limiting toxicities. Five participants completed the 36-month extended observation study, and no further serious adverse events were observed. Conclusions Three repeated administrations of hUCB-MSCs into the lateral ventricle via an Ommaya reservoir were feasible, relatively and sufficiently safe, and well-tolerated. Currently, we are undergoing an extended follow-up study for those who participated in a phase IIa trial where upon completion, we hope to gain a deeper understanding of the clinical efficacy of MSC AD therapy. Trial registration ClinicalTrials.gov NCT02054208. Registered on 4 February 2014. ClinicalTrials.gov NCT03172117. Registered on 1 June 2017 Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00897-2.
Collapse
Affiliation(s)
- Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 80 Ilwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyung Rae Cho
- Department of Neurosurgery, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 80 Ilwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Na Kyung Lee
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Hee Jung
- Department of Neurology, Myongji Hospital, Hanyang University, Goyang, Republic of Korea
| | - Jun Pyo Kim
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jung Il Lee
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Seongbeom Park
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 80 Ilwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sung Tae Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Whan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 80 Ilwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam, Republic of Korea
| | - Duk L Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 80 Ilwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea. .,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea. .,Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea. .,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Tavakol-Afshari J, Boroumand AR, Farkhad NK, Adhami Moghadam A, Sahab-Negah S, Gorji A. Safety and efficacy of bone marrow derived-mesenchymal stem cells transplantation in patients with amyotrophic lateral sclerosis. Regen Ther 2021; 18:268-274. [PMID: 34466632 PMCID: PMC8377537 DOI: 10.1016/j.reth.2021.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/11/2021] [Accepted: 07/29/2021] [Indexed: 01/22/2023] Open
Abstract
Stem cell-based treatments have emerged as potentially effective approaches to delay the progression of amyotrophic lateral sclerosis (ALS). This study was designed as a single-center, prospective, and open-label study without a placebo control group to assess the safety and efficacy of concurrent intrathecal (IT) and intravenous (IV) administration of autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) in patients with ALS. Autologous BM-MSCs were isolated and expanded under standard conditions. Fifteen patients were neurologically examined before BM-MSCs transplantation (1 × 10 6 cells/kg BW) to evaluate the rate of pre-treatment disease progression. To assess the safety and efficacy, patients were examined at 1, 3, and 6 months following the treatment with BM-MSCs. Adverse reactions were assessed, and the clinical outcome was determined by the evaluation of the ALS functional rating scale-revised (ALSFRS-R) and forced vital capacity (FVC). No serious adverse reaction was observed after combined IT and IV administration of BM-MSCs. The mean ALSFRS-R and FVC values remained stable during the first 3 months of the treatment. However, a significant reduction in ALSFRS-R and FVC levels was observed in these patients 6 months after BM-MSCs administration. Our study revealed that the concurrent IT and IV application of BM-MSCs in patients with ALS is a safe procedure. Furthermore, our data indicate a temporary delay in the progression of ALS after a single combined IT and IV administration of BM-MSCs. Further studies are required to explore if the repeated applications of BM-MSCs could prolong survival and delay the progression of ALS.
Collapse
Affiliation(s)
| | - Amir Reza Boroumand
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najmeh Kaffash Farkhad
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Adhami Moghadam
- Department of Internal Medicine and Critical Care, Islamic Azad University, Mashhad, Iran
- Specialty of Internal Medicine and Critical Care, Head of Army Hospital ICU and Intensive, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Corresponding author. Neuroscience Research Center, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Corresponding author. Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, D-48149, Germany.
| |
Collapse
|
14
|
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
|
15
|
Huang J, Xian B, Peng Y, Zeng B, Li W, Li Z, Xie Y, Zhao M, Zhang H, Zhou M, Yu H, Wu P, Liu X, Huang B. Migration of pre-induced human peripheral blood mononuclear cells from the transplanted to contralateral eye in mice. Stem Cell Res Ther 2021; 12:168. [PMID: 33691753 PMCID: PMC7945672 DOI: 10.1186/s13287-021-02180-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/20/2021] [Indexed: 11/26/2022] Open
Abstract
Background Retina diseases may lead to blindness as they often afflict both eyes. Stem cell transplantation into the affected eye(s) is a promising therapeutic strategy for certain retinal diseases. Human peripheral blood mononuclear cells (hPBMCs) are a good source of stem cells, but it is unclear whether pre-induced hPBMCs can migrate from the injected eye to the contralateral eye for bilateral treatment. We examine the possibility of bilateral cell transplantation from unilateral cell injection. Methods One hundred and sixty-one 3-month-old retinal degeneration 1 (rd1) mice were divided randomly into 3 groups: an untreated group (n = 45), a control group receiving serum-free Dulbecco’s modified Eagle’s medium (DMEM) injection into the right subretina (n = 45), and a treatment group receiving injection of pre-induced hPBMCs into the right subretina (n = 71). Both eyes were examined by full-field electroretinogram (ERG), immunofluorescence, flow cytometry, and quantitative real-time polymerase chain reaction (qRT-PCR) at 1 and 3 months post-injection. Results At both 1 and 3 months post-injection, labeled pre-induced hPBMCs were observed in the retinal inner nuclear layer of the contralateral (left untreated) eye as well as the treated eye as evidenced by immunofluorescence staining for a human antigen. Flow cytometry of fluorescently label cells and qRT-PCR of hPBMCs genes confirmed that transplanted hPBMCs migrated from the treated to the contralateral untreated eye and remained viable for up to 3 months. Further, full-field ERG showed clear light-evoked a and b waves in both treated and untreated eyes at 3 months post-transplantation. Labeled pre-induced hPBMCs were also observed in the contralateral optic nerve but not in the blood circulation, suggesting migration via the optic chiasm. Conclusion It may be possible to treat binocular eye diseases by unilateral stem cell injection. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02180-5.
Collapse
Affiliation(s)
- Jianfa Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Bikun Xian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.,The Second People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Yuting Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.,Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Baozhu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhiquan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yaojue Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Hening Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Minyi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Huan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Peixin Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Bing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
16
|
Giunti D, Marini C, Parodi B, Usai C, Milanese M, Bonanno G, Kerlero de Rosbo N, Uccelli A. Role of miRNAs shuttled by mesenchymal stem cell-derived small extracellular vesicles in modulating neuroinflammation. Sci Rep 2021; 11:1740. [PMID: 33462263 PMCID: PMC7814007 DOI: 10.1038/s41598-021-81039-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are characterized by neuroprotective, immunomodulatory, and neuroregenerative properties, which support their therapeutic potential for inflammatory/neurodegenerative diseases, including multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). One mode of action through which MSCs exert their immunomodulatory effects is release of extracellular vesicles that carry proteins, mRNAs, and microRNAs (miRNAs), which, once transferred, modify the function of target cells. We identified nine miRNAs significantly dysregulated in IFN-γ-primed MSCs, but present at different levels in their derived small extracellular vesicles (s-EV). We show that miR-467f and miR-466q modulate the pro-inflammatory phenotype of activated N9 microglia cells and of primary microglia acutely isolated from late symptomatic SOD1G93A mice, a murine ALS model, by downregulating Tnf and Il1b expression. Further analysis of the mode of action of miR-467f and miR-466q indicated that they dampen the pro-inflammatory phenotype of microglia by modulating p38 MAPK signaling pathway via inhibition of expression of their target genes, Map3k8 and Mk2. Finally, we demonstrated that in vivo administration of s-EV leads to decreased expression of neuroinflammation markers in the spinal cord of EAE-affected mice, albeit without affecting disease course. Overall, our data suggest that MSC-derived exosomes could affect neuroinflammation possibly through specific immunomodulatory miRNAs acting on microglia.
Collapse
Affiliation(s)
- Debora Giunti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Marini
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Benedetta Parodi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council (CNR), Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Giambattista Bonanno
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Pharmacy (DIFAR), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Nicole Kerlero de Rosbo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.
| |
Collapse
|
17
|
Studying ALS: Current Approaches, Effect on Potential Treatment Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1241:195-217. [PMID: 32383122 DOI: 10.1007/978-3-030-41283-8_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is one of the most common neurodegenerative diseases, characterized by inevitable progressive paralysis. To date, only two disease modifying therapeutic options are available for the patients with ALS, although they show very modest effect on disease course. The main reason of failure in the field of pharmacological correction of ALS is inability to untangle complex relationships taking place during ALS initiation and progression. Traditional methods of research, based on morphology or transgenic animal models studying provided lots of information about ALS throughout the years. However, translation of these results to humans was unsuccessful due to incomplete recapitulation of molecular pathology and overall inadequacy of the models used in the research.In this review we summarize current knowledge regarding ALS molecular pathology with depiction of novel methods applied recently for the studies. Furthermore we describe present and potential treatment strategies that are based on the recent findings in ALS disease mechanisms.
Collapse
|
18
|
Hernández R, Jiménez-Luna C, Perales-Adán J, Perazzoli G, Melguizo C, Prados J. Differentiation of Human Mesenchymal Stem Cells towards Neuronal Lineage: Clinical Trials in Nervous System Disorders. Biomol Ther (Seoul) 2020; 28:34-44. [PMID: 31649208 PMCID: PMC6939692 DOI: 10.4062/biomolther.2019.065] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been proposed as an alternative therapy to be applied into several pathologies of the nervous system. These cells can be obtained from adipose tissue, umbilical cord blood and bone marrow, among other tissues, and have remarkable therapeutic properties. MSCs can be isolated with high yield, which adds to their ability to differentiate into non-mesodermal cell types including neuronal lineage both in vivo and in vitro. They are able to restore damaged neural tissue, thus being suitable for the treatment of neural injuries, and possess immunosuppressive activity, which may be useful for the treatment of neurological disorders of inflammatory etiology. Although the long-term safety of MSC-based therapies remains unclear, a large amount of both pre-clinical and clinical trials have shown functional improvements in animal models of nervous system diseases following transplantation of MSCs. In fact, there are several ongoing clinical trials evaluating the possible benefits this cell-based therapy could provide to patients with neurological damage, as well as their clinical limitations. In this review we focus on the potential of MSCs as a therapeutic tool to treat neurological disorders, summarizing the state of the art of this topic and the most recent clinical studies.
Collapse
Affiliation(s)
- Rosa Hernández
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - Cristina Jiménez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland
| | - Jesús Perales-Adán
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| |
Collapse
|
19
|
Vieira S, Strymecka P, Stanaszek L, Silva-Correia J, Drela K, Fiedorowicz M, Malysz-Cymborska I, Rogujski P, Janowski M, Reis RL, Lukomska B, Walczak P, Oliveira JM. Methacrylated gellan gum and hyaluronic acid hydrogel blends for image-guided neurointerventions. J Mater Chem B 2020; 8:5928-5937. [DOI: 10.1039/d0tb00877j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mn-Based gellan gum hydrogels for cell delivery and real-time tracking on image-guided neuro-procedures.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Paulina Strymecka
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Luiza Stanaszek
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Katarzyna Drela
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Izabela Malysz-Cymborska
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - Piotr Rogujski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Miroslaw Janowski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Barbara Lukomska
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Piotr Walczak
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - J. Miguel Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| |
Collapse
|
20
|
Watanabe TK. A Review of Stem Cell Therapy for Acquired Brain Injuries and Neurodegenerative Central Nervous System Diseases. PM R 2019; 10:S151-S156. [PMID: 30269801 DOI: 10.1016/j.pmrj.2018.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been the subject of much discussion regarding their potential role in enhancing central nervous system function for a number of pathologic conditions. Much of the current research has been in preclinical trials, with clinical trials in the phase I or I/II stage. Nevertheless, there is considerable interest in the public about the potential regenerative role that stem cells may have in improving function for these neurologic conditions. This review will describe the different types of stem cells that are available, review their possible effects, and discuss some of the variables that investigators need to consider when designing their studies. Current clinical research in the areas of stroke, traumatic brain injury, and neurodegenerative diseases (amyotrophic lateral sclerosis and Parkinson disease) will be reviewed. As this article is aimed at a rehabilitation audience, outcome measures, and the role of concurrent rehabilitation therapies will also be mentioned.
Collapse
Affiliation(s)
- Thomas K Watanabe
- MossRehab at Elkins Park / Einstein Healthcare Network, 60 Township Line Road, Elkins Park, PA 19027(∗).
| |
Collapse
|
21
|
The Anti-Apoptotic Effect of ASC-Exosomes in an In Vitro ALS Model and Their Proteomic Analysis. Cells 2019; 8:cells8091087. [PMID: 31540100 PMCID: PMC6770878 DOI: 10.3390/cells8091087] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/17/2022] Open
Abstract
Stem cell therapy represents a promising approach in the treatment of several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). The beneficial effect of stem cells is exerted by paracrine mediators, as exosomes, suggesting a possible potential use of these extracellular vesicles as non-cell based therapy. We demonstrated that exosomes isolated from adipose stem cells (ASC) display a neuroprotective role in an in vitro model of ALS. Moreover, the internalization of ASC-exosomes by the cells was shown and the molecules and the mechanisms by which exosomes could exert their beneficial effect were addressed. We performed for the first time a comprehensive proteomic analysis of exosomes derived from murine ASC. We identified a total of 189 proteins and the shotgun proteomics analysis revealed that the exosomal proteins are mainly involved in cell adhesion and negative regulation of the apoptotic process. We correlated the protein content to the anti-apoptotic effect of exosomes observing a downregulation of pro-apoptotic proteins Bax and cleaved caspase-3 and upregulation of anti-apoptotic protein Bcl-2 α, in an in vitro model of ALS after cell treatment with exosomes. Overall, this study shows the neuroprotective effect of ASC-exosomes after their internalization and their global protein profile, that could be useful to understand how exosomes act, demonstrating that they can be employed as therapy in neurodegenerative diseases.
Collapse
|
22
|
Gouel F, Rolland AS, Devedjian JC, Burnouf T, Devos D. Past and Future of Neurotrophic Growth Factors Therapies in ALS: From Single Neurotrophic Growth Factor to Stem Cells and Human Platelet Lysates. Front Neurol 2019; 10:835. [PMID: 31428042 PMCID: PMC6688198 DOI: 10.3389/fneur.2019.00835] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that typically results in death within 3–5 years after diagnosis. To date, there is no curative treatment and therefore an urgent unmet need of neuroprotective and/or neurorestorative treatments. Due to their spectrum of capacities in the central nervous system—e.g., development, plasticity, maintenance, neurogenesis—neurotrophic growth factors (NTF) have been exploited for therapeutic strategies in ALS for decades. In this review we present the initial strategy of using single NTF by different routes of administration to the use of stem cells transplantation to express a multiple NTFs-rich secretome to finally focus on a new biotherapy based on the human platelet lysates, the natural healing system containing a mix of pleitropic NTF and having immunomodulatory function. This review highlights that this latter treatment may be crucial to power the neuroprotection and/or neurorestoration therapy requested in this devastating disease.
Collapse
Affiliation(s)
- Flore Gouel
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1171, University Hospital Center, LICEND COEN Center, Lille, France
| | - Anne-Sophie Rolland
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1171, University Hospital Center, LICEND COEN Center, Lille, France
| | - Jean-Christophe Devedjian
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1171, University Hospital Center, LICEND COEN Center, Lille, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - David Devos
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1171, University Hospital Center, LICEND COEN Center, Lille, France.,Department of Neurology, Lille University, INSERM UMRS_1171, University Hospital Center, LICEND COEN Center, Lille, France
| |
Collapse
|
23
|
Rajangam T, Moon KS, Kim D, Kang J, Lee S, Oh SJ, Kim SH. Therapeutic Effect of a Xeno-Free Three-Dimensional Stem Cell Mass in a Hind Limb Ischemia Model. Tissue Eng Part A 2019; 25:314-332. [DOI: 10.1089/ten.tea.2018.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Thanavel Rajangam
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Kyoung-Sik Moon
- Korea Institute of Toxicology, Daejeon, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejon, Republic of Korea
| | - Dokyun Kim
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Jungmi Kang
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Sunyeong Lee
- Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Seung Ja Oh
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| |
Collapse
|
24
|
Gubert F, Bonacossa-Pereira I, Decotelli AB, Furtado M, Vasconcelos-Dos-Santos A, Mendez-Otero R, Santiago MF. Bone-marrow mononuclear cell therapy in a mouse model of amyotrophic lateral sclerosis: Functional outcomes from different administration routes. Brain Res 2019; 1712:73-81. [PMID: 30735638 DOI: 10.1016/j.brainres.2019.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic degenerative disease that mainly affects motor neurons, leading to progressive paralysis and death. Recently, cell therapy has emerged as a therapeutic alternative for several neurological diseases, including ALS, and bone-marrow cells are one of the major cell sources. Considering the importance of pre-clinical trials to determine the best therapeutic protocol and the hope of translating this protocol to the clinical setting, we tested bone-marrow mononuclear cell (BMMC) therapy administered by different routes in the SOD1G93A model of ALS. BMMCs were isolated from non-transgenic, age matched animals and administered intravenously (IV), intramuscularly (IM), and intravenously and intramuscular concomitantly (IV + IM). BMMC therapy had no significant beneficial effects when injected IV or IM, but delayed disease progression when these two routes were used concomitantly. BMMC IV + IM treatment reduced the number of microglia cells in the spinal cord and partially protected of neuromuscular-junction innervation, but had no effect in preventing motor-neuron loss. This study showed that injection of BMMC IV + IM had better results when compared to each route in isolation, highlighting the importance of targeting multiple anatomical regions in the treatment of ALS.
Collapse
Affiliation(s)
- Fernanda Gubert
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Igor Bonacossa-Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Ana B Decotelli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Michelle Furtado
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcelo F Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
25
|
Rando A, Pastor D, Viso-León MC, Martínez A, Manzano R, Navarro X, Osta R, Martínez S. Intramuscular transplantation of bone marrow cells prolongs the lifespan of SOD1 G93A mice and modulates expression of prognosis biomarkers of the disease. Stem Cell Res Ther 2018; 9:90. [PMID: 29625589 PMCID: PMC5889612 DOI: 10.1186/s13287-018-0843-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by progressive muscle weakness, paralysis and death. There is no effective treatment for ALS and stem cell therapy has arisen as a potential therapeutic approach. METHODS SOD1 mutant mice were used to study the potential neurotrophic effect of bone marrow cells grafted into quadriceps femoris muscle. RESULTS Bone marrow intramuscular transplants resulted in increased longevity with improved motor function and decreased motoneuron degeneration in the spinal cord. Moreover, the increment of the glial-derived neurotrophic factor and neurotrophin 4 observed in the grafted muscles suggests that this partial neuroprotective effect is mediated by neurotrophic factor release at the neuromuscular junction level. Finally, certain neurodegeneration and muscle disease-specific markers, which are altered in the SOD1G93A mutant mouse and may serve as molecular biomarkers for the early detection of ALS in patients, have been studied with encouraging results. CONCLUSIONS This work demonstrates that stem cell transplantation in the muscle prolonged the lifespan, increased motoneuron survival and slowed disease progression, which was also assessed by genetic expression analysis.
Collapse
Affiliation(s)
- Amaya Rando
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Diego Pastor
- Centro de Investigación Deporte, Universidad Miguel Hernández de Elche, Alicante, Spain
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Mari Carmen Viso-León
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Anna Martínez
- Grupo de Neuroplasticidad y Regeneración, Instituto de Neurociencias y Departamento de Biología Celular, Fisiología e Inmunología, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Raquel Manzano
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Xavier Navarro
- Grupo de Neuroplasticidad y Regeneración, Instituto de Neurociencias y Departamento de Biología Celular, Fisiología e Inmunología, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Rosario Osta
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Salvador Martínez
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| |
Collapse
|
26
|
Suh MR, Choi WA, Choi YC, Lee JW, Hong JH, Park J, Kang SW. Long-Term Outcome of Amyotrophic Lateral Sclerosis in Korean Subjects. Ann Rehabil Med 2017; 41:1055-1064. [PMID: 29354583 PMCID: PMC5773426 DOI: 10.5535/arm.2017.41.6.1055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/03/2017] [Indexed: 11/16/2022] Open
Abstract
Objective To report the latest long-term outcome of amyotrophic lateral sclerosis (ALS) and to analyze the predictors of prognosis. Methods Subjects who were diagnosed with ALS between January 2005 and December 2009 at a single institute were followed up until death or up to December 2014. Data regarding age, sex, date of onset, date of diagnosis, presence of bulbar symptoms on onset, date of initiation of non-invasive ventilation (NIV), and the date of tracheostomy were collected. Survival was assessed using Kaplan-Meier curves and multivariate analyses of the risk of death were performed using the Cox proportional hazards model. Results Among 212 suspicious subjects, definite ALS was diagnosed in 182 subjects. The survival rate at 3 and 5 years from onset was 61.5% and 40.1%, respectively, and the survival rate at 3 and 5 years post-diagnosis was 49.5% and 24.2%, respectively. Further, 134 patients (134/182, 73.6%) were initiated on NIV, and among them, 90 patients (90/182, 49.5%) underwent tracheostomy. Male gender and onset age of ≥65 years were independent predictors of adverse survival. Conclusion The analysis of long term survival in ALS showed excellent outcomes considering the overall poor prognosis of this disease.
Collapse
Affiliation(s)
- Mi Ri Suh
- Department of Medicine, The Graduate School, Yonsei University, Seoul, Korea.,Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Won Ah Choi
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jang Woo Lee
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jung Hwa Hong
- Research Institute, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jihyun Park
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
27
|
Ciervo Y, Ning K, Jun X, Shaw PJ, Mead RJ. Advances, challenges and future directions for stem cell therapy in amyotrophic lateral sclerosis. Mol Neurodegener 2017; 12:85. [PMID: 29132389 PMCID: PMC5683324 DOI: 10.1186/s13024-017-0227-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative condition where loss of motor neurons within the brain and spinal cord leads to muscle atrophy, weakness, paralysis and ultimately death within 3–5 years from onset of symptoms. The specific molecular mechanisms underlying the disease pathology are not fully understood and neuroprotective treatment options are minimally effective. In recent years, stem cell transplantation as a new therapy for ALS patients has been extensively investigated, becoming an intense and debated field of study. In several preclinical studies using the SOD1G93A mouse model of ALS, stem cells were demonstrated to be neuroprotective, effectively delayed disease onset and extended survival. Despite substantial improvements in stem cell technology and promising results in preclinical studies, several questions still remain unanswered, such as the identification of the most suitable and beneficial cell source, cell dose, route of delivery and therapeutic mechanisms. This review will cover publications in this field and comprehensively discuss advances, challenges and future direction regarding the therapeutic potential of stem cells in ALS, with a focus on mesenchymal stem cells. In summary, given their high proliferation activity, immunomodulation, multi-differentiation potential, and the capacity to secrete neuroprotective factors, adult mesenchymal stem cells represent a promising candidate for clinical translation. However, technical hurdles such as optimal dose, differentiation state, route of administration, and the underlying potential therapeutic mechanisms still need to be assessed.
Collapse
Affiliation(s)
- Yuri Ciervo
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.,Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.,Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Xu Jun
- Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK
| | - Richard J Mead
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.
| |
Collapse
|
28
|
Sironi F, Vallarola A, Violatto MB, Talamini L, Freschi M, De Gioia R, Capelli C, Agostini A, Moscatelli D, Tortarolo M, Bigini P, Introna M, Bendotti C. Multiple intracerebroventricular injections of human umbilical cord mesenchymal stem cells delay motor neurons loss but not disease progression of SOD1G93A mice. Stem Cell Res 2017; 25:166-178. [PMID: 29154076 DOI: 10.1016/j.scr.2017.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 11/02/2017] [Accepted: 11/04/2017] [Indexed: 12/13/2022] Open
Abstract
Stem cell therapy is considered a promising approach in the treatment of amyotrophic lateral sclerosis (ALS) and mesenchymal stem cells (MSCs) seem to be the most effective in ALS animal models. The umbilical cord (UC) is a source of highly proliferating fetal MSCs, more easily collectable than other MSCs. Recently we demonstrated that human (h) UC-MSCs, double labeled with fluorescent nanoparticles and Hoechst-33258 and transplanted intracerebroventricularly (ICV) into SOD1G93A transgenic mice, partially migrated into the spinal cord after a single injection. This prompted us to assess the effect of repeated ICV injections of hUC-MSCs on disease progression in SOD1G93A mice. Although no transplanted cells migrated to the spinal cord, a partial but significant protection of motor neurons (MNs) was found in the lumbar spinal cord of hUC-MSCs-treated SOD1G93A mice, accompanied by a shift from a pro-inflammatory (IL-6, IL-1β) to anti-inflammatory (IL-4, IL-10) and neuroprotective (IGF-1) environment in the lumbar spinal cord, probably linked to the activation of p-Akt survival pathway in both motor neurons and reactive astrocytes. However, this treatment neither prevented the muscle denervation nor delayed the disease progression of mice, emphasizing the growing evidence that protecting the motor neuron perikarya is not sufficient to delay the ALS progression.
Collapse
Affiliation(s)
- Francesca Sironi
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Antonio Vallarola
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Martina Bruna Violatto
- Department of Biochemistry and Molecular Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Laura Talamini
- Department of Biochemistry and Molecular Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Mattia Freschi
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Roberta De Gioia
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Chiara Capelli
- USS Center of Cellular Therapy"G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Azzurra Agostini
- Department of Chemistry, Material and Chemical Engineering "G. Natta", Politecnico di Milano, Milano, Italy
| | - Davide Moscatelli
- Department of Chemistry, Material and Chemical Engineering "G. Natta", Politecnico di Milano, Milano, Italy
| | - Massimo Tortarolo
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Paolo Bigini
- Department of Biochemistry and Molecular Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Martino Introna
- USS Center of Cellular Therapy"G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Caterina Bendotti
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy.
| |
Collapse
|
29
|
Fang F, Peters TL, Beard JD, Umbach DM, Keller J, Mariosa D, Allen KD, Ye W, Sandler DP, Schmidt S, Kamel F. Blood Lead, Bone Turnover, and Survival in Amyotrophic Lateral Sclerosis. Am J Epidemiol 2017; 186:1057-1064. [PMID: 29020133 DOI: 10.1093/aje/kwx176] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022] Open
Abstract
Blood lead and bone turnover may be associated with the risk of amyotrophic lateral sclerosis (ALS). We aimed to assess whether these factors were also associated with time from ALS diagnosis to death through a survival analysis of 145 ALS patients enrolled during 2007 in the National Registry of Veterans with ALS. Associations of survival time with blood lead and plasma biomarkers of bone resorption (C-terminal telopeptides of type I collagen (CTX)) and bone formation (procollagen type I amino-terminal peptide (PINP)) were estimated using Cox models adjusted for age at diagnosis, diagnostic certainty, diagnostic delay, site of onset, and score on the Revised ALS Functional Rating Scale. Hazard ratios were calculated for each doubling of biomarker concentration. Blood lead, plasma CTX, and plasma PINP were mutually adjusted for one another. Increased lead (hazard ratio (HR) = 1.38; 95% confidence interval (CI): 1.03, 1.84) and CTX (HR = 2.03; 95% CI: 1.42, 2.89) were both associated with shorter survival, whereas higher PINP was associated with longer survival (HR = 0.59; 95% CI: 0.42, 0.83), after ALS diagnosis. No interactions were observed between lead or bone turnover and other prognostic indicators. Lead toxicity and bone metabolism may be involved in ALS pathophysiology.
Collapse
|
30
|
Gowing G, Svendsen S, Svendsen CN. Ex vivo gene therapy for the treatment of neurological disorders. PROGRESS IN BRAIN RESEARCH 2017; 230:99-132. [PMID: 28552237 DOI: 10.1016/bs.pbr.2016.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ex vivo gene therapy involves the genetic modification of cells outside of the body to produce therapeutic factors and their subsequent transplantation back into patients. Various cell types can be genetically engineered. However, with the explosion in stem cell technologies, neural stem/progenitor cells and mesenchymal stem cells are most often used. The synergy between the effect of the new cell and the additional engineered properties can often provide significant benefits to neurodegenerative changes in the brain. In this review, we cover both preclinical animal studies and clinical human trials that have used ex vivo gene therapy to treat neurological disorders with a focus on Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, and stroke. We highlight some of the major advances in this field including new autologous sources of pluripotent stem cells, safer ways to introduce therapeutic transgenes, and various methods of gene regulation. We also address some of the remaining hurdles including tunable gene regulation, in vivo cell tracking, and rigorous experimental design. Overall, given the current outcomes from researchers and clinical trials, along with exciting new developments in ex vivo gene and cell therapy, we anticipate that successful treatments for neurological diseases will arise in the near future.
Collapse
Affiliation(s)
- Genevieve Gowing
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Soshana Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
31
|
Czarzasta J, Habich A, Siwek T, Czapliński A, Maksymowicz W, Wojtkiewicz J. Stem cells for ALS: An overview of possible therapeutic approaches. Int J Dev Neurosci 2017; 57:46-55. [PMID: 28088365 DOI: 10.1016/j.ijdevneu.2017.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an unusual, fatal, neurodegenerative disorder leading to the loss of motor neurons. After diagnosis, the average lifespan ranges from 3 to 5 years, and death usually results from respiratory failure. Although the pathogenesis of ALS remains unclear, multiple factors are thought to contribute to the progression of ALS, such as network interactions between genes, environmental exposure, impaired molecular pathways and many others. The neuroprotective properties of neural stem cells (NSCs) and the paracrine signaling of mesenchymal stem cells (MSCs) have been examined in multiple pre-clinical trials of ALS with promising results. The data from these initial trials indicate a reduction in the rate of disease progression. The mechanism through which stem cells achieve this reduction is of major interest. Here, we review the to-date pre-clinical and clinical therapeutic approaches employing stem cells, and discuss the most promising ones.
Collapse
Affiliation(s)
- Joanna Czarzasta
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland.
| | - Aleksandra Habich
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Tomasz Siwek
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Adam Czapliński
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland; Neurocentrum Bellevue, Neurology, Zurich, Switzerland
| | - Wojciech Maksymowicz
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland; Laboratory of Regenerative Medicine, University of Warmia and Mazury, Olsztyn, Poland; Foundation for nerve cells regeneration, Olsztyn, Poland
| |
Collapse
|
32
|
Gene therapy and respiratory neuroplasticity. Exp Neurol 2016; 287:261-267. [PMID: 27697480 DOI: 10.1016/j.expneurol.2016.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/19/2016] [Accepted: 09/27/2016] [Indexed: 01/01/2023]
Abstract
Breathing is a life-sustaining behavior that in mammals is accomplished by activation of dedicated muscles responsible for inspiratory and expiratory forces acting on the lung and chest wall. Motor control is exerted by specialized pools of motoneurons in the medulla and spinal cord innervated by projections from multiple centers primarily in the brainstem that act in concert to generate both the rhythm and pattern of ventilation. Perturbations that prevent the accomplishment of the full range of motor behaviors by respiratory muscles commonly result in significant morbidity and increased mortality. Recent developments in gene therapy and novel targeting strategies have contributed to deeper understanding of the organization of respiratory motor systems. Gene therapy has received widespread attention and substantial progress has been made in recent years with the advent of improved tools for vector design. Genes can be delivered via a variety of plasmids, synthetic or viral vectors and cell therapies. In recent years, adeno-associated viruses (AAV) have become one of the most commonly used vector systems, primarily because of the extensive characterization conducted to date and the versatility in targeting strategies. Recent studies highlight the power of using AAV to selectively and effectively transduce respiratory motoneurons and muscle fibers with promising therapeutic effects. This brief review summarizes current evidence for the use of gene therapy in respiratory disorders with a primary focus on interventions that address motor control and neuroplasticity, including regeneration, in the respiratory system.
Collapse
|
33
|
Gubert F, Satiago MF. Prospects for bone marrow cell therapy in amyotrophic lateral sclerosis: how far are we from a clinical treatment? Neural Regen Res 2016; 11:1216-9. [PMID: 27651758 PMCID: PMC5020809 DOI: 10.4103/1673-5374.189167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that causes progressive muscular atrophy and death within 3-5 years after its onset. Despite the significant advances in knowledge of ALS pathology, no effective treatment is available. Therefore, it is imperative to search for new alternatives to treat ALS. Cell therapy, especially using bone-marrow cells, has showed to be very useful to protect the neural tissue in different brain disease or traumatic lesions. In ALS, most published results show beneficial effects of the use bone marrow cells, especially mesenchymal stromal cells. However, until now, the best outcome extends animal's lifespan by only a few weeks. It is essential to continue the search for a really effective therapy, testing different cells, routes and time-windows of administration. Studying the mechanisms that initiate and spread the degenerative process is also important to find out an effective therapy. Therefore, we discussed here some progresses that have been made using bone-marrow cell therapy as a therapeutic tool for ALS.
Collapse
Affiliation(s)
- Fernanda Gubert
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Marcelo F. Satiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
34
|
Xian B, Zhang Y, Peng Y, Huang J, Li W, Wang W, Zhang M, Li K, Zhang H, Zhao M, Liu X, Huang B. Adult Human Peripheral Blood Mononuclear Cells Are Capable of Producing Neurocyte or Photoreceptor-Like Cells That Survive in Mouse Eyes After Preinduction With Neonatal Retina. Stem Cells Transl Med 2016; 5:1515-1524. [PMID: 27458266 DOI: 10.5966/sctm.2015-0395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 04/18/2016] [Indexed: 02/07/2023] Open
Abstract
: Adult human peripheral blood mononuclear cells (hPBMCs) exhibit pluripotency in vitro and so may be a valuable cell source for regenerative therapies. The efficacy of such therapies depends on the survival, differentiation, migration, and integration capacity of hPBMCs in specific tissues. In this study, we examined these capacities of transplanted hPBMCs in mouse retina as well functional improvement after transplant. We isolated hPBMCs and preinduced them for 4 days in media preconditioned with postnatal day 1 rat retina explants. Preinduction increased the proportions of hPBMCs expressing neural stem cell, neural progenitor cell, or photoreceptor markers as revealed by immunofluorescent staining, flow cytometry, and quantitative real-time polymerase chain reaction. Preinduced hPBMCs were transplanted into the subretinal space of retinal degenerative slow (RDS) and retinal degeneration 1 (RD1) mice. At 1, 3, and 6 months after transplantation, treated eyes of RDS mice were collected and cell phenotype was studied by immunofluorescent staining. Preinduced hPBMCs survived in the subretinal space; migrated away from the injection site and into multiple retinal layers; and expressed neural stem cell, neuronal, and photoreceptor markers. Finally, we assessed RD1 retinal function after subretinal transplantation and found significant improvement at 3 months after transplantation. The ease of harvesting, viability in vivo, capacity to express neuronal and photoreceptor proteins, and capacity for functional enhancement suggest that hPBMCs are potential candidates for cell replacement therapy to treat retinal degenerative diseases. SIGNIFICANCE This study provides support for the use of peripheral blood mononuclear cells (PBMCs) as a potential source of pluripotent stem cells for treating retinal degeneration. First, this study demonstrated that PBMCs can differentiate into retinal neuron-like cells in vitro and in vivo. Second, some transplanted cells expressed markers for neural progenitors, mature neurons, or photoreceptors at 1, 3, and 6 months after subretinal injection. Finally, this study showed that PBMC transplantation can improve the function of a degenerated retina.
Collapse
Affiliation(s)
- Bikun Xian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yichi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yuting Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianfa Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wencong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Min Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hening Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Bing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Arnal-Pastor M, Martínez-Ramos C, Vallés-Lluch A, Pradas MM. Influence of scaffold morphology on co-cultures of human endothelial and adipose tissue-derived stem cells. J Biomed Mater Res A 2016; 104:1523-33. [DOI: 10.1002/jbm.a.35682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 11/08/2022]
Affiliation(s)
- M. Arnal-Pastor
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; C. de Vera s/n Valencia 46022 Spain
| | - C. Martínez-Ramos
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; C. de Vera s/n Valencia 46022 Spain
| | - A. Vallés-Lluch
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; C. de Vera s/n Valencia 46022 Spain
| | - M. Monleón Pradas
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; C. de Vera s/n Valencia 46022 Spain
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine; Valencia Spain
| |
Collapse
|
36
|
Suzuki M, Svendsen CN. Ex Vivo Gene Therapy Using Human Mesenchymal Stem Cells to Deliver Growth Factors in the Skeletal Muscle of a Familial ALS Rat Model. Methods Mol Biol 2016; 1382:325-36. [PMID: 26611598 DOI: 10.1007/978-1-4939-3271-9_24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic protein and molecule delivery to target sites by transplanted human stem cells holds great promise for ex vivo gene therapy. Our group has demonstrated the therapeutic benefits of ex vivo gene therapy targeting the skeletal muscles in a transgenic rat model of familial amyotrophic lateral sclerosis (ALS). We used human mesenchymal stem cells (hMSCs) and genetically modified them to release neuroprotective growth factors such as glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF). Intramuscular growth factor delivery via hMSCs can enhance neuromuscular innervation and motor neuron survival in a rat model of ALS (SOD1(G93A) transgenic rats). Here, we describe the protocol of ex vivo delivery of growth factors via lentiviral vector-mediated genetic modification of hMSCs and hMSC transplantation into the skeletal muscle of a familial ALS rat model.
Collapse
Affiliation(s)
- Masatoshi Suzuki
- Department of Comparative Biosciences, The Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, 4124 Veterinary Medicine Building, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| |
Collapse
|
37
|
|
38
|
Stem Cells and Regenerative Medicine: Myth or Reality of the 21th Century. Stem Cells Int 2015; 2015:734731. [PMID: 26300923 PMCID: PMC4537770 DOI: 10.1155/2015/734731] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/22/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton's Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ's reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.
Collapse
|
39
|
Scarrott JM, Herranz-Martín S, Alrafiah AR, Shaw PJ, Azzouz M. Current developments in gene therapy for amyotrophic lateral sclerosis. Expert Opin Biol Ther 2015; 15:935-47. [DOI: 10.1517/14712598.2015.1044894] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
|