1
|
Petrou P, Kassis I, Levi Y, Yaghmour N, Epstein T, Ginzberg A, Karussis D. Kinetics of serum NFL and GFAP and changes in cognitive functions, in MS patients treated with repeated administrations of autologous mesenchymal stem cells (MSC-NG01). J Neuroimmunol 2025; 403:578613. [PMID: 40215559 DOI: 10.1016/j.jneuroim.2025.578613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Intrathecal injection (IT) of autologous mesenchymal stem cells (MSC) showed robust beneficial effects a previous randomized study from our center, in patients with progressive multiple sclerosis (MS) (NCT02166021). We evaluated here the effect of repeated MSC-NG01 transplantations on serum biomarkers of neuroinflammation and neurodegeneration, namely, neurofilaments light chains (NFL) and glial fibrillary acidic protein (GFAP), in an open-label extension trial. METHODS 23 patients with progressive types of MS, who participated in the NCT02166021-trial, were included. Patients were treated with 2-3 intrathecal injections of MSC-NG01 and followed up for a period of >12 months. Safety/tolerability and various efficacy measurements, including EDSS/FS, timed 25-ft (T25FW) walking, cognitive functions, and serum levels of the biomarkers NFL and GFAP, were assessed. RESULTS Serum levels of NFL and GFAP showed a gradual and consistent reduction after the intrathecal treatments with MSC-NG01, in multiple measurements. The mean NFL reduction at last observation after one year was 33.2 % (p < 0.001, Wilcoxon-paired test). Serum levels of GFAP were reduced in all tested patients (p < 0.0004, Wilcoxon-paired test). A significant improvement was observed in T25-FW and in the sum of all functional systems (FS) at the final visit of 12 months. SDMT cognitive test was also improved by a mean of >3 degrees (p = 0.0008). CONCLUSION Treatment of progressive MS patients with IT injections of autologous MSC-NG01 induced a reduction in both NFL and GFAP biomarkers levels, paralleled by beneficial effects on cognition, neurological functional tests and quality of life. These data indicate significant effects of MSC-transplantation on neurodegeneration and neuroinflammation.
Collapse
Affiliation(s)
- Panayiota Petrou
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel.
| | - Ibrahim Kassis
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Yarden Levi
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Nour Yaghmour
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Tehila Epstein
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Ariel Ginzberg
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Dimitrios Karussis
- Unit of Neuroimmunology and Multiple Sclerosis Center and The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| |
Collapse
|
2
|
Nowaczewska-Kuchta A, Ksiazek-Winiarek D, Glabinski A. Interaction Between Neutrophils and Elements of the Blood-Brain Barrier in the Context of Multiple Sclerosis and Ischemic Stroke. Int J Mol Sci 2025; 26:4437. [PMID: 40362673 PMCID: PMC12072651 DOI: 10.3390/ijms26094437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
The blood-brain barrier (BBB) is a semi-permeable membrane in physiological conditions, but in pathologies like multiple sclerosis (MS) and ischemic stroke (IS), its permeability increases. In this review, we focus on neutrophils and their interaction with cellular components of the BBB: endothelial cells (EC), pericytes (PC), and astrocytes (AC). Nowadays, neutrophils receive more attention, mostly due to advanced research techniques that show the complexity of their population. Additionally, neutrophils have the ability to secrete extracellular vesicles (EVs), reactive oxygen species (ROS) and cytokines, which both destroy and restore the BBB. Astrocytes, PCs, and ECs also have dual roles in the pathogenesis of MS and IS. The interaction between neutrophils and cellular components of the BBB provides us with a wider insight into the pathogenesis of common diseases in the central nervous system. Further, we comprehensively review knowledge about the influence of neutrophils on the BBB in the context of MS and IS. Moreover, we describe new therapeutic strategies for patients with MS and IS like cell-based therapies and therapies that use the neutrophil function.
Collapse
Affiliation(s)
| | | | - Andrzej Glabinski
- Department of Neurology and Stroke, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (A.N.-K.); (D.K.-W.)
| |
Collapse
|
3
|
Mitrache Desaga AC, Noui Y, Silfverberg T, Carlson K, Burman J. Patient-reported outcomes of quality of life before and after autologous hematopoietic stem cell transplantation for multiple sclerosis. Mult Scler 2025; 31:455-463. [PMID: 39957535 PMCID: PMC11956378 DOI: 10.1177/13524585251315363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (AHSCT) is a therapeutic intervention for multiple sclerosis (MS) that has gained increased attention in the last decade. The impact of this intervention on the quality of life (QoL) of patients with MS remains unclear. OBJECTIVE The aim of this study was to investigate the impact of AHSCT on QoL in patients with MS using Multiple Sclerosis Impact Scale (MSIS-29) scores. METHODS In this observational retrospective cohort study, patients with relapsing-remitting MS treated with AHSCT in Sweden from 2004, when the first transplant was performed, until 31 December 2019, were considered for participation. Anonymized outcome data were extracted from the Swedish MS registry in May 2022. RESULTS Out of 213 patients assessed for eligibility in the study, 96 were included in the final analysis. After a median follow-up of 5.2 (IQR 3.2-6.8) years, 58% improved, 14% remained unchanged and 28% worsened in the physical domain of the MSIS-29. In the psychological domain, 63% improved, 18% remained unchanged and 19% worsened. Improvements in both domains occurred early, within the first year following intervention. CONCLUSIONS Treatment intervention with AHSCT is associated with a clinically meaningful improvement in QoL.
Collapse
Affiliation(s)
| | - Yassine Noui
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Thomas Silfverberg
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Center for Clinical Research Dalarna, Uppsala University, Falun, Sweden
| | - Kristina Carlson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Henry-Ojo HO, Liu F, Narayanan SP. Targeting SMOX Preserves Optic Nerve Myelin, Axonal Integrity, and Visual Function in Multiple Sclerosis. Biomolecules 2025; 15:158. [PMID: 40001462 PMCID: PMC11853291 DOI: 10.3390/biom15020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Multiple sclerosis (MS) is a highly disabling chronic neurological condition affecting young adults. Inflammation, demyelination, and axonal damage are key pathological features of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Our previous work demonstrated that inhibiting spermine oxidase (SMOX) with MDL72527, a selective irreversible pharmacological inhibitor, significantly reduced clinical symptoms, retinal ganglion cell (RGC) loss, and optic nerve inflammation in EAE mice. The present study explored the broader therapeutic potential of SMOX inhibition, focusing on myelin preservation, axonal integrity, and visual function in the EAE model. Electron microscopy of optic nerve cross-sections showed significant preservation of myelin thickness and axonal integrity due to SMOX inhibition. The quantitative assessment showed that g-ratio and axon count metrics were significantly improved in MDL72527-treated EAE mice compared to their vehicle-treated counterparts. Immunofluorescence studies confirmed these findings, showing increased preservation of myelin and axonal proteins in MDL72527-treated EAE mice compared to the vehicle-treated group. Functional assessment studies (Electroretinography) demonstrated significant improvement in RGC function and axonal conduction in EAE mice treated with MDL72527. Furthermore, SMOX inhibition downregulated the expression of galectin3 (Gal3), a mediator of neuroinflammation, indicating Gal3's role in SMOX-mediated neuroprotection. This study provides compelling evidence for the potential of SMOX inhibition as a therapeutic strategy in multiple sclerosis and other demyelinating disorders.
Collapse
Affiliation(s)
- Harry O. Henry-Ojo
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| | - Fang Liu
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| | - S. Priya Narayanan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30907, USA; (H.O.H.-O.); (F.L.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30907, USA
| |
Collapse
|
5
|
Yue Y, Deng B, Zeng Y, Li W, Qiu X, Hu P, Shen L, Ruan T, Zhou R, Li S, Ying J, Xiong T, Qu Y, Luan Z, Mu D. Oligodendrocyte Progenitor Cell Transplantation Reduces White Matter Injury in a Fetal Goat Model. CNS Neurosci Ther 2024; 30:e70178. [PMID: 39690788 DOI: 10.1111/cns.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Preterm white matter injury (PWMI) is the most common type of brain injury in preterm infants, in which, oligodendrocyte progenitor cells (OPCs) are predominantly damaged. In this study, human OPCs (hOPCs) were administered to a fetal goat model of PWMI to examine the differentiation potential and therapeutic effects of the cells on PWMI. METHODS Preterm goat fetuses were subjected to hypoxic-ischemia (HI) via intermittent umbilical cord occlusion (5 min × 5). Twenty million hOPCs were administered via a nasal catheter 12 h after an HI insult, and brain tissues were collected 14 or 21 days after the HI insult. Myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) were detected by immunofluorescence and western blotting techniques. The percentage of myelinated nerve fibers and g-ratio were examined using transmission electron microscopy. Inflammatory cells were detected by immunohistochemistry. Inflammatory and neurotrophic factors were measured using enzyme-linked immunosorbent assay. RESULTS Our results showed that intermittent umbilical cord occlusion induced PWMI in fetal goats. Transplanted hOPCs can survive in periventricular and subcortical white matter. Further, transplanted hOPCs expressed markers of mature oligodendrocytes (MBP and MAG) and few cells expressed markers of preoligodendrocytes (NG2 and A2B5), suggesting that these cells can differentiate into mature oligodendrocytes in the brain. In addition, hOPCs administration increased MBP and MAG levels, percentage of myelinated nerve fibers, and thickness of the myelin sheath, indicating a reduction in PWMI. Furthermore, hOPCs did not increase the inflammatory response after HI. Interestingly, hOPC administration decreased tumor necrosis factor-alpha and increased glial-derived neurotrophic factor and brain-derived neurotrophic factor levels after HI, suggesting that additional mechanisms mediate the inflammatory microenvironment and neuroprotective effects. CONCLUSIONS Exogenous hOPCs can differentiate into mature oligodendrocytes in fetal goats and alleviate HI-induced PWMI. Transplantation of hOPCs is a promising strategy for treating PWMI.
Collapse
Affiliation(s)
- Yan Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Bixin Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Yan Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Wenxing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Xia Qiu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Peng Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - LiuHong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tiechao Ruan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Ruixi Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shiping Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Junjie Ying
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Tao Xiong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Yi Qu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Zuo Luan
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dezhi Mu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Luciani M, Garsia C, Beretta S, Cifola I, Peano C, Merelli I, Petiti L, Miccio A, Meneghini V, Gritti A. Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice. Nat Commun 2024; 15:9433. [PMID: 39487141 PMCID: PMC11530573 DOI: 10.1038/s41467-024-53613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NSCs) hold promise for treating neurodegenerative and demyelinating disorders. However, comprehensive studies on their identity and safety remain limited. In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional characteristics with human fetal neural stem cells (hfNSCs) while exhibiting divergent profiles from glioblastoma stem cells. Long-term transplantation studies in mice showed robust and stable engraftment of hiPSC-NSCs, with predominant differentiation into glial cells and no evidence of tumor formation. Additionally, we identified the Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) as a regulator of astroglial differentiation in hiPSC-NSCs. These findings provide valuable transcriptional and epigenetic reference datasets to prospectively define the maturation stage of NSCs derived from different hiPSC sources and demonstrate the long-term safety of hiPSC-NSCs, reinforcing their potential as a viable alternative to hfNSCs for clinical applications.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Garsia
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Rozzano, Milan, Italy
- Human Technopole, Via Rita Levi Montalcini 1, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Luca Petiti
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Annarita Miccio
- IMAGINE Institute, Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
7
|
Panghal A, Flora SJS. Nano-based approaches for the treatment of neuro-immunological disorders: a special emphasis on multiple sclerosis. DISCOVER NANO 2024; 19:171. [PMID: 39466516 PMCID: PMC11519283 DOI: 10.1186/s11671-024-04135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Multiple sclerosis (MS) is a neuroimmunological disorder which causes axonal damage, demyelination and paralysis. Although numerous therapeutics have been developed for the effective treatment of MS and a few have been approved in recent decades, complete remission and treatment of MS remain a matter of concern. Nanotechnology is a potential approach for manipulating the properties of materials at the molecular level to attain desired properties. This approach is effective in the treatment of several CNS disorders by enhancing drug delivery, bioavailability and efficacy. We have briefly discussed the neuroimmunological disorders with a particular emphasis on MS. We also explored nanoengineered drug delivery systems, describing several nano-formulations for the treatment of MS, challenges and future of nanotechnology.
Collapse
Affiliation(s)
- Archna Panghal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-SAS Nagar, Mohali, 160102, India
| | - S J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-SAS Nagar, Mohali, 160102, India.
- Era College of Pharmaceutical Sciences, Era Lucknow Medical University, Sarfarajgang, Lucknow, 226002, India.
| |
Collapse
|
8
|
Medina R, Derias AM, Lakdawala M, Speakman S, Lucke-Wold B. Overview of emerging therapies for demyelinating diseases. World J Clin Cases 2024; 12:6361-6373. [PMID: 39464332 PMCID: PMC11438674 DOI: 10.12998/wjcc.v12.i30.6361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
This paper provides an overview of autoimmune disorders of the central nervous system, specifically those caused by demyelination. We explore new research regarding potential therapeutic interventions, particularly those aimed at inducing remyelination. Remyelination is a detailed process, involving many cell types-oligodendrocyte precursor cells (OPCs), astrocytes, and microglia-and both the innate and adaptive immune systems. Our discussion of this process includes the differentiation potential of neural stem cells, the function of adult OPCs, and the impact of molecular mediators on myelin repair. Emerging therapies are also explored, with mechanisms of action including the induction of OPC differentiation, the transplantation of mesenchymal stem cells, and the use of molecular mediators. Further, we discuss current medical advancements in relation to many myelin-related disorders, including multiple sclerosis, optic neuritis, neuromyelitis optica spectrum disorder, myelin oligodendrocyte glycoprotein antibody-associated disease, transverse myelitis, and acute disseminated encephalomyelitis. Beyond these emerging systemic therapies, we also introduce the dimethyl fumarate/silk fibroin nerve conduit and its potential role in the treatment of peripheral nerve injuries. Despite these aforementioned scientific advancements, this paper maintains the need for ongoing research to deepen our understanding of demyelinating diseases and advance therapeutic strategies that enhance affected patients' quality of life.
Collapse
Affiliation(s)
- Robert Medina
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Ann-Marie Derias
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Maria Lakdawala
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Skye Speakman
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
9
|
Olejnik P, Roszkowska Z, Adamus S, Kasarełło K. Multiple sclerosis: a narrative overview of current pharmacotherapies and emerging treatment prospects. Pharmacol Rep 2024; 76:926-943. [PMID: 39177889 PMCID: PMC11387431 DOI: 10.1007/s43440-024-00642-0] [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: 06/13/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease characterized by pathological processes of demyelination, subsequent axonal loss, and neurodegeneration within the central nervous system. Despite the availability of numerous disease-modifying therapies that effectively manage this condition, there is an emerging need to identify novel therapeutic targets, particularly for progressive forms of MS. Based on contemporary insights into disease pathophysiology, ongoing efforts are directed toward developing innovative treatment modalities. Primarily, monoclonal antibodies have been extensively investigated for their efficacy in influencing specific pathological pathways not yet targeted. Emerging approaches emphasizing cellular mechanisms, such as chimeric antigen receptor T cell therapy targeting immunological cells, are attracting increasing interest. The evolving understanding of microglia and the involvement of ferroptotic mechanisms in MS pathogenesis presents further avenues for targeted therapies. Moreover, innovative treatment strategies extend beyond conventional approaches to encompass interventions that target alterations in microbiota composition and dietary modifications. These adjunctive therapies hold promise as complementary methods for the holistic management of MS. This narrative review aims to summarize current therapies and outline potential treatment methods for individuals with MS.
Collapse
Affiliation(s)
- Piotr Olejnik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Roszkowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Sylwia Adamus
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Kaja Kasarełło
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
10
|
Kråkenes T, Sandvik CE, Ytterdal M, Gavasso S, Evjenth EC, Bø L, Kvistad CE. The Therapeutic Potential of Exosomes from Mesenchymal Stem Cells in Multiple Sclerosis. Int J Mol Sci 2024; 25:10292. [PMID: 39408622 PMCID: PMC11477223 DOI: 10.3390/ijms251910292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Although treatment for multiple sclerosis (MS) has undergone a revolution in the last decades, at least two important barriers remain: alleviation of innate inflammation driving disease progression and promotion of remyelination and neural regeneration. Mesenchymal stem cells (MSCs) possess immunomodulatory properties and promote remyelination in murine MS models. The main therapeutic mechanism has, however, been attributed to their potent paracrine capacity, and not to in vivo tissue implantation. Studies have demonstrated that exosomes released as part of the cells' secretome effectively encapsulate the beneficial properties of MSCs. These membrane-enclosed nanoparticles contain a variety of proteins and nucleic acids and serve as mediators of intercellular communication. In vitro studies have demonstrated that exosomes from MSCs modulate activated microglia from an inflammatory to an anti-inflammatory phenotype and thereby dampen the innate inflammation. Rodent studies have also demonstrated potent immunomodulation and remyelination with improved outcomes following exosome administration. Thus, exosomes from MSCs may represent a potential cell-free treatment modality to prevent disease progression and promote remyelination in MS. In this narrative review, we summarize the current knowledge of exosomes from MSCs as a potential treatment for MS and discuss the remaining issues before successful translation into clinical trials.
Collapse
Affiliation(s)
- Torbjørn Kråkenes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Casper Eugen Sandvik
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Marie Ytterdal
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Sonia Gavasso
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Elisabeth Claire Evjenth
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Lars Bø
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Christopher Elnan Kvistad
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| |
Collapse
|
11
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
12
|
Santos SIP, Ortiz-Peñuela SJ, de Paula Filho A, Tomiyama ALMR, Coser LDO, da Silveira JC, Martins DDS, Ciena AP, de Oliveira ALR, Ambrósio CE. Oligodendrocyte precursor cell-derived exosomes combined with cell therapy promote clinical recovery by immunomodulation and gliosis attenuation. Front Cell Neurosci 2024; 18:1413843. [PMID: 39109218 PMCID: PMC11301646 DOI: 10.3389/fncel.2024.1413843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 01/22/2025] Open
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system characterized by autoimmune destruction of the myelin sheath, leading to irreversible and progressive functional deficits in patients. Pre-clinical studies involving the use of neural stem cells (NSCs) have already demonstrated their potential in neuronal regeneration and remyelination. However, the exclusive application of cell therapy has not proved sufficient to achieve satisfactory therapeutic levels. Recognizing these limitations, there is a need to combine cell therapy with other adjuvant protocols. In this context, extracellular vesicles (EVs) can contribute to intercellular communication, stimulating the production of proteins and lipids associated with remyelination and providing trophic support to axons. This study aimed to evaluate the therapeutic efficacy of the combination of NSCs and EVs derived from oligodendrocyte precursor cells (OPCs) in an animal model of multiple sclerosis. OPCs were differentiated from NSCs and had their identity confirmed by gene expression analysis and immunocytochemistry. Exosomes were isolated by differential ultracentrifugation and characterized by Western, transmission electron microscopy and nanoparticle tracking analysis. Experimental therapy of C57BL/6 mice induced with experimental autoimmune encephalomyelitis (EAE) were grouped in control, treated with NSCs, treated with OPC-derived EVs and treated with a combination of both. The treatments were evaluated clinically using scores and body weight, microscopically using immunohistochemistry and immunological profile by flow cytometry. The animals showed significant clinical improvement and weight gain with the treatments. However, only the treatments involving EVs led to immune modulation, changing the profile from Th1 to Th2 lymphocytes. Fifteen days after treatment revealed a reduction in reactive microgliosis and astrogliosis in the groups treated with EVs. However, there was no reduction in demyelination. The results indicate the potential therapeutic use of OPC-derived EVs to attenuate inflammation and promote recovery in EAE, especially when combined with cell therapy.
Collapse
Affiliation(s)
- Sarah Ingrid Pinto Santos
- Faculty of Animal Science and Food Engineering, University of São Paulo (FZEA/USP), São Paulo, Brazil
| | | | - Alessandro de Paula Filho
- Faculty of Animal Science and Food Engineering, University of São Paulo (FZEA/USP), São Paulo, Brazil
| | | | | | | | | | | | | | - Carlos Eduardo Ambrósio
- Faculty of Animal Science and Food Engineering, University of São Paulo (FZEA/USP), São Paulo, Brazil
| |
Collapse
|
13
|
Afshar Hezarkhani L, Veysi K, Rahmani A, Salari N, Hasheminezhad R, Nasr V, Mohammadi M. Safety and Efficacy of Bone Marrow and Adipose Tissue-Derived Mesenchymal Stem Cells for the Treatment of Ischemic Stroke: A Systematic Review. Cardiol Rev 2024:00045415-990000000-00214. [PMID: 38358290 DOI: 10.1097/crd.0000000000000671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Neurological diseases, including ischemic stroke, are considered a big challenge for public health due to their high prevalence and lack of definitive and effective treatments. Addressing these issues requires innovative therapeutic approaches and among the limited methods available, stem cells have shown promise in improving central nervous system repair by enhancing myelin regeneration and neuronal recovery. To advance this field of research, this systematic review aims to assess the safety and effectiveness of mesenchymal stem cells (MSCs) derived from both bone marrow and adipose tissue for the treatment of ischemic stroke. This study conducted a systematic review in the electronic databases PubMed, Scopus, Web of Science, Embase, ScienceDirect, and Google Scholar to assess the efficacy and safety of MSCs generated from bone marrow and adipose tissue for the treatment of ischemic stroke. It was extracted without a time limit until April 2023. The studies were then transferred to the information management program (EndNote) and duplicates were eliminated. The remaining studies were then examined using the entry and exit criteria and the 3 stages of primary, secondary, and qualitative evaluation, and finally, the results of the final studies were extracted. According to the initial search in the desired databases, 1028 possible related articles were identified and transferred to the information management software (EndNote). After removing 390 duplicate studies, 608 studies were excluded based on inclusion and exclusion criteria. Finally, 37 final studies were included in the systematic review process. Based on the investigations, it was evident that the administration of MSCs derived from both bone marrow and adipose tissue holds significant promise as an effective and safe treatment approach for ischemic stroke. The results consistently showed acceptable outcomes in the studies and this evidence can be recommended for the clinical application of this treatment. Also, the findings of this study report that the use of adipose tissue and bone marrow MSCs in the treatment of ischemic stroke can be used as a practical method.
Collapse
Affiliation(s)
- Leila Afshar Hezarkhani
- From the Neuroscience Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kazhal Veysi
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Adibeh Rahmani
- Center for Musculoskeletal Biomechanics and Regeneration, Julius Wolff Institut, Charité, Berlin, Germany
| | - Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Razie Hasheminezhad
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vahideh Nasr
- Department of Neurology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Mohammadi
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
14
|
Jamali F, Aldughmi M, Atiani S, Al-Radaideh A, Dahbour S, Alhattab D, Khwaireh H, Arafat S, Jaghbeer JA, Rahmeh R, Abu Moshref K, Bawaneh H, Hassuneh MR, Hourani B, Ababneh O, Alghwiri A, Awidi A. Human Umbilical Cord-Derived Mesenchymal Stem Cells in the Treatment of Multiple Sclerosis Patients: Phase I/II Dose-Finding Clinical Study. Cell Transplant 2024; 33:9636897241233045. [PMID: 38450623 PMCID: PMC10921855 DOI: 10.1177/09636897241233045] [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: 09/12/2023] [Revised: 01/08/2024] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neuro-inflammatory disease resulting in disabilities that negatively impact patients' life quality. While current treatment options do not reverse the course of the disease, treatment using mesenchymal stromal/stem cells (MSC) is promising. There has yet to be a consensus on the type and dose of MSC to be used in MS. This work aims to study the safety and efficacy of two treatment protocols of MSCs derived from the umbilical cord (UC-MSCs) and their secretome. The study included two groups of MS patients; Group A received two intrathecal doses of UC-MSCs, and Group B received a single dose. Both groups received UC-MSCs conditioned media 3 months post-treatment. Adverse events in the form of a clinical checklist and extensive laboratory tests were performed. Whole transcriptome analysis was performed on patients' cells at baseline and post-treatment. Results showed that all patients tolerated the cellular therapy without serious adverse events. The general disability scale improved significantly in both groups at 6 months post-treatment. Examining specific aspects of the disease revealed more parameters that improved in Group A compared to Group B patients, including a significant increase in the (CD3+CD4+) expressing lymphocytes at 12 months post-treatment. In addition, better outcomes were noted regarding lesion load, cortical thickness, manual dexterity, and information processing speed. Both protocols impacted the transcriptome of treated participants with genes, transcription factors, and microRNAs (miRNAs) differentially expressed compared to baseline. Inflammation-related and antigen-presenting (HLA-B) genes were downregulated in both groups. In contrast, TNF-alpha, TAP-1, and miR142 were downregulated only in Group A. The data presented indicate that both protocols are safe. Furthermore, it suggests that administering two doses of stem cells can be more beneficial to MS patients. Larger multisite studies should be initiated to further examine similar or higher doses of MSCs.
Collapse
Affiliation(s)
- Fatima Jamali
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Mayis Aldughmi
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Serin Atiani
- Data Science Department, Princess Sumaya University for Technology, Amman, Jordan
| | - Ali Al-Radaideh
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Jordan University Hospital, The University of Jordan, Amman, Jordan
- Laboratory of Nanomedicine, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Said Dahbour
- Department of Medical Imaging, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Dana Alhattab
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Department of Medical Radiography, School of Health Sciences, University of Doha for Science and Technology, Doha, Qatar
| | - Hind Khwaireh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Sally Arafat
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Joud Al Jaghbeer
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Reem Rahmeh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | | | - Hisham Bawaneh
- Hematology Department, Jordan University Hospital, Amman, Jordan
| | - Mona R. Hassuneh
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biology, Faculty of Sciences, The University of Jordan, Amman, Jordan
| | - Bayan Hourani
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Osameh Ababneh
- Department of Ophthalmology, Jordan University Hospital, School of Medicine, The University of Jordan, Amman, Jordan
| | - Alia Alghwiri
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Hematology Department, Jordan University Hospital, Amman, Jordan
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
15
|
Christodoulou MV, Petkou E, Atzemoglou N, Gkorla E, Karamitrou A, Simos YV, Bellos S, Bekiari C, Kouklis P, Konitsiotis S, Vezyraki P, Peschos D, Tsamis KI. Cell replacement therapy with stem cells in multiple sclerosis, a systematic review. Hum Cell 2024; 37:9-53. [PMID: 37985645 PMCID: PMC10764451 DOI: 10.1007/s13577-023-01006-1] [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: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system (CNS), characterized by demyelination and axonal loss. It is induced by attack of autoreactive lymphocytes on the myelin sheath and endogenous remyelination failure, eventually leading to accumulation of neurological disability. Disease-modifying agents can successfully address inflammatory relapses, but have low efficacy in progressive forms of MS, and cannot stop the progressive neurodegenerative process. Thus, the stem cell replacement therapy approach, which aims to overcome CNS cell loss and remyelination failure, is considered a promising alternative treatment. Although the mechanisms behind the beneficial effects of stem cell transplantation are not yet fully understood, neurotrophic support, immunomodulation, and cell replacement appear to play an important role, leading to a multifaceted fight against the pathology of the disease. The present systematic review is focusing on the efficacy of stem cells to migrate at the lesion sites of the CNS and develop functional oligodendrocytes remyelinating axons. While most studies confirm the improvement of neurological deficits after the administration of different stem cell types, many critical issues need to be clarified before they can be efficiently introduced into clinical practice.
Collapse
Affiliation(s)
- Maria Veatriki Christodoulou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Ermioni Petkou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Natalia Atzemoglou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Eleni Gkorla
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Aikaterini Karamitrou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Yannis V Simos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Stefanos Bellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Chryssa Bekiari
- Laboratory of Anatomy and Histology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Panos Kouklis
- Laboratory of Biology, Department of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Konstantinos I Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.
- Department of Neurology, University Hospital of Ioannina, Ioannina, Greece.
| |
Collapse
|
16
|
Ghosh S, Bhatti GK, Sharma PK, Kandimalla R, Mastana SS, Bhatti JS. Potential of Nano-Engineered Stem Cells in the Treatment of Multiple Sclerosis: A Comprehensive Review. Cell Mol Neurobiol 2023; 44:6. [PMID: 38104307 PMCID: PMC11397842 DOI: 10.1007/s10571-023-01434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023]
Abstract
Multiple sclerosis (MS) is a chronic and degrading autoimmune disorder mainly targeting the central nervous system, leading to progressive neurodegeneration, demyelination, and axonal damage. Current treatment options for MS are limited in efficacy, generally linked to adverse side effects, and do not offer a cure. Stem cell therapies have emerged as a promising therapeutic strategy for MS, potentially promoting remyelination, exerting immunomodulatory effects and protecting against neurodegeneration. Therefore, this review article focussed on the potential of nano-engineering in stem cells as a therapeutic approach for MS, focusing on the synergistic effects of combining stem cell biology with nanotechnology to stimulate the proliferation of oligodendrocytes (OLs) from neural stem cells and OL precursor cells, by manipulating neural signalling pathways-PDGF, BMP, Wnt, Notch and their essential genes such as Sox, bHLH, Nkx. Here we discuss the pathophysiology of MS, the use of various types of stem cells in MS treatment and their mechanisms of action. In the context of nanotechnology, we present an overview of its applications in the medical and research field and discuss different methods and materials used to nano-engineer stem cells, including surface modification, biomaterials and scaffolds, and nanoparticle-based delivery systems. We further elaborate on nano-engineered stem cell techniques, such as nano script, nano-exosome hybrid, nano-topography and their potentials in MS. The article also highlights enhanced homing, engraftment, and survival of nano-engineered stem cells, targeted and controlled release of therapeutic agents, and immunomodulatory and tissue repair effects with their challenges and limitations. This visual illustration depicts the process of utilizing nano-engineering in stem cells and exosomes for the purpose of delivering more accurate and improved treatments for Multiple Sclerosis (MS). This approach targets specifically the creation of oligodendrocytes, the breakdown of which is the primary pathological factor in MS.
Collapse
Affiliation(s)
- Sushruta Ghosh
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University, Rajasthan, India
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University, Rajasthan, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
- Department of Applied Biology, CSIR-Indian Institute of Technology, Hyderabad, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India.
| |
Collapse
|
17
|
Kråkenes T, Wergeland S, Al-Sharabi N, Mohamed-Ahmed S, Fromreide S, Costea DE, Mustafa K, Bø L, Kvistad CE. The neuroprotective potential of mesenchymal stem cells from bone marrow and human exfoliated deciduous teeth in a murine model of demyelination. PLoS One 2023; 18:e0293908. [PMID: 37943848 PMCID: PMC10635499 DOI: 10.1371/journal.pone.0293908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/21/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is characterized by chronic inflammation, demyelination, and axonal degeneration within the central nervous system (CNS), for which there is no current treatment available with the ability to promote neuroprotection or remyelination. Some aspects of the progressive form of MS are displayed in the murine cuprizone model, where demyelination is induced by the innate immune system without major involvement of the adaptive immune system. Mesenchymal stem cells (MSCs) are multipotent cells with immunomodulatory and neuroprotective potential. In this study, we aimed to assess the neuroprotective potential of MSCs from bone marrow (BM-MSCs) and stem cells from human exfoliated deciduous teeth (SHED) in the cuprizone model. METHODS Human BM-MSCs and SHED were isolated and characterized. Nine-week-old female C57BL/6 mice were randomized to receive either human BM-MSCs, human SHED or saline intraperitoneally. Treatments were administered on day -1, 14 and 21. Outcomes included levels of local demyelination and inflammation, and were assessed with immunohistochemistry and histology. RESULTS BM-MSCs were associated with increased myelin content and reduced microglial activation whereas mice treated with SHED showed reduced microglial and astroglial activation. There were no differences between treatment groups in numbers of mature oligodendrocytes or axonal injury. MSCs were identified in the demyelinated corpus callosum in 40% of the cuprizone mice in both the BM-MSC and SHED group. CONCLUSION Our results suggest a neuroprotective effect of MSCs in a toxic MS model, with demyelination mediated by the innate immune system.
Collapse
Affiliation(s)
- Torbjørn Kråkenes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Niyaz Al-Sharabi
- Tissue Engineering Group, Center of Translational Oral Research (TOR), Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Samih Mohamed-Ahmed
- Tissue Engineering Group, Center of Translational Oral Research (TOR), Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Siren Fromreide
- Center for Cancer Biomarkers CCBIO and Gades Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Daniela-Elana Costea
- Center for Cancer Biomarkers CCBIO and Gades Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Kamal Mustafa
- Tissue Engineering Group, Center of Translational Oral Research (TOR), Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Lars Bø
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
18
|
Chen X, Wang Y, Ji J, Li C, Zhuang W, Luo J, Shi Y, Lin Q, Wu J, Li A, Wang J, Meng Y, Zhang S, Lang X, Liu X, Sun B, Li H, Liu Y. Electroacupuncture at ST36 acupoint regulates stem cells during experimental autoimmune encephalomyelitis. Int Immunopharmacol 2023; 124:110856. [PMID: 37647680 DOI: 10.1016/j.intimp.2023.110856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Electroacupuncture (EA) is given to assist in the treatment of MS, which is an effective therapeutic method. However, the therapy mechanism of EA related to stem cells in the treatment of MS is not yet known. In this study, we used a classic animal model of multiple sclerosis: experimental autoimmune encephalomyelitis (EAE) to evaluate the therapeutic effect of EA at Zusanli (ST36) acupoint in EAE and shed light on its potential roles in the effects of stem cells in vivo. METHODS The EAE animal models were established. From the first day after immunization, EAE model mice received EA at ST36 acupoint, named the EA group. The weight and clinical score of the three groups were recorded for 28 days. The demyelination, inflammatory cell infiltration, and markers of neural stem cells (NSCs), hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs) were compared. RESULTS We showed that EAE mice treated with EA at ST36 acupoint, were suppressed in demyelination and inflammatory cell infiltration, and thus decreased clinical score and weight loss and mitigated the development of EAE when compared with the EAE group. Moreover, our data revealed that the proportions of NSCs, HSCs, and MSCs increased in the EA group compared with the EAE group. CONCLUSIONS Our study suggested that EA at ST36 acupoint was an effective nonpharmacological therapeutic protocol that not only reduced the CNS demyelination and inflammatory cell infiltration in EAE disease but also increased the proportions of various stem cells. Further study is necessary to better understand how EA at the ST36 acupoint affects EAE.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yanping Wang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jiayu Ji
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Changyu Li
- Department of Neurosurgery, Hainan Cancer Hospital, Haikou, China
| | - Wei Zhuang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jingyu Luo
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yu Shi
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Qian Lin
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Junfeng Wu
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Anqi Li
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jing Wang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yanting Meng
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Xiujuan Lang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University, Harbin, China.
| |
Collapse
|
19
|
Wu D, Jin Y, Xing Y, Abate MD, Abbasian M, Abbasi-Kangevari M, Abbasi-Kangevari Z, Abd-Allah F, Abdelmasseh M, Abdollahifar MA, Abdulah DM, Abedi A, Abedi V, Abidi H, Aboagye RG, Abolhassani H, Abuabara K, Abyadeh M, Addo IY, Adeniji KN, Adepoju AV, Adesina MA, Sakilah Adnani QE, Afarideh M, Aghamiri S, Agodi A, Agrawal A, Aguilera Arriagada CE, Ahmad A, Ahmad D, Ahmad S, Ahmad S, Ahmadi A, Ahmed A, Ahmed A, Aithala JP, Ajadi AA, Ajami M, Akbarzadeh-Khiavi M, Alahdab F, AlBataineh MT, Alemi S, Saeed Al-Gheethi AA, Ali L, Alif SM, Almazan JU, Almustanyir S, Alqahtani JS, Alqasmi I, Khan Altaf IU, Alvis-Guzman N, Alvis-Zakzuk NJ, Al-Worafi YM, Aly H, Amani R, Amu H, Amusa GA, Andrei CL, Ansar A, Ansariniya H, Anyasodor AE, Arabloo J, Arefnezhad R, Arulappan J, Asghari-Jafarabadi M, Ashraf T, Atata JA, Athari SS, Atlaw D, Wahbi Atout MM, Aujayeb A, Awan AT, Ayatollahi H, Azadnajafabad S, Azzam AY, Badawi A, Badiye AD, Bagherieh S, Baig AA, Bantie BB, Barchitta M, Bardhan M, Barker-Collo SL, Barone-Adesi F, Batra K, Bayileyegn NS, Behnoush AH, Belgaumi UI, Bemanalizadeh M, Bensenor IM, Beyene KA, Bhagavathula AS, Bhardwaj P, Bhaskar S, Bhat AN, Bitaraf S, Bitra VR, Boloor A, Bora K, Botelho JS, et alWu D, Jin Y, Xing Y, Abate MD, Abbasian M, Abbasi-Kangevari M, Abbasi-Kangevari Z, Abd-Allah F, Abdelmasseh M, Abdollahifar MA, Abdulah DM, Abedi A, Abedi V, Abidi H, Aboagye RG, Abolhassani H, Abuabara K, Abyadeh M, Addo IY, Adeniji KN, Adepoju AV, Adesina MA, Sakilah Adnani QE, Afarideh M, Aghamiri S, Agodi A, Agrawal A, Aguilera Arriagada CE, Ahmad A, Ahmad D, Ahmad S, Ahmad S, Ahmadi A, Ahmed A, Ahmed A, Aithala JP, Ajadi AA, Ajami M, Akbarzadeh-Khiavi M, Alahdab F, AlBataineh MT, Alemi S, Saeed Al-Gheethi AA, Ali L, Alif SM, Almazan JU, Almustanyir S, Alqahtani JS, Alqasmi I, Khan Altaf IU, Alvis-Guzman N, Alvis-Zakzuk NJ, Al-Worafi YM, Aly H, Amani R, Amu H, Amusa GA, Andrei CL, Ansar A, Ansariniya H, Anyasodor AE, Arabloo J, Arefnezhad R, Arulappan J, Asghari-Jafarabadi M, Ashraf T, Atata JA, Athari SS, Atlaw D, Wahbi Atout MM, Aujayeb A, Awan AT, Ayatollahi H, Azadnajafabad S, Azzam AY, Badawi A, Badiye AD, Bagherieh S, Baig AA, Bantie BB, Barchitta M, Bardhan M, Barker-Collo SL, Barone-Adesi F, Batra K, Bayileyegn NS, Behnoush AH, Belgaumi UI, Bemanalizadeh M, Bensenor IM, Beyene KA, Bhagavathula AS, Bhardwaj P, Bhaskar S, Bhat AN, Bitaraf S, Bitra VR, Boloor A, Bora K, Botelho JS, Buchbinder R, Calina D, Cámera LA, Carvalho AF, Kai Chan JS, Chattu VK, Abebe EC, Chichagi F, Choi S, Chou TC, Chu DT, Coberly K, Costa VM, Couto RA, Cruz-Martins N, Dadras O, Dai X, Damiani G, Dascalu AM, Dashti M, Debela SA, Dellavalle RP, Demetriades AK, Demlash AA, Deng X, Desai HD, Desai R, Rahman Dewan SM, Dey S, Dharmaratne SD, Diaz D, Dibas M, Dinis-Oliveira RJ, Diress M, Do TC, Doan DK, Dodangeh M, Dodangeh M, Dongarwar D, Dube J, Dziedzic AM, Ed-Dra A, Edinur HA, Eissazade N, Ekholuenetale M, Ekundayo TC, Elemam NM, Elhadi M, Elmehrath AO, Abdou Elmeligy OA, Emamverdi M, Emeto TI, Esayas HL, Eshetu HB, Etaee F, Fagbamigbe AF, Faghani S, Fakhradiyev IR, Fatehizadeh A, Fathi M, Feizkhah A, Fekadu G, Fereidouni M, Fereshtehnejad SM, Fernandes JC, Ferrara P, Fetensa G, Filip I, Fischer F, Foroutan B, Foroutan M, Fukumoto T, Ganesan B, Belete Gemeda BN, Ghamari SH, Ghasemi M, Gholamalizadeh M, Gill TK, Gillum RF, Goldust M, Golechha M, Goleij P, Golinelli D, Goudarzi H, Guan SY, Guo Y, Gupta B, Gupta VB, Gupta VK, Haddadi R, Hadi NR, Halwani R, Haque S, Hasan I, Hashempour R, Hassan A, Hassan TS, Hassanzadeh S, Hassen MB, Haubold J, Hayat K, Heidari G, Heidari M, Heidari-Soureshjani R, Herteliu C, Hessami K, Hezam K, Hiraike Y, Holla R, Hosseini MS, Huynh HH, Hwang BF, Ibitoye SE, Ilic IM, Ilic MD, Iranmehr A, Iravanpour F, Ismail NE, Iwagami M, Iwu CC, Jacob L, Jafarinia M, Jafarzadeh A, Jahankhani K, Jahrami H, Jakovljevic M, Jamshidi E, Jani CT, Janodia MD, Jayapal SK, Jayaram S, Jeganathan J, Jonas JB, Joseph A, Joseph N, Joshua CE, Vaishali K, Kaambwa B, Kabir A, Kabir Z, Kadashetti V, Kaliyadan F, Kalroozi F, Kamal VK, Kandel A, Kandel H, Kanungo S, Karami J, Karaye IM, Karimi H, Kasraei H, Kazemian S, Kebede SA, Keikavoosi-Arani L, Keykhaei M, Khader YS, Khajuria H, Khamesipour F, Khan EA, Khan IA, Khan M, Khan MJ, Khan MA, Khan MA, Khatatbeh H, Khatatbeh MM, Khateri S, Khayat Kashani HR, Kim MS, Kisa A, Kisa S, Koh HY, Kolkhir P, Korzh O, Kotnis AL, Koul PA, Koyanagi A, Krishan K, Kuddus M, Kulkarni VV, Kumar N, Kundu S, Kurmi OP, La Vecchia C, Lahariya C, Laksono T, Lám J, Latief K, Lauriola P, Lawal BK, Thu Le TT, Bich Le TT, Lee M, Lee SW, Lee WC, Lee YH, Lenzi J, Levi M, Li W, Ligade VS, Lim SS, Liu G, Liu X, Llanaj E, Lo CH, Machado VS, Maghazachi AA, Mahmoud MA, Mai TA, Majeed A, Sanaye PM, Makram OM, Rad EM, Malhotra K, Malik AA, Malik I, Mallhi TH, Malta DC, Mansournia MA, Mantovani LG, Martorell M, Masoudi S, Masoumi SZ, Mathangasinghe Y, Mathews E, Mathioudakis AG, Maugeri A, Mayeli M, Carabeo Medina JR, Meles GG, Mendes JJ, Menezes RG, Mestrovic T, Michalek IM, Micheletti Gomide Nogueira de Sá AC, Mihretie ET, Nhat Minh LH, Mirfakhraie R, Mirrakhimov EM, Misganaw A, Mohamadkhani A, Mohamed NS, Mohammadi F, Mohammadi S, Mohammed S, Mohammed S, Mohan S, Mohseni A, Mokdad AH, Momtazmanesh S, Monasta L, Moni MA, Moniruzzaman M, Moradi Y, Morovatdar N, Mostafavi E, Mousavi P, Mukoro GD, Mulita A, Mulu GB, Murillo-Zamora E, Musaigwa F, Mustafa G, Muthu S, Nainu F, Nangia V, Swamy SN, Natto ZS, Navaraj P, Nayak BP, Nazri-Panjaki A, Negash H, Nematollahi MH, Nguyen DH, Hien Nguyen HT, Nguyen HQ, Nguyen PT, Nguyen VT, Niazi RK, Nikolouzakis TK, Nnyanzi LA, Noreen M, Nzoputam CI, Nzoputam OJ, Oancea B, Oh IH, Okati-Aliabad H, Okonji OC, Okwute PG, Olagunju AT, Olatubi MI, Olufadewa II, Ordak M, Otstavnov N, Owolabi MO, Mahesh P, Padubidri JR, Pak A, Pakzad R, Palladino R, Pana A, Pantazopoulos I, Papadopoulou P, Pardhan S, Parthasarathi A, Pashaei A, Patel J, Pathan AR, Patil S, Paudel U, Pawar S, Pedersini P, Pensato U, Pereira DM, Pereira J, Pereira MO, Pereira RB, Peres MF, Perianayagam A, Perna S, Petcu IR, Pezeshki PS, Pham HT, Philip AK, Piradov MA, Podder I, Podder V, Poddighe D, Sady Prates EJ, Qattea I, Radfar A, Raee P, Rafiei A, Raggi A, Rahim F, Rahimi M, Rahimifard M, Rahimi-Movaghar V, Rahman MO, Ur Rahman MH, Rahman M, Rahman MA, Rahmani AM, Rahmani M, Rahmani S, Rahmanian V, Ramasubramani P, Rancic N, Rao IR, Rashedi S, Rashid AM, Ravikumar N, Rawaf S, Mohamed Redwan EM, Rezaei N, Rezaei N, Rezaei N, Rezaeian M, Ribeiro D, Rodrigues M, Buendia Rodriguez JA, Roever L, Romero-Rodríguez E, Saad AM, Saddik B, Sadeghian S, Saeed U, Safary A, Safdarian M, Safi SZ, Saghazadeh A, Sagoe D, Sharif-Askari FS, Sharif-Askari NS, Sahebkar A, Sahoo H, Sahraian MA, Sajid MR, Sakhamuri S, Sakshaug JW, Saleh MA, Salehi L, Salehi S, Farrokhi AS, Samadzadeh S, Samargandy S, Samieefar N, Samy AM, Sanadgol N, Sanjeev RK, Sawhney M, Saya GK, Schuermans A, Senthilkumaran S, Sepanlou SG, Sethi Y, Shafie M, Shah H, Shahid I, Shahid S, Shaikh MA, Sharfaei S, Sharma M, Shayan M, Shehata HS, Sheikh A, Shetty JK, Shin JI, Shirkoohi R, Shitaye NA, Shivakumar K, Shivarov V, Shobeiri P, Siabani S, Sibhat MM, Siddig EE, Simpson CR, Sinaei E, Singh H, Singh I, Singh JA, Singh P, Singh S, Siraj MS, Al Mamun Sohag A, Solanki R, Solikhah S, Solomon Y, Soltani-Zangbar MS, Sun J, Szeto MD, Tabarés-Seisdedos R, Tabatabaei SM, Tabish M, Taheri E, Tahvildari A, Talaat IM, Lukenze Tamuzi JJ, Tan KK, Tat NY, Oliaee RT, Tavasol A, Temsah MH, Thangaraju P, Tharwat S, Tibebu NS, Vera Ticoalu JH, Tillawi T, Tiruye TY, Tiyuri A, Tovani-Palone MR, Tripathi M, Tsegay GM, Tualeka AR, Ty SS, Ubah CS, Ullah S, Ullah S, Umair M, Umakanthan S, Upadhyay E, Vahabi SM, Vaithinathan AG, Tahbaz SV, Valizadeh R, Varthya SB, Vasankari TJ, Venketasubramanian N, Verras GI, Villafañe JH, Vlassov V, Vo DC, Waheed Y, Waris A, Welegebrial BG, Westerman R, Wickramasinghe DP, Wickramasinghe ND, Willekens B, Woldegeorgis BZ, Woldemariam M, Xiao H, Yada DY, Yahya G, Yang L, Yazdanpanah F, Yon DK, Yonemoto N, You Y, Zahir M, Zaidi SS, Zangiabadian M, Zare I, Zeineddine MA, Zemedikun DT, Zeru NG, Zhang C, Zhao H, Zhong C, Zielińska M, Zoladl M, Zumla A, Guo C, Tam LS. Global, regional, and national incidence of six major immune-mediated inflammatory diseases: findings from the global burden of disease study 2019. EClinicalMedicine 2023; 64:102193. [PMID: 37731935 PMCID: PMC10507198 DOI: 10.1016/j.eclinm.2023.102193] [Show More Authors] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND The causes for immune-mediated inflammatory diseases (IMIDs) are diverse and the incidence trends of IMIDs from specific causes are rarely studied. The study aims to investigate the pattern and trend of IMIDs from 1990 to 2019. METHODS We collected detailed information on six major causes of IMIDs, including asthma, inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis, psoriasis, and atopic dermatitis, between 1990 and 2019, derived from the Global Burden of Disease study in 2019. The average annual percent change (AAPC) in number of incidents and age standardized incidence rate (ASR) on IMIDs, by sex, age, region, and causes, were calculated to quantify the temporal trends. FINDINGS In 2019, rheumatoid arthritis, atopic dermatitis, asthma, multiple sclerosis, psoriasis, inflammatory bowel disease accounted 1.59%, 36.17%, 54.71%, 0.09%, 6.84%, 0.60% of overall new IMIDs cases, respectively. The ASR of IMIDs showed substantial regional and global variation with the highest in High SDI region, High-income North America, and United States of America. Throughout human lifespan, the age distribution of incident cases from six IMIDs was quite different. Globally, incident cases of IMIDs increased with an AAPC of 0.68 and the ASR decreased with an AAPC of -0.34 from 1990 to 2019. The incident cases increased across six IMIDs, the ASR of rheumatoid arthritis increased (0.21, 95% CI 0.18, 0.25), while the ASR of asthma (AAPC = -0.41), inflammatory bowel disease (AAPC = -0.72), multiple sclerosis (AAPC = -0.26), psoriasis (AAPC = -0.77), and atopic dermatitis (AAPC = -0.15) decreased. The ASR of overall and six individual IMID increased with SDI at regional and global level. Countries with higher ASR in 1990 experienced a more rapid decrease in ASR. INTERPRETATION The incidence patterns of IMIDs varied considerably across the world. Innovative prevention and integrative management strategy are urgently needed to mitigate the increasing ASR of rheumatoid arthritis and upsurging new cases of other five IMIDs, respectively. FUNDING The Global Burden of Disease Study is funded by the Bill and Melinda Gates Foundation. The project funded by Scientific Research Fund of Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital (2022QN38).
Collapse
|
20
|
Kasarełło K, Seta M, Sulejczak D, Snarski E, Cudnoch-Jędrzejewska A. Effect of Hematopoietic Stem Cell Transplantation and Post-Transplantation Cyclophosphamide on the Microglia Phenotype in Rats with Experimental Allergic Encephalomyelitis. Arch Immunol Ther Exp (Warsz) 2023; 71:10. [PMID: 36964399 PMCID: PMC10039091 DOI: 10.1007/s00005-023-00675-y] [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: 10/02/2022] [Accepted: 02/16/2023] [Indexed: 03/26/2023]
Abstract
Microglia are the resident immune cells of the central nervous system, playing a role in the inflammatory process development and resolution, presenting two main phenotypes, pro-inflammatory M1, and anti-inflammatory M2. Therapies affecting the microglia phenotype may be beneficial in treating inflammatory neurodegenerative diseases. In our experiments, we used the animal multiple sclerosis model, experimental allergic encephalomyelitis (EAE). Rats were treated during the pre- or symptomatic phase of the disease with cyclophosphamide, followed by hematopoietic stem cell transplantation, and with/without post-transplantation cyclophosphamide. Our study aimed to analyze the microglia phenotype in animals subjected to this treatment. The number of M1 cells in the spinal cord, and inducible nitric oxide synthase (iNOS) levels in the brain were similar in all experimental groups. The differences were observed in M2 cells number and arginase 1 (Arg1) levels, which were decreased in EAE animals, and increased after treatment in the symptomatic phase of EAE, and in the pre-symptomatic phase, but only with post-transplantation cyclophosphamide. Analysis of gene expression in the brain showed decreased iNOS expression in EAE animals treated in the symptomatic phase of EAE and no differences in Arg1 expression. Results indicate that treatment applied to experimental animals influences the microglia phenotype, promoting differentiation towards M2 cells.
Collapse
Affiliation(s)
- Kaja Kasarełło
- Chair and Department of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Martyna Seta
- Chair and Department of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Emilian Snarski
- Chair and Department of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
21
|
Cohen JA, Lublin FD, Lock C, Pelletier D, Chitnis T, Mehra M, Gothelf Y, Aricha R, Lindborg S, Lebovits C, Levy Y, Motamed Khorasani A, Kern R. Evaluation of neurotrophic factor secreting mesenchymal stem cells in progressive multiple sclerosis. Mult Scler 2023; 29:92-106. [PMID: 36113170 PMCID: PMC9896300 DOI: 10.1177/13524585221122156] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autologous mesenchymal stem cell neurotrophic factor-secreting cells (NurOwn®) have the potential to modify underlying disease mechanisms in progressive multiple sclerosis (PMS). OBJECTIVE This open-label phase II study was conducted to evaluate safety/efficacy of three intrathecal cell treatments. METHODS Eighteen participants with non-relapsing PMS were treated. The primary endpoint was safety. Secondary endpoints included: cerebrospinal fluid (CSF) biomarkers; timed 25-foot walk speed, nine-hole peg test (9-HPT), low-contrast letter acuity, symbol digit modalities test, and 12-item multiple sclerosis (MS) walking scale. Seventeen participants received all treatments. RESULTS No deaths/adverse events related to worsening of MS, clinical/magnetic resonance imaging (MRI) evidence of disease activation, and clinically significant changes in safety lab results were reported. Two participants developed symptoms of low back and leg pain, consistent with a diagnosis of arachnoiditis, occurring in one of three intrathecal treatments in both participants. Nineteen percent of treated participants achieved pre-specified ⩾ 25% improvements in timed 25-foot walk speed/nine-HPT at 28 weeks compared to baseline, along with consistent efficacy signals for pre-specified response criteria across other secondary efficacy outcomes. CSF neuroprotective factors increased, and inflammatory biomarkers decreased after treatment, consistent with the proposed mechanism of action. CONCLUSION Based on these encouraging preliminary findings, further confirmation in a randomized study is warranted.
Collapse
Affiliation(s)
- Jeffrey A Cohen
- JA Cohen Department of Neurology, Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | - Fred D Lublin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christoper Lock
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Daniel Pelletier
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Tanuja Chitnis
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Munish Mehra
- Department of Statistics, Tigermed, Somerset, NJ, USA
| | - Yael Gothelf
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| | - Revital Aricha
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| | - Stacy Lindborg
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| | - Chaim Lebovits
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| | - Yossef Levy
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| | - Afsaneh Motamed Khorasani
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
- Department of Medical Affairs, Eonian Stanzas LLC, Potomac, MD, USA
| | - Ralph Kern
- Department of Research & Development, Brainstorm Cell Therapeutics, New York, NY, USA
| |
Collapse
|
22
|
Damavandi AR, Mirmosayyeb O, Ebrahimi N, Zalpoor H, khalilian P, Yahiazadeh S, Eskandari N, Rahdar A, Kumar PS, Pandey S. Advances in nanotechnology versus stem cell therapy for the theranostics of multiple sclerosis disease. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02698-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Marzban M, Rustamzadeh A, Asghari A, Terme Y, Amichi AG, Ghanbarzehi V, Holaso AS, Hosseini F, Shahraki M, Sadafi P, Hashemzahi E, Honardar M, Iravankhah M, Baloochi M, Yarmohammadi A, Ebrahimi P. Stem cell therapy for cuprizone model of multiple sclerosis focusing on the effectiveness of different injection methods and cell labeling. Acta Histochem 2022; 124:151953. [PMID: 36116321 DOI: 10.1016/j.acthis.2022.151953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/01/2022]
Abstract
Multiple Sclerosis (MS) is a chronic and autoimmune disease of the central nervous system that causes inflammation in the brain and spinal cord, progressive degeneration of central nervous system tissue, damage to neuronal axons, and loss of function of central nervous system neurons. Experimental encephalomyelitis is an alternative animal model of MS that can simulate the symptoms of this disease. Cuprizone is one of the factors creating this model. Various researchers are testing the use of different cells to reduce the symptoms of cuprizone-demyelinated mice. The different injection methods explained in this article include intracerebral, intraperitoneal, intravenous, and intranasal. The intracerebral method, in contrast to the intranasal method, was widely employed by researchers. In each technique, the researchers try to inject a specific type of stem cell (SC) and monitor their efficiency. For monitoring SCs various labeling procedures are available, however, there is an upward trend in using magnetic resonance imaging (MRI). Two main barriers to using this method are high cost and complexity. In the current review, we try to make review cell therapy in the cuprizone model of MS.
Collapse
Affiliation(s)
- Mohsen Marzban
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aria Asghari
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Yousef Terme
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Vahid Ghanbarzehi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Fateme Hosseini
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mahya Shahraki
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Paniz Sadafi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Erfan Hashemzahi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Minoo Honardar
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Marziyeh Iravankhah
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mehdi Baloochi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amin Yarmohammadi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Pirooz Ebrahimi
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria, Italy
| |
Collapse
|
24
|
Brod SA. The genealogy, methodology, similarities and differences of immune reconstitution therapies for multiple sclerosis and neuromyelitis optica. Autoimmun Rev 2022; 21:103170. [PMID: 35963569 DOI: 10.1016/j.autrev.2022.103170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 11/09/2022]
Abstract
Immune reconstitution therapies (IRTs) are a type of short course procedure or pharmaceutical agent within the MS pharmacopeia. They emanate from oncology and induce transient incomplete lympho-ablation with or without myelo-ablation, resulting in potential prolonged immunomodulation. Thus, they provide significant prophylaxis from disease activity without retreatment. Modern IRT for autoimmunity encompasses a heterogeneous group of pulsed lympho- and non-myelo-ablative treatments designed to re-boot the adaptive immune system in a quasi-permanent manner - a re-induction of ontogeny. IRT is the extensive debulking of an auto-aggressive immune system to attempt to reach the Holy Grail of immune tolerance. This incomplete yet significant lympho-ablation induces lymphoproliferation, reduces pathogenic clonal cells, causes thymopoiesis and results in the induction of immune tolerance. Lympho-ablation with immune reconstitution can result in minimal residual autoimmunity. There is a resetting of the immune thermostat - i.e., the immunostat. IRTs have the potential to provide prolonged periods of disease inactivity without retreatment in part through the immunological results of their pulsatile lymphocyte depletion. It is vital to increase our understanding of how IRTs alter a patient's immune response to the antigenic target of the disease so that we can devise newer, more durable and safer forms of such agents. What common features do extant IRTs (i.e., stem cell transplant, alemtuzumab and oral cladribine) have to produce the durable therapeutic response without long term treatment in neuroimmunological diseases such as MS (multiple sclerosis) and NMOSD (neuromyelitis optica spectrum disorders)? Can we learn from these critical features to predict what other maneuvers or agents might effect similar clinical results with equal or greater efficacy and safety?
Collapse
Affiliation(s)
- Staley A Brod
- Division of MS/Neuro-immunology, Department of Neurology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
25
|
Thamm K, Möbus K, Towers R, Baertschi S, Wetzel R, Wobus M, Segeletz S. A chemically defined biomimetic surface for enhanced isolation efficiency of high-quality human mesenchymal stromal cells under xenogeneic/serum-free conditions. Cytotherapy 2022; 24:1049-1059. [PMID: 35931601 DOI: 10.1016/j.jcyt.2022.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/23/2022] [Accepted: 06/10/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are one of the most frequently used cell types in regenerative medicine and cell therapy. Generating sufficient cell numbers for MSC-based therapies is constrained by (i) their low abundance in tissues of origin, which imposes the need for significant ex vivo cell expansion; (ii) donor-specific characteristics, including MSC frequency/quality, that decline with disease state and increasing age; and (iii) cellular senescence, which is promoted by extensive cell expansion and results in decreased therapeutic functionality. The final yield of a manufacturing process is therefore primarily determined by the applied isolation procedure and its efficiency in isolating therapeutically active cells from donor tissue. To date, MSCs are predominantly isolated using media supplemented with either serum or its derivatives, which poses safety and consistency issues. METHODS To overcome these limitations while enabling robust MSC production with constant high yield and quality, the authors developed a chemically defined biomimetic surface coating called isoMATRIX (denovoMATRIX GmbH, Dresden, Germany) and tested its performance during isolation of MSCs. RESULTS The isoMATRIX facilitates the isolation of significantly higher numbers of MSCs in xenogeneic (xeno)/serum-free and chemically defined conditions. The isolated cells display a smaller cell size and higher proliferation rate than those derived from a serum-containing isolation procedure and a strong immunomodulatory capacity. The high proliferation rates can be maintained up to 5 passages after isolation and cells even benefit from a switch towards a proliferation-specific MSC matrix (myMATRIX MSC) (denovoMATRIX GmbH, Dresden, Germany). CONCLUSION In sum, isoMATRIX promotes enhanced xeno/serum-free and chemically defined isolation of human MSCs and supports consistent and reliable cell performance for improved stem cell-based therapies.
Collapse
Affiliation(s)
| | - Kristin Möbus
- Universitätskrankenhaus Carl Gustav Carus der Technischen Universität Dresden, Dresden, Germany
| | - Russell Towers
- Universitätskrankenhaus Carl Gustav Carus der Technischen Universität Dresden, Dresden, Germany
| | | | | | - Manja Wobus
- Universitätskrankenhaus Carl Gustav Carus der Technischen Universität Dresden, Dresden, Germany
| | | |
Collapse
|
26
|
Kvistad SAS, Burman J, Lehmann AK, Tolf A, Zjukovskaja C, Melve GK, Bø L, Torkildsen Ø. Impact of previous disease-modifying treatment on safety and efficacy in patients with MS treated with AHSCT. J Neurol Neurosurg Psychiatry 2022; 93:844-848. [PMID: 35508373 PMCID: PMC9304086 DOI: 10.1136/jnnp-2022-328797] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Autologous haematopoietic stem cell transplantation (AHSCT) is a highly effective treatment for multiple sclerosis (MS). The impact of previous long-lasting disease-modifying treatments (DMT) for safety and efficacy of AHSCT is unknown. OBJECTIVE To explore whether previous DMTs with long-lasting effects on the immune system (anti-CD20 therapy, alemtuzumab and cladribine) affect treatment-related complications, long-term outcome and risk of new MS disease activity in patients treated with AHSCT. METHODS Retrospective observational study of 104 relapsing remitting patients with MS treated by AHSCT in Sweden and Norway from 2011 to 2021, grouped according to the last DMT used ≤6 months prior to AHSCT. The primary outcomes were early AHSCT-related complications (mortality, neutropenic fever and hospitalisation length), long-term complications (secondary autoimmunity) and proportion of patients with No Evidence of Disease Activity (NEDA-3 status): no new relapses, no MRI activity and no disease progression during the follow-up. RESULTS The mean follow-up time was 39.5 months (range 1-95). Neutropenic fever was a common AHSCT-related complication affecting 69 (66%) patients. There was no treatment-related mortality. During the follow-up period, 20 patients (19%) were diagnosed with autoimmunity. Occurrence of neutropenic fever, hospitalisation length or secondary autoimmunity did not vary dependent on the last DMT used prior to AHSCT. A total of 84 patients (81%) achieved NEDA-3 status, including all patients (100%) using rituximab, alemtuzumab or cladribine before AHSCT. CONCLUSION This study provides level 4 evidence that AHSCT in patients previously treated with alemtuzumab, cladribine or rituximab is safe and efficacious.
Collapse
Affiliation(s)
- Silje Agnethe Stokke Kvistad
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway .,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Joachim Burman
- Department of Neuroscience, Uppsala Universitet, Uppsala, Sweden
| | - Anne Kristine Lehmann
- Department of Medicine, Section of Hematology, Haukeland University Hospital, Bergen, Norway
| | - Andreas Tolf
- Department of Neuroscience, Uppsala Universitet, Uppsala, Sweden.,Department of Neurology, Akademiska sjukhuset, Uppsala, Sweden
| | | | - Guro Kristin Melve
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lars Bø
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Norwegian Multiple Sclerosis Competence Centre, Haukeland University Hospital, Bergen, Norway
| | - Øivind Torkildsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Neurology, Norwegian Multiple Sclerosis Competence Centre, Bergen, Norway
| |
Collapse
|
27
|
Karussis D, Kassis I, Petrou P. Therapy with mesenchymal stem cell transplantation in multiple sclerosis is ready for prime time: YES. Mult Scler 2022; 28:1324-1326. [PMID: 35786042 DOI: 10.1177/13524585211062173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Dimitrios Karussis
- Multiple Sclerosis Center and Neuroimmunology Unit, The Hadassah University Hospital-Ein Kerem, Jerusalem, Israel
| | - Ibrahim Kassis
- Multiple Sclerosis Center and Neuroimmunology Unit, The Hadassah University Hospital-Ein Kerem, Jerusalem, Israel
| | - Panayiota Petrou
- Multiple Sclerosis Center and Neuroimmunology Unit, The Hadassah University Hospital-Ein Kerem, Jerusalem, Israel
| |
Collapse
|
28
|
Gureev AP, Samoylova NA, Potanina DV, Popov VN. The Effect of Methylene Blue and Its Metabolite—Azure I—on Bioenergetic Parameters of Intact Mouse Brain Mitochondria. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2022; 16:148-153. [PMID: 35601460 PMCID: PMC9113384 DOI: 10.1134/s1990750822020044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Methylene blue, a phenothiazine dye, that is widely used in medicine and is under clinical trials as an agent for treatment of Alzheimer’s disease. One of the factors of the unique therapeutic effect of methylene blue is its redox properties, allowing implementation of alternative electron transport: the dye accepts electrons from reducing equivalents in mitochondria and transfer them to other components of the respiratory chain or molecular oxygen. Azure I, an N-dimethylated metabolite of methylene blue, is potentially a more effective compound than methylene blue, but its ability for alternative electron transport has not been studied yet. We have shown that in contrast to methylene blue, azure I is unable to restore the membrane potential in isolated mouse brain mitochondria, inhibited by rotenone and, therefore, is unable to perform bypass of the respiratory chain complex I. Moreover, addition of azure I does not affect the rate of mitochondrial respiration in contrast to methylene blue, which increases the rate of non-phosphorylation respiration. At the same time, both dyes stimulate an increase in H2O2 production. Thus, only methylene blue is capable of alternative electron transport, while azure I does not produce complex I bypass. This limits its therapeutic application only as a mitochondrial-targeted agent, but does not question its antidepressant effects.
Collapse
Affiliation(s)
- A. P. Gureev
- Voronezh State University, Universitetskaya pl. 1, 394018 Voronezh, Russia
- Voronezh State University of Engineering Technologies, pr. Revolyutsii 19, 394036 Voronezh, Russia
| | - N. A. Samoylova
- Voronezh State University, Universitetskaya pl. 1, 394018 Voronezh, Russia
| | - D. V. Potanina
- Voronezh State University, Universitetskaya pl. 1, 394018 Voronezh, Russia
| | - V. N. Popov
- Voronezh State University, Universitetskaya pl. 1, 394018 Voronezh, Russia
- Voronezh State University of Engineering Technologies, pr. Revolyutsii 19, 394036 Voronezh, Russia
| |
Collapse
|
29
|
Sreter JA, Foxall TL, Varga K. Intracellular and Extracellular Antifreeze Protein Significantly Improves Mammalian Cell Cryopreservation. Biomolecules 2022; 12:669. [PMID: 35625597 PMCID: PMC9139014 DOI: 10.3390/biom12050669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 12/03/2022] Open
Abstract
Cell cryopreservation is an essential part of the biotechnology, food, and health care industries. There is a need to develop more effective, less toxic cryoprotective agents (CPAs) and methods, especially for mammalian cells. We investigated the impact of an insect antifreeze protein from Anatolica polita (ApAFP752) on mammalian cell cryopreservation using the human embryonic kidney cell line HEK 293T. An enhanced green fluorescent protein (EGFP)-tagged antifreeze protein, EGFP-ApAFP752, was transfected into the cells and the GFP was used to determine the efficiency of transfection. AFP was assessed for its cryoprotective effects intra- and extracellularly and both simultaneously at different concentrations with and without dimethyl sulfoxide (DMSO) at different concentrations. Comparisons were made to DMSO or medium alone. Cells were cryopreserved at -196 °C for ≥4 weeks. Upon thawing, cellular viability was determined using trypan blue, cellular damage was assessed by lactate dehydrogenase (LDH) assay, and cellular metabolism was measured using a metabolic activity assay (MTS). The use of this AFP significantly improved cryopreserved cell survival when used with DMSO intracellularly. Extracellular AFP also significantly improved cell survival when included in the DMSO freezing medium. Intra- and extracellular AFP used together demonstrated the most significantly increased cryoprotection compared to DMSO alone. These findings present a potential method to improve the viability of cryopreserved mammalian cells.
Collapse
Affiliation(s)
- Jonathan A. Sreter
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA;
| | - Thomas L. Foxall
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA;
| | - Krisztina Varga
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA;
| |
Collapse
|
30
|
Liu H, Chen B, Zhu Q. Potential application of hydrogel to the diagnosis and treatment of multiple sclerosis. J Biol Eng 2022; 16:10. [PMID: 35395765 PMCID: PMC8991948 DOI: 10.1186/s13036-022-00288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/12/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system. This disorder may cause progressive and permanent impairment, placing significant physical and psychological strain on sufferers. Each progress in MS therapy marks a significant advancement in neurological research. Hydrogels can serve as a scaffold with high water content, high expansibility, and biocompatibility to improve MS cell proliferation in vitro and therapeutic drug delivery to cells in vivo. Hydrogels may also be utilized as biosensors to detect MS-related proteins. Recent research has employed hydrogels as an adjuvant imaging agent in immunohistochemistry assays. Following an overview of the development and use of hydrogels in MS diagnostic and therapy, this review discussed hydrogel’s advantages and future opportunities in the diagnosis and treatment of MS. Graphical abstract ![]()
Collapse
Affiliation(s)
- Haochuan Liu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China
| | - Bing Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| | - Qingsan Zhu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| |
Collapse
|
31
|
Rice CM, Sarkar P, Walsh P, Owen D, Bidgood C, Smith P, Kane NM, Asghar S, Marks DI, Scolding NJ. Repeat infusion of autologous bone marrow cells in progressive multiple sclerosis - A phase I extension study (SIAMMS II). Mult Scler Relat Disord 2022; 61:103782. [PMID: 35397289 DOI: 10.1016/j.msard.2022.103782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND During the safety and feasibility 'Study of Intravenous Autologous Marrow in Multiple Sclerosis (SIAMMS)', intravenous infusion of autologous marrow was well tolerated. The efficacy of the approach is being explored in a placebo-controlled randomised controlled trial (ACTiMuS, NCT01815632) but it is not known whether repeated infusions will be required to optimise benefit. The objective of the current study was to explore the safety and feasibility of repeat treatment with intravenous autologous bone marrow for patients with progressive multiple sclerosis (MS). METHODS 'SIAMMS II' was a prospective, single centre phase I extension study in which participants in the SIAMMS study were offered repeat bone marrow harvest and infusion of autologous, unfractionated bone marrow as a day-case procedure. The primary outcome measure was number of adverse events and secondary outcome measures included change in clinical rating scales of disability, global evoked potential and cranial magnetic resonance imaging (MRI). RESULTS In total, 4 of the 6 participants in the SIAMMS study had repeat bone marrow harvest and infusion of filtered autologous marrow as a day case procedure which was well tolerated. There were no serious adverse effects. Additional outcome measures including clinical scales, global evoked potentials and cranial MRI were stable. CONCLUSION SIAMMS II demonstrates the safety and feasibility of repeated, non-myeloablative autologous bone marrow-derived cell therapy in progressive MS.
Collapse
Affiliation(s)
- Claire M Rice
- Clinical Neuroscience, Bristol Medical School, University of Bristol, Level 1 Learning and Research Building, Southmead Hospital, Bristol, BS10 5NBww, UK; Department of Neurology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK.
| | - Pamela Sarkar
- Clinical Neuroscience, Bristol Medical School, University of Bristol, Level 1 Learning and Research Building, Southmead Hospital, Bristol, BS10 5NBww, UK; Department of Neurology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Peter Walsh
- Department of Neurophysiology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Denise Owen
- Department of Neurology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Clare Bidgood
- Adult BMT Unit, Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust, St Michael's Hill, Bristol BS2 8BJ, UK
| | - Paul Smith
- Department of Neuroradiology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Nick M Kane
- Department of Neurophysiology, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Suhail Asghar
- NHS Blood and Transplant, North Bristol Park, Filton, Bristol, UK
| | - David I Marks
- Clinical Neuroscience, Bristol Medical School, University of Bristol, Level 1 Learning and Research Building, Southmead Hospital, Bristol, BS10 5NBww, UK; Adult BMT Unit, Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust, St Michael's Hill, Bristol BS2 8BJ, UK
| | - Neil J Scolding
- Clinical Neuroscience, Bristol Medical School, University of Bristol, Level 1 Learning and Research Building, Southmead Hospital, Bristol, BS10 5NBww, UK
| |
Collapse
|
32
|
The Integration of Cell Therapy and Biomaterials as Treatment Strategies for Remyelination. Life (Basel) 2022; 12:life12040474. [PMID: 35454965 PMCID: PMC9027199 DOI: 10.3390/life12040474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic degenerative autoimmune disease of the central nervous system that causes inflammation, demyelinating lesions, and axonal damage and is associated with a high rate of early-onset disability. Disease-modifying therapies are used to mitigate the inflammatory process in MS but do not promote regeneration or remyelination; cell therapy may play an important role in these processes, modulating inflammation and promoting the repopulation of oligodendrocytes, which are responsible for myelin repair. The development of genetic engineering has led to the emergence of stable, biocompatible biomaterials that may promote a favorable environment for exogenous cells. This review summarizes the available evidence about the effects of transplantation of different types of stem cells reported in studies with several animal models of MS and clinical trials in human patients. We also address the advantages of combining cell therapy with biomaterials.
Collapse
|
33
|
Bebo BF, Allegretta M, Landsman D, Zackowski KM, Brabazon F, Kostich WA, Coetzee T, Ng AV, Marrie RA, Monk KR, Bar-Or A, Whitacre CC. Pathways to cures for multiple sclerosis: A research roadmap. Mult Scler 2022; 28:331-345. [PMID: 35236198 PMCID: PMC8948371 DOI: 10.1177/13524585221075990] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Multiple Sclerosis (MS) is a growing global health challenge affecting nearly 3 million people. Progress has been made in the understanding and treatment of MS over the last several decades, but cures remain elusive. The National MS Society is focused on achieving cures for MS. Objectives: Cures for MS will be hastened by having a roadmap that describes knowledge gaps, milestones, and research priorities. In this report, we share the Pathways to Cures Research Roadmap and recommendations for strategies to accelerate the development of MS cures. Methods: The Roadmap was developed through engagement of scientific thought leaders and people affected by MS from North America and the United Kingdom. It also included the perspectives of over 300 people living with MS and was endorsed by many leading MS organizations. Results: The Roadmap consist of three distinct but overlapping cure pathways: (1) stopping the MS disease process, (2) restoring lost function by reversing damage and symptoms, and (3) ending MS through prevention. Better alignment and focus of global resources on high priority research questions are also recommended. Conclusions: We hope the Roadmap will inspire greater collaboration and alignment of global resources that accelerate scientific breakthroughs leading to cures for MS.
Collapse
Affiliation(s)
- Bruce F Bebo
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Mark Allegretta
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Douglas Landsman
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Kathy M Zackowski
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Fiona Brabazon
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Walter A Kostich
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | - Timothy Coetzee
- National Multiple Sclerosis Society 733 3rd Ave New York, NY 10017 USA
| | | | - Ruth Ann Marrie
- Department of Internal Medicine (Neurology), University of Manitoba, Winnipeg, MB, Canada
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Neurotherapeutics, Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
34
|
Petrou P, Kassis I, Ginzberg A, Hallimi M, Karussis D. Effects of Mesenchymal Stem Cell Transplantation on Cerebrospinal Fluid Biomarkers in Progressive Multiple Sclerosis. Stem Cells Transl Med 2022. [PMID: 35641166 DOI: 10.1093/stcltm/szab017.pmid:35641166;pmcid:pmc8895488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Neurofilament light chains (NF-L) were shown to serve as a reliable biomarker of neurodegeneration in multiple sclerosis (MS). The chemokine receptor CXCL13 was shown to correlate with CNS inflammatory activity and to predict the future progression of MS. OBJECTIVE To evaluate the levels of NF-L and CXCL13 in the cerebrospinal fluid (CSF) following treatment with mesenchymal stem cells (MSC) in patients with progressive MS. METHODS The CSF samples were obtained from 48 patients with progressive MS who participated in a double-blind randomized phase II clinical trial that tested the effects of intrathecal (IT) or intravenous (IV) transplantation of mesenchymal stem cells (MSC), at baseline (before the first injection of the MSC) and at 6 months following treatment with MSC, or sham treatment. The CSF specimens were tested in a blinded way, using a single-molecule array (SIMOA) technique. FINDINGS The CSF levels of NF-L were significantly lower at 6 months following treatment with MSC-IT when compared with the baseline, pre-treatment measurements (P = .026, Wilcoxon paired test). Nine out of 15 tested patients in the MSC-IT group had a reduction in NF-L levels of more than 50% (median decrease: -4449 pg/mL) when compared with 5/15 in the MSC-IV group (median decrease: -151 pg/mL) and 1/15 in the placebo group (median increase: +2450 pg/mL) (P = .001 for MSC-IT vs. placebo, chi-square test). CXCL13 levels were also reduced at 6 months following MSC-IT treatment but not to a statistically significant level. CONCLUSIONS Our findings indicate possible neuroprotective effects of MSC transplantation in patients with MS. CLINICAL TRIAL REGISTRATION NCT02166021.
Collapse
Affiliation(s)
- Panayiota Petrou
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ibrahim Kassis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ariel Ginzberg
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Michelle Hallimi
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Dimitrios Karussis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
35
|
Rg1 exerts protective effect in CPZ-induced demyelination mouse model via inhibiting CXCL10-mediated glial response. Acta Pharmacol Sin 2022; 43:563-576. [PMID: 34103690 PMCID: PMC8888649 DOI: 10.1038/s41401-021-00696-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023]
Abstract
Myelin damage and abnormal remyelination processes lead to central nervous system dysfunction. Glial activation-induced microenvironment changes are characteristic features of the diseases with myelin abnormalities. We previously showed that ginsenoside Rg1, a main component of ginseng, ameliorated MPTP-mediated myelin damage in mice, but the underlying mechanisms are unclear. In this study we investigated the effects of Rg1 and mechanisms in cuprizone (CPZ)-induced demyelination mouse model. Mice were treated with CPZ solution (300 mg· kg-1· d-1, ig) for 5 weeks; from week 2, the mice received Rg1 (5, 10, and 20 mg· kg-1· d-1, ig) for 4 weeks. We showed that Rg1 administration dose-dependently alleviated bradykinesia and improved CPZ-disrupted motor coordination ability in CPZ-treated mice. Furthermore, Rg1 administration significantly decreased demyelination and axonal injury in pathological assays. We further revealed that the neuroprotective effects of Rg1 were associated with inhibiting CXCL10-mediated modulation of glial response, which was mediated by NF-κB nuclear translocation and CXCL10 promoter activation. In microglial cell line BV-2, we demonstrated that the effects of Rg1 on pro-inflammatory and migratory phenotypes of microglia were related to CXCL10, while Rg1-induced phagocytosis of microglia was not directly related to CXCL10. In CPZ-induced demyelination mouse model, injection of AAV-CXCL10 shRNA into mouse lateral ventricles 3 weeks prior CPZ treatment occluded the beneficial effects of Rg1 administration in behavioral and pathological assays. In conclusion, CXCL10 mediates the protective role of Rg1 in CPZ-induced demyelination mouse model. This study provides new insight into potential disease-modifying therapies for myelin abnormalities.
Collapse
|
36
|
Yan X, Liu Y, Yu S, Huang D, Hu R. Repair Effects of Bone Marrow Mesenchymal Stem Cells on Demyelination of Trigeminal Ganglion in Rats with Trigeminal Neuralgia. J Pain Res 2022; 15:613-622. [PMID: 35250305 PMCID: PMC8894102 DOI: 10.2147/jpr.s347907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The current study investigated the effects of bone marrow mesenchymal stem cells (BMSCs) on pain behavior in rats with trigeminal neuralgia induced by infraorbital nerve chronic constriction injury (ION-CCI), and the repair effects of BMSCs on pathological changes in trigeminal ganglion demyelination. Methods BMSCs or phosphate-buffered saline (PBS) alone were injected around trigeminal ganglion in ION-CCI rats via a rat brain stereotaxic apparatus. Mechanical pain threshold (von Frey test) and face grooming behavior were measured in each group. Recovery of demyelination of trigeminal ganglion was observed via electron microscopy 2 weeks later, and BMSC differentiation was observed via immunofluorescence. Results Rats in the BMSC group exhibited significant improvements in mechanical pain threshold and face grooming behavior compared with the PBS group. BMSCs could repair demyelinating changes in trigeminal ganglion in ION-CCI rats. Only cells expressing GFAP, S-100, and p75 were observed via immunofluorescence, and no PKH67-labeled BMSCs were observed in the trigeminal ganglion. No BMSC differentiation was observed in the trigeminal ganglion. Conclusion Injection of BMSCs around the trigeminal ganglion could relieve trigeminal neuralgia effectively and repair trigeminal ganglion demyelination. No differentiation of BMSCs injected around the trigeminal ganglion into Schwann cells was observed. The mechanism of trigeminal neuralgia demyelination repair requires further investigation.
Collapse
Affiliation(s)
- Xuebin Yan
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yi Liu
- Department of Anesthesia, The First Hospital of Changsha, Changsha, People’s Republic of China
| | - Shanzi Yu
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Dong Huang
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Rong Hu
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Correspondence: Rong Hu, Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China, Tel +86 18973162969, Email
| |
Collapse
|
37
|
OCT4-induced oligodendrocyte progenitor cells promote remyelination and ameliorate disease. NPJ Regen Med 2022; 7:4. [PMID: 35027563 PMCID: PMC8758684 DOI: 10.1038/s41536-021-00199-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/30/2021] [Indexed: 12/23/2022] Open
Abstract
The generation of human oligodendrocyte progenitor cells (OPCs) may be therapeutically valuable for human demyelinating diseases such as multiple sclerosis. Here, we report the direct reprogramming of human somatic cells into expandable induced OPCs (iOPCs) using a combination of OCT4 and a small molecule cocktail. This method enables generation of A2B5+ (an early marker for OPCs) iOPCs within 2 weeks retaining the ability to differentiate into MBP-positive mature oligodendrocytes. RNA-seq analysis revealed that the transcriptome of O4+ iOPCs was similar to that of O4+ OPCs and ChIP-seq analysis revealed that putative OCT4-binding regions were detected in the regulatory elements of CNS development-related genes. Notably, engrafted iOPCs remyelinated the brains of adult shiverer mice and experimental autoimmune encephalomyelitis mice with MOG-induced 14 weeks after transplantation. In conclusion, our study may contribute to the development of therapeutic approaches for neurological disorders, as well as facilitate the understanding of the molecular mechanisms underlying glial development.
Collapse
|
38
|
Allanach JR, Farrell JW, Mésidor M, Karimi-Abdolrezaee S. Current status of neuroprotective and neuroregenerative strategies in multiple sclerosis: A systematic review. Mult Scler 2022; 28:29-48. [PMID: 33870797 PMCID: PMC8688986 DOI: 10.1177/13524585211008760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/13/2021] [Accepted: 03/21/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Immune-mediated demyelination and consequent degeneration of oligodendrocytes and axons are hallmark features of multiple sclerosis (MS). Remyelination declines in progressive MS, causing permanent axonal loss and irreversible disabilities. Strategies aimed at enhancing remyelination are critical to attenuate disease progression. OBJECTIVE We systematically reviewed recent advances in neuroprotective and regenerative therapies for MS, covering preclinical and clinical studies. METHODS We searched three biomedical databases using defined keywords. Two authors independently reviewed articles for inclusion based on pre-specified criteria. The data were extracted from each study and assessed for risk of bias. RESULTS Our search identified 7351 studies from 2014 to 2020, of which 221 met the defined criteria. These studies reported 262 interventions, wherein 92% were evaluated in animal models. These interventions comprised protein, RNA, lipid and cellular biologics, small molecules, inorganic compounds, and dietary and physiological interventions. Small molecules were the most highly represented strategy, followed by antibody therapies and stem cell transplantation. CONCLUSION While significant strides have been made to develop regenerative treatments for MS, the current evidence illustrates a skewed representation of the types of strategies that advance to clinical trials. Further examination is thus required to address current barriers to implementing experimental treatments in clinical settings.
Collapse
Affiliation(s)
- Jessica R Allanach
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - John W. Farrell
- Department of Health and Human Performance, Texas State University, San Marcos, TX, USA
| | - Miceline Mésidor
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada/Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada/Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
39
|
Petrou P, Kassis I, Ginzberg A, Hallimi M, Karussis D. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:55-58. [PMID: 35641166 PMCID: PMC8895488 DOI: 10.1093/stcltm/szab017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/14/2021] [Indexed: 11/25/2022] Open
Abstract
Background Neurofilament light chains (NF-L) were shown to serve as a reliable biomarker of neurodegeneration in multiple sclerosis (MS). The chemokine receptor CXCL13 was shown to correlate with CNS inflammatory activity and to predict the future progression of MS. Objective To evaluate the levels of NF-L and CXCL13 in the cerebrospinal fluid (CSF) following treatment with mesenchymal stem cells (MSC) in patients with progressive MS. Methods The CSF samples were obtained from 48 patients with progressive MS who participated in a double-blind randomized phase II clinical trial that tested the effects of intrathecal (IT) or intravenous (IV) transplantation of mesenchymal stem cells (MSC), at baseline (before the first injection of the MSC) and at 6 months following treatment with MSC, or sham treatment. The CSF specimens were tested in a blinded way, using a single-molecule array (SIMOA) technique. Findings The CSF levels of NF-L were significantly lower at 6 months following treatment with MSC-IT when compared with the baseline, pre-treatment measurements (P = .026, Wilcoxon paired test). Nine out of 15 tested patients in the MSC-IT group had a reduction in NF-L levels of more than 50% (median decrease: −4449 pg/mL) when compared with 5/15 in the MSC-IV group (median decrease: −151 pg/mL) and 1/15 in the placebo group (median increase: +2450 pg/mL) (P = .001 for MSC-IT vs. placebo, chi-square test). CXCL13 levels were also reduced at 6 months following MSC-IT treatment but not to a statistically significant level. Conclusions Our findings indicate possible neuroprotective effects of MSC transplantation in patients with MS. Clinical trial registration NCT02166021
Collapse
Affiliation(s)
- Panayiota Petrou
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ibrahim Kassis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ariel Ginzberg
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Michelle Hallimi
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Dimitrios Karussis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
- Corresponding author: Dimitrios Karussis, MD, PhD, Unit of Neuroimmunology, Hadassah Medical Organization, Ein-Karem, Jerusalem, Israel, IL-91120. Tel: +972-2-6776639;
| |
Collapse
|
40
|
Borhani-Haghighi M, Mohamadi Y. The protective effects of neural stem cells and neural stem cells-conditioned medium against inflammation-induced prenatal brain injury. J Neuroimmunol 2021; 360:577707. [PMID: 34507013 DOI: 10.1016/j.jneuroim.2021.577707] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
Intrauterine inflammation affects fetal development of the nervous system and may cause prenatal brain injury in offspring. Previously, neural stem cells have been extensively used as a therapeutic choice for nervous system diseases. Recently, the therapeutic ability of conditioned medium, harvested from cultured stem cells, has captured the attention of researchers in the field. Our study aimed to compare the therapeutic effect of neural stem cells (NSCs) or NSC-conditioned medium (NSC-CM) after prenatal brain injury. The animal model was induced by intraperitoneal injection of lipopolysaccharide into the pregnant mice and NSCs or NSC-CM were transplanted into the lateral ventricle of embryos in treatment groups. Inflammation and apoptosis were evaluated postpartum in offspring via measuring the expression of NLRP3 gene and protein, the expression and the activity of caspase-3, and the expression of pro-inflammatory cytokines by real-time PCR, immunohistochemistry, western blotting, ELISA, and colorimetric assay kit. A rotarod test was performed for motor function evaluation. Data showed that although NSC-CM fought against the inflammation and apoptosis and improved the motor function, NSCs acted more efficiently. In conclusion, the results of our study contend that NSCs have a better therapeutic effect than CM in prenatal brain injury.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomical Sciences, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
41
|
Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat Commun 2021; 12:6495. [PMID: 34764248 PMCID: PMC8586224 DOI: 10.1038/s41467-021-26834-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) ameliorate a wide range of diseases in preclinical models, but the lack of clarity around their mechanisms of action has impeded their clinical utility. The therapeutic effects of MSCs are often attributed to bioactive molecules secreted by viable MSCs. However, we found that MSCs underwent apoptosis in the lung after intravenous administration, even in the absence of host cytotoxic or alloreactive cells. Deletion of the apoptotic effectors BAK and BAX prevented MSC death and attenuated their immunosuppressive effects in disease models used to define MSC potency. Mechanistically, apoptosis of MSCs and their efferocytosis induced changes in metabolic and inflammatory pathways in alveolar macrophages to effect immunosuppression and reduce disease severity. Our data reveal a mode of action whereby the host response to dying MSCs is key to their therapeutic effects; findings that have broad implications for the effective translation of cell-based therapies. Mesenchymal stromal cells (MSCs) demonstrate therapeutic benefits in multiple diseases, but the mechanisms remain unclear as infused MSCs do not persist in the body. Here, the authors show that MSC apoptosis is an important mechanistic element, as MSCs rendered genetically incapable of apoptosis lose their ability to ameliorate disease.
Collapse
|
42
|
Gureev AP, Samoylova NA, Potanina DV, Popov VN. [Effect of methylene blue and its metabolite - azure I - on bioenergetic parameters of intact mice brain mitochondria]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:485-490. [PMID: 34964442 DOI: 10.18097/pbmc20216706485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Methylene blue is a phenothiazine dye that is widely used in medicine and clinical trials for the treatment of Alzheimer's disease. One of the factors of the unique therapeutic effect of methylene blue is its redox properties, allowing implementation of alternative electron transport - the dye accepts electrons from reducing equivalents in the mitochondria and transfer it them to other components of the respiratory chain or molecular oxygen. Azure I, an N-dimethylated metabolite of methylene blue, is potentially a more effective compound than methylene blue, but its ability for alternative electron transport has not been studied. We have shown that azure I, unlike methylene blue, is unable to restore the membrane potential in isolated mouse brain mitochondria, inhibited by rotenone and, therefore, is unable to perform bypass of the respiratory chain Complex I. Moreover, the addition of azure I does not affect the rate of mitochondrial respiration in contrast to methylene blue, which increases the rate of non-phosphorylation respiration. At the same time, both dyes stimulate an increase in H2O2 production. As a consequence, only methylene blue is capable of alternative electron transport, while azure I does not produce complex I bypass. This limits its therapeutic application only as a mitochondrial-targeted drug, but not as a substance with a potentially powerful antidepressant effect.
Collapse
Affiliation(s)
- A P Gureev
- Voronezh State University, Voronezh, Russia; Voronezh State University of Engineering Technologies, Voronezh, Russia
| | | | | | - V N Popov
- Voronezh State University, Voronezh, Russia; Voronezh State University of Engineering Technologies, Voronezh, Russia
| |
Collapse
|
43
|
Hohlfeld R. Mesenchymal stem cells for multiple sclerosis: hype or hope? Lancet Neurol 2021; 20:881-882. [PMID: 34687621 DOI: 10.1016/s1474-4422(21)00324-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Centre, Faculty of Medicine, Ludwig Maximilians University Munich, Munich D-81377, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
44
|
Lotfy A, Ali NS, Abdelgawad M, Salama M. Mesenchymal stem cells as a treatment for multiple sclerosis: a focus on experimental animal studies. Rev Neurosci 2021; 31:161-179. [PMID: 31605598 DOI: 10.1515/revneuro-2019-0040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a progressive and debilitating neurological condition in which the immune system abnormally attacks the myelin sheath insulating the nerves. Mesenchymal stem cells (MSCs) are found in most adult tissues and play a significant systemic role in self-repair. MSCs have promising therapeutic effects in many diseases, such as autoimmune diseases, including MS. MSCs have been tested in MS animal models, such as experimental autoimmune encephalomyelitis. Other studies have combined other agents with MSCs, genetically modified MSCs, or used culture medium from MSCs. In this review, we will summarize these studies and compare the main factors in each study, such as the source of MSCs, the type of animal model, the route of injection, the number of injected cells, and the mechanism of action.
Collapse
Affiliation(s)
- Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt, e-mail:
| | - Nourhan S Ali
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Mohamed Salama
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansourah, Ad Daqahliyah, Egypt.,Institute of Global Health and Human Ecology (IGHHE), American University in Cairo (AUC), Cairo, Egypt
| |
Collapse
|
45
|
Patel J, Prasad R, Bryant C, Connolly H, Teasdale B, Moosajee S. Multiple sclerosis and its impact on dental care. Br Dent J 2021; 231:281-286. [PMID: 34508197 DOI: 10.1038/s41415-021-3333-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/24/2021] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis affects approximately 130,000 people in the UK. Due to the wide variation in the symptoms associated with this condition and their variable severity, the provision of dental care for affected patients must be determined by an individual's specific needs. This will often vary significantly over time. This paper reviews the aetiology, presentation and current management of multiple sclerosis, and explores the impact of these on oral health and the provision of dental care.
Collapse
Affiliation(s)
- Jashme Patel
- Department of Oral Surgery, King´s College Dental Hospital, Bessemer Road, London, SE5 9RW, UK.
| | - Ria Prasad
- Department of Special Care, King´s College Dental Hospital, Bessemer Road, London, SE5 9RW, UK
| | - Cathy Bryant
- King´s College Hospital NHS Foundation Trust, London, UK
| | - Hannah Connolly
- Department of Community and Special Care Dentistry, King´s College Dental Hospital, Bessemer Road, London, SE5 9RW, UK
| | - Brogan Teasdale
- Department of Dental Hygiene and Therapy, King´s College Dental Hospital, Bessemer Road, London, SE5 9RW, UK
| | - Sukina Moosajee
- Department of Special Care Dentistry, King´s College NHS Foundation Trust, Dental Institute, Denmark Hill, London, SE5 9RS, UK
| |
Collapse
|
46
|
Woods WA, Chowdhury F, Tzerakis N, Adams CF, Chari DM. Developing a New Strategy for Delivery of Neural Transplant Populations Using Precursor Cell Sprays and Specialized Cell Media. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- William A. Woods
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| | - Farhana Chowdhury
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| | - Nikolaos Tzerakis
- Department of Neurosurgery University Hospital of North Midlands ST4 6QG UK
| | | | - Divya M. Chari
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| |
Collapse
|
47
|
Smith JA, Nicaise AM, Ionescu RB, Hamel R, Peruzzotti-Jametti L, Pluchino S. Stem Cell Therapies for Progressive Multiple Sclerosis. Front Cell Dev Biol 2021; 9:696434. [PMID: 34307372 PMCID: PMC8299560 DOI: 10.3389/fcell.2021.696434] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system characterized by demyelination and axonal degeneration. MS patients typically present with a relapsing-remitting (RR) disease course, manifesting as sporadic attacks of neurological symptoms including ataxia, fatigue, and sensory impairment. While there are several effective disease-modifying therapies able to address the inflammatory relapses associated with RRMS, most patients will inevitably advance to a progressive disease course marked by a gradual and irreversible accrual of disabilities. Therapeutic intervention in progressive MS (PMS) suffers from a lack of well-characterized biological targets and, hence, a dearth of successful drugs. The few medications approved for the treatment of PMS are typically limited in their efficacy to active forms of the disease, have little impact on slowing degeneration, and fail to promote repair. In looking to address these unmet needs, the multifactorial therapeutic benefits of stem cell therapies are particularly compelling. Ostensibly providing neurotrophic support, immunomodulation and cell replacement, stem cell transplantation holds substantial promise in combatting the complex pathology of chronic neuroinflammation. Herein, we explore the current state of preclinical and clinical evidence supporting the use of stem cells in treating PMS and we discuss prospective hurdles impeding their translation into revolutionary regenerative medicines.
Collapse
Affiliation(s)
- Jayden A. Smith
- Cambridge Innovation Technologies Consulting (CITC) Limited, Cambridge, United Kingdom
| | - Alexandra M. Nicaise
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rosana-Bristena Ionescu
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Regan Hamel
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
48
|
Petrou P, Kassis I, Ginzberg A, Halimi M, Yaghmour N, Abramsky O, Karussis D. Long-Term Clinical and Immunological Effects of Repeated Mesenchymal Stem Cell Injections in Patients With Progressive Forms of Multiple Sclerosis. Front Neurol 2021; 12:639315. [PMID: 34135843 PMCID: PMC8202001 DOI: 10.3389/fneur.2021.639315] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Mesenchymal stem cells (MSC) were shown to possess immunomodulatory and neurotrophic effects. Our previous trials, have shown that intrathecal (IT) and intravenous (IV) administration of MSCs were safe and provided indications of beneficial clinical effects. Methods: This is an open prospective study to evaluate the safety and the long-term clinical and immunological effects of multiple injections of autologous MSCs in 24 patients with active-progressive MS. At inclusion, the mean age of the patients was 47.0 ± 9.22, and the mean EDSS score was 6.75 ± 0.68 (range: 5.5–7.5). Patients were initially treated with 1 ×106 MSCS/kg of body weight (IT + IV) and subsequently with up to additional eight courses of MSCs, at intervals of 6–12 months. The duration of the trial was 4 years. Results: No serious, treatment-related adverse events were observed during the follow-up period. Twenty-two of the 24 patients were either stable or improved at the last follow-up visit. Ten patients had a lower than baseline EDSS at the last follow-up (nine were among those who received >2 treatments and one in the subgroup of ≤ 2 treatments, p = 0.04). The mean EDSS score reduced from 6.75 ± 0.68 at baseline to 6.42 ± 0.84 at the last visit, during a median follow-up period of 27.8 months (p = 0.028). Immunological follow-up showed a transient upregulation of CD4+CD25+FoxP3+ cells and downregulation of the proliferative ability of lymphocytes. Conclusions: Repeated MSC treatments in patients with progressive MS were shown safe at the short/intermediate term and induced clinical benefits (especially in patients treated with >2 injections) that lasted for up to 4 years, paralleled by short-term immunomodulatory effects. Clinical Trial Registration:www.ClinicalTrials.gov, identifier: NCT04823000.
Collapse
Affiliation(s)
- Panayiota Petrou
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ibrahim Kassis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Ariel Ginzberg
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Michel Halimi
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Nour Yaghmour
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Oded Abramsky
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| | - Dimitrios Karussis
- Multiple Sclerosis Center/Neuroimmunology Unit, Department of Neurology, The Agnes-Ginges Center for Neurogenetics, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
49
|
Li H, Lian G, Wang G, Yin Q, Su Z. A review of possible therapies for multiple sclerosis. Mol Cell Biochem 2021; 476:3261-3270. [PMID: 33886059 DOI: 10.1007/s11010-021-04119-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/23/2021] [Indexed: 01/22/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune chronic inflammatory disease of the central nervous system with a wide range of symptoms, like executive function defect, cognitive dysfunction, blurred vision, decreased sensation, spasticity, fatigue, and other symptoms. This neurological disease is characterized by the destruction of the blood-brain barrier, loss of myelin, and damage to neurons. It is the result of immune cells crossing the blood-brain barrier into the central nervous system and attacking self-antigens. Heretofore, many treatments proved that they can retard the progression of the disease even though there is no cure. Therefore, treatments aimed at improving patients' quality of life and reducing adverse drug reactions and costs are essential. In this review, the treatment approaches to alleviate the progress of MS include the following: pharmacotherapy, antibody therapy, cell therapy, gene therapy, and surgery. The current treatment methods of MS are described in terms of the prevention of myelin shedding, the promotion of myelin regeneration, and the protection of neurons.
Collapse
Affiliation(s)
- Hui Li
- Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China
| | - Gaojian Lian
- Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China
| | - Guang Wang
- Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China
| | - Qianmei Yin
- Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China
| | - Zehong Su
- Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan Province, China.
| |
Collapse
|
50
|
Kassis I, Ben-Zwi M, Petrou P, Halimi M, Karussis D. Synergistic neuroprotective effects of Fingolimod and mesenchymal stem cells (MSC) in experimental autoimmune encephalomyelitis. Immunol Lett 2021; 233:11-19. [PMID: 33676976 DOI: 10.1016/j.imlet.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 01/20/2023]
Abstract
Fingolimod (Gilenya™) is an effective oral medication approved for relapsing-remitting multiple sclerosis (MS), albeit less effective in chronic disease. Its main mechanism of action is through peripheral immunomodulation but neuroprotective effects may also be involved. Mesenchymal stem cells (MSC) were shown to exert immunomodulatory and neurotrophic effects in the model of multiple sclerosis (experimental autoimmune encephalomyelitis-EAE). The use of combination treatments in chronic diseases such as MS, has long been advocated and may result in improvement of the beneficial effects of each one of them. We tested the in vitro effects of Fingolimod (FTY720) on MSC and the in vivo effect of such combination treatment in the model of EAE. Fingolimod did not affect in any detrimental way the basic features of MSCs and it promoted their migration and proliferation ability .Moreover, Fingolimod induced neurotrophic factors secretion and suppressed the production of pro-inflammatory cytokines from astrocytes and microglia, in vitro. In vivo, the combined treatment of FTY720 and MSC (either by the intravenous or the intra-cerebroventricular route of administration) resulted in synergistic clinical beneficial effects compared to FTY720 or MSC alone, paralleled by a significant reduction of inflammatory CNS infiltrations and of axonal loss. These data may indicate a synergism of fingolimod with MSC and may support future combinations of immunomodulatory drugs with cellular therapies for the improvement of the benefits in progressive forms of MS.
Collapse
Affiliation(s)
- Ibrahim Kassis
- Unit of Neuroimmunology and Multiple Sclerosis Center, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel.
| | - Moriel Ben-Zwi
- Unit of Neuroimmunology and Multiple Sclerosis Center, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Panayiota Petrou
- Unit of Neuroimmunology and Multiple Sclerosis Center, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Michele Halimi
- Unit of Neuroimmunology and Multiple Sclerosis Center, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| | - Dimitrios Karussis
- Unit of Neuroimmunology and Multiple Sclerosis Center, Hadassah University Hospital, Jerusalem, Ein-Kerem, Israel
| |
Collapse
|