1
|
Ebert ET, Schwinghamer KM, Siahaan TJ. Delivery of Neuroregenerative Proteins to the Brain for Treatments of Neurodegenerative Brain Diseases. Life (Basel) 2024; 14:1456. [PMID: 39598254 PMCID: PMC11595909 DOI: 10.3390/life14111456] [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: 09/21/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Neurodegenerative brain diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), and Parkinson's disease (PD) are difficult to treat. Unfortunately, many therapeutic agents for neurodegenerative disease only halt the progression of these diseases and do not reverse neuronal damage. There is a demand for finding solutions to reverse neuronal damage in the central nervous system (CNS) of patients with neurodegenerative brain diseases. Therefore, the purpose of this review is to discuss the potential for therapeutic agents like specific neurotrophic and growth factors in promoting CNS neuroregeneration in brain diseases. We discuss how BDNF, NGF, IGF-1, and LIF could potentially be used for the treatment of brain diseases. The molecule's different mechanisms of action in stimulating neuroregeneration and methods to analyze their efficacy are described. Methods that can be utilized to deliver these proteins to the brain are also discussed.
Collapse
Affiliation(s)
| | | | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; (E.T.E.); (K.M.S.)
| |
Collapse
|
2
|
Islek Z, Ucisik MH, Sahin F. Astrocytes Can Be Key Players Against Cerebral Leishmaniasis: In Vitro Co-Culture Model for the Assessment of Infection. Parasite Immunol 2024; 46:e13071. [PMID: 39449623 DOI: 10.1111/pim.13071] [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: 07/12/2024] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024]
Abstract
Leishmaniasis is a neglected tropical disease, caused by protozoan parasites of Leishmania (L.), and is transmitted by bite of phlebotomine sandflies. There are several studies on central nervous system infection to indicate that Leishmania can cross the blood-brain barrier, resulting in neurological manifestations, known as "cerebral leishmaniasis." This study highlighted the notions: (i) polarisation of bone marrow-derived macrophages (BMDM) incubated following stimulation with lipopolysaccharide (LPS) or soluble Leishmania antigen (SLA), (ii) quantification of parasites within co-culture of Leishmania-infected macrophages, and astrocytes, and (iii) effect of interferon-gamma (IFN-γ) on the infection rate of co-culture populations. Accordingly, 83% of overall macrophage population was identified on day 7 for CD11b and F4/80 macrophage markers. Flow cytometry analysis revealed significant increases in CD11b and F4/80 surface markers in LPS and SLA-stimulated BMDMs at 24 h, compared to untreated cells. TNF-α levels increased significantly in both LPS and SLA-treated BMDMs after 48 h. Additionally, SLA treatment induced a more elongated, spindle-like shape in the cells, indicative of M2 macrophage polarisation over the M1 phenotype. When non-infected astrocytes with/without stimulation with IFN-γ before co-culture, gp63 FITC-labelled parasite populations (%) in co-culture decreased to 25% at 72 h, thus indicating a lower infection rate in a time-dependent manner. IFN-γ and IL-6 levels significantly increased to 71.66 ± 3.51 and 184 ± 14.42 pg/mL, resulting in the inflammatory response in the co-culture system at 48 h (p ≤ 0.0001), when compared to the control (30 ± 2.52 pg/mL for IFN-γ and 8.66 ± 2.37 pg/mL for IL-6) at 0 h of the incubation. It is the first study to emphasize the communication between Leishmania-infected macrophages and astrocytes regarding Leishmania parasite load. The results suggest that astrocytes can lead to the reduction in Leishmania parasites, thereby controlling the incidence of cerebral leishmaniasis.
Collapse
Affiliation(s)
- Zeynep Islek
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Ataşehir/Istanbul, Turkey
| | - Mehmet Hikmet Ucisik
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Ataşehir/Istanbul, Turkey
| | - Fikrettin Sahin
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Ataşehir/Istanbul, Turkey
| |
Collapse
|
3
|
Wu L, Lu J, Lan T, Zhang D, Xu H, Kang Z, Peng F, Wang J. Stem cell therapies: a new era in the treatment of multiple sclerosis. Front Neurol 2024; 15:1389697. [PMID: 38784908 PMCID: PMC11111935 DOI: 10.3389/fneur.2024.1389697] [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: 02/22/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Multiple Sclerosis (MS) is an immune-mediated condition that persistently harms the central nervous system. While existing treatments can slow its course, a cure remains elusive. Stem cell therapy has gained attention as a promising approach, offering new perspectives with its regenerative and immunomodulatory properties. This article reviews the application of stem cells in MS, encompassing various stem cell types, therapeutic potential mechanisms, preclinical explorations, clinical research advancements, safety profiles of clinical applications, as well as limitations and challenges, aiming to provide new insights into the treatment research for MS.
Collapse
Affiliation(s)
- Lei Wu
- Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Tianye Lan
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Hanying Xu
- Changchun University of Chinese Medicine, Changchun, China
| | - Zezheng Kang
- Changchun University of Chinese Medicine, Changchun, China
| | - Fang Peng
- Hunan Provincial People's Hospital, Changsha, China
| | - Jian Wang
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
4
|
Ren R, Jiang J, Li X, Zhang G. Research progress of autoimmune diseases based on induced pluripotent stem cells. Front Immunol 2024; 15:1349138. [PMID: 38720903 PMCID: PMC11076788 DOI: 10.3389/fimmu.2024.1349138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.
Collapse
Affiliation(s)
| | | | | | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| |
Collapse
|
5
|
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
|
6
|
Zolfaghari Baghbadorani P, Rayati Damavandi A, Moradi S, Ahmadi M, Bemani P, Aria H, Mottedayyen H, Rayati Damavandi A, Eskandari N, Fathi F. Current advances in stem cell therapy in the treatment of multiple sclerosis. Rev Neurosci 2023; 34:613-633. [PMID: 36496351 DOI: 10.1515/revneuro-2022-0102] [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: 08/10/2022] [Accepted: 11/18/2022] [Indexed: 08/04/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease related to the central nervous system (CNS) with a significant global burden. In this illness, the immune system plays an essential role in its pathophysiology and progression. The currently available treatments are not recognized as curable options and, at best, might slow the progression of MS injuries to the CNS. However, stem cell treatment has provided a new avenue for treating MS. Stem cells may enhance CNS healing and regulate immunological responses. Likewise, stem cells can come from various sources, including adipose, neuronal, bone marrow, and embryonic tissues. Choosing the optimal cell source for stem cell therapy is still a difficult verdict. A type of stem cell known as mesenchymal stem cells (MSCs) is obtainable from different sources and has a strong immunomodulatory impact on the immune system. According to mounting data, the umbilical cord and adipose tissue may serve as appropriate sources for the isolation of MSCs. Human amniotic epithelial cells (hAECs), as novel stem cell sources with immune-regulatory effects, regenerative properties, and decreased antigenicity, can also be thought of as a new upcoming contender for MS treatment. Overall, the administration of stem cells in different sets of animal and clinical trials has shown immunomodulatory and neuroprotective results. Therefore, this review aims to discuss the different types of stem cells by focusing on MSCs and their mechanisms, which can be used to treat and improve the outcomes of MS disease.
Collapse
Affiliation(s)
| | - Amirmasoud Rayati Damavandi
- Students' Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Keshavarz Blvrd, Vesal Shirazi St., Tehran 1417613151, Iran
| | - Samira Moradi
- School of Medicine, Hormozgan University of Medical Sciences Chamran Blvrd., Hormozgan 7919693116, Bandar Abbass, Iran
| | - Meysam Ahmadi
- School of Medicine, Shiraz University of Medical Sciences, Fars, Zand St., Shiraz 7134814336, Iran
| | - Peyman Bemani
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fars, Ibn Sina Sq., Fasa 7461686688, Iran
| | - Hossein Mottedayyen
- Department of Immunology, School of Medicine, Kashan University of Medical Sciences, Ravandi Blvrd, Isfahan, Kashan 8715988141, Iran
| | - Amirhossein Rayati Damavandi
- Student's Research Committee, Pharmaceutical Sciences Branch, Islamic Azad University, Yakhchal St., Tehran 193951498, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Farshid Fathi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| |
Collapse
|
7
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
8
|
Colombo E, De Angelis A, Bassani C, Ruffini F, Ottoboni L, Garzetti L, Finardi A, Martino G, Furlan R, Farina C. iAstrocytes do not restrain T cell proliferation in vitro. BMC Neurosci 2023; 24:33. [PMID: 37286983 DOI: 10.1186/s12868-023-00806-3] [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: 01/18/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
The cross-talk between T cells and astrocytes occurring under physiological and, even more, neuroinflammatory conditions may profoundly impact the generation of adaptive immune responses in the nervous tissue. In this study, we used a standardized in vitro co-culture assay to investigate the immunomodulatory properties of astrocytes differing for age, sex, and species. Mouse neonatal astrocytes enhanced T cell vitality but suppressed T lymphocyte proliferation in response to mitogenic stimuli or myelin antigens, regardless of the Th1, Th2 or Th17 T cell phenotype. Studies comparing glia cells from adult and neonatal animals showed that adult astrocytes were more efficient in inhibiting T lymphocyte activation than neonatal astrocytes, regardless of their sex. Differently from primary cultures, mouse and human astrocytes derived from reprogrammed fibroblasts did not interfere with T cell proliferation. Overall, we describe a standardized astrocyte-T cell interaction in vitro assay and demonstrate that primary astrocytes and iAstrocytes may differ in modulating T cell function.
Collapse
Affiliation(s)
- Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Anthea De Angelis
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Claudia Bassani
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesca Ruffini
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Linda Ottoboni
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Livia Garzetti
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Annamaria Finardi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gianvito Martino
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Roberto Furlan
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
9
|
Sasson DC, Islam S, Duan K, Dash BC, Hsia HC. TNF-α Preconditioning Promotes a Proangiogenic Phenotype in hiPSC-Derived Vascular Smooth Muscle Cells. Cell Mol Bioeng 2023; 16:231-240. [PMID: 37456784 PMCID: PMC10338418 DOI: 10.1007/s12195-023-00764-0] [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: 10/04/2022] [Accepted: 03/29/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction hiPSC-VSMCs have been suggested as therapeutic agents for wound healing and revascularization through the secretion of proangiogenic factors. However, methods of increasing cell paracrine secretion and survivability have thus far yielded inconsistent results. This study investigates the effect of pre-conditioning of hiPSC-VSMCs with TNF-α and their integration into 3D collagen scaffolds on cellular viability and secretome. Methods hiPSC-VSMCs were dual-plated in a 2D environment. TNF-α was introduced to one plate. Following incubation, cells from each plate were divided and added to type-I collagen scaffolds. TNF-α was introduced to two sets of scaffolds, one from each 2D plate. Following incubation, scaffolds were harvested for their media, tested for cell survivability, cytotoxicity, and imaged. Intra-media VEGF and bFGF levels were evaluated using ELISA testing. Results hiPSC-VSMCs exposed to TNF-α during collagen scaffold proliferation and preconditioning showed an increase in cell viability and less cytotoxicity compared to non-exposed cells and solely-preconditioned cells. Significant increases in bFGF expression were found in pre-conditioned cell groups with further increases found in cells subsequently exposed during intra-scaffold conditioning. A significant increase in VEGF expression was found in cell groups exposed during both pre-conditioning and intra-scaffold conditioning. Fibroblasts treated with any conditioned media demonstrated increased migration potential. Conclusions Conditioning hiPSC-VSMCs embedded in scaffolds with TNF-α improves cellular viability and increases the secretion of paracrine factors necessary for wound healing mechanisms such as migration. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00764-0.
Collapse
Affiliation(s)
- Daniel C Sasson
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, 330 Cedar Street, Boardman Bldg, 3rd Floor, New Haven, CT 06510 USA
| | - Sara Islam
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, 330 Cedar Street, Boardman Bldg, 3rd Floor, New Haven, CT 06510 USA
| | - Kaiti Duan
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, 330 Cedar Street, Boardman Bldg, 3rd Floor, New Haven, CT 06510 USA
| | - Biraja C Dash
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, 330 Cedar Street, Boardman Bldg, 3rd Floor, New Haven, CT 06510 USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT USA
| | - Henry C Hsia
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, 330 Cedar Street, Boardman Bldg, 3rd Floor, New Haven, CT 06510 USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT USA
| |
Collapse
|
10
|
Zhang R, Zhang Y, Wu T, Tian W, Luo J, Shi Y, Su D, Shu H, Tian J. Bibliometric analysis of research topics on blood-brain barrier breakdown and cognitive function over the last two decades (2000-2021). Front Aging Neurosci 2023; 15:1108561. [PMID: 37323140 PMCID: PMC10268002 DOI: 10.3389/fnagi.2023.1108561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 05/02/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Blood-brain barrier (BBB) breakdown is closely associated with cognitive dysfunction. This study aimed to categorize and summarize research topics on the correlation between BBB breakdown and its effects on cognitive function. Methods Bibliometric analysis methods were used to quantitatively and qualitatively assess research progress and predict future research hotspots. Relevant publications from the Web of the Science Core Collection were extracted on November 5, 2022 and analyzed to predict trends and hotspots in the field. Results We identified 5518 articles published from 2000 to 2021 about the BBB and cognition. The number of manuscripts on this topic increased steadily during this time period, especially after 2013. We found that the number of articles published in China increased gradually and is in second place behind the United States of America (USA). In the research field of BBB breakdown and cognitive function, the USA is still far ahead. Keyword burst detection suggested that cognitive impairment, neurodegeneration disease and neuroinflammation are emerging research hotspots. Discussion The mechanisms of BBB integrity breakdown and its effects on the deterioration of cognitive function are complex, and clinical treatment of the affected diseases has been a hot topic in the field over the past 22 years. Looking forward, this body of research is aimed at improving or maintaining patients' cognitive abilities, by finding preventive measures and to provide a basis for finding new treatments of cognitive disorders.
Collapse
|
11
|
Zhang Q, Chen Z, Zhang K, Zhu J, Jin T. FGF/FGFR system in the central nervous system demyelinating disease: Recent progress and implications for multiple sclerosis. CNS Neurosci Ther 2023; 29:1497-1511. [PMID: 36924298 PMCID: PMC10173727 DOI: 10.1111/cns.14176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND With millions of victims worldwide, multiple sclerosis is the second most common cause of disability among young adults. Although formidable advancements have been made in understanding the disease, the neurodegeneration associated with multiple sclerosis is only partially counteracted by current treatments, and effective therapy for progressive multiple sclerosis remains an unmet need. Therefore, new approaches are required to delay demyelination and the resulting disability and to restore neural function by promoting remyelination and neuronal repair. AIMS The article reviews the latest literature in this field. MATERIALS AND METHODS The fibroblast growth factor (FGF) signaling pathway is a promising target in progressive multiple sclerosis. DISCUSSION FGF signal transduction contributes to establishing the oligodendrocyte lineage, neural stem cell proliferation and differentiation, and myelination of the central nervous system. Furthermore, FGF signaling is implicated in the control of neuroinflammation. In recent years, interventions targeting FGF, and its receptor (FGFR) have been shown to ameliorate autoimmune encephalomyelitis symptoms in multiple sclerosis animal models moderately. CONCLUSION Here, we summarize the recent findings and investigate the role of FGF/FGFR signaling in the onset and progression, discuss the potential therapeutic advances, and offer fresh insights into managing multiple sclerosis.
Collapse
Affiliation(s)
- Qingxiang Zhang
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Kaili Zhang
- Stomatology College of Inner Mongolia Medical University, Hohhot, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Borda M, Aquino JB, Mazzone GL. Cell-based experimental strategies for myelin repair in multiple sclerosis. J Neurosci Res 2023; 101:86-111. [PMID: 36164729 DOI: 10.1002/jnr.25129] [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: 05/15/2022] [Revised: 08/21/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS), diagnosed at a mean age of 32 years. CNS glia are crucial players in the onset of MS, primarily involving astrocytes and microglia that can cause/allow massive oligodendroglial cells death, without immune cell infiltration. Current therapeutic approaches are aimed at modulating inflammatory reactions during relapsing episodes, but lack the ability to induce very significant repair mechanisms. In this review article, different experimental approaches based mainly on the application of different cell types as therapeutic strategies applied for the induction of myelin repair and/or the amelioration of the disease are discussed. Regarding this issue, different cell sources were applied in various experimental models of MS, with different results, both in significant improvements in remyelination and the reduction of neuroinflammation and glial activation, or in neuroprotection. All cell types tested have advantages and disadvantages, which makes it difficult to choose a better option for therapeutic application in MS. New strategies combining cell-based treatment with other applications would result in further improvements and would be good candidates for MS cell therapy and myelin repair.
Collapse
Affiliation(s)
- Maximiliano Borda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
| | - Jorge B Aquino
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| | - Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| |
Collapse
|
13
|
Genchi A, Brambilla E, Sangalli F, Radaelli M, Bacigaluppi M, Furlan R, Andolfo A, Drago D, Magagnotti C, Scotti GM, Greco R, Vezzulli P, Ottoboni L, Bonopane M, Capilupo D, Ruffini F, Belotti D, Cabiati B, Cesana S, Matera G, Leocani L, Martinelli V, Moiola L, Vago L, Panina-Bordignon P, Falini A, Ciceri F, Uglietti A, Sormani MP, Comi G, Battaglia MA, Rocca MA, Storelli L, Pagani E, Gaipa G, Martino G. Neural stem cell transplantation in patients with progressive multiple sclerosis: an open-label, phase 1 study. Nat Med 2023; 29:75-85. [PMID: 36624312 PMCID: PMC9873560 DOI: 10.1038/s41591-022-02097-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/17/2022] [Indexed: 01/11/2023]
Abstract
Innovative pro-regenerative treatment strategies for progressive multiple sclerosis (PMS), combining neuroprotection and immunomodulation, represent an unmet need. Neural precursor cells (NPCs) transplanted in animal models of multiple sclerosis have shown preclinical efficacy by promoting neuroprotection and remyelination by releasing molecules sustaining trophic support and neural plasticity. Here we present the results of STEMS, a prospective, therapeutic exploratory, non-randomized, open-label, single-dose-finding phase 1 clinical trial ( NCT03269071 , EudraCT 2016-002020-86), performed at San Raffaele Hospital in Milan, Italy, evaluating the feasibility, safety and tolerability of intrathecally transplanted human fetal NPCs (hfNPCs) in 12 patients with PMS (with evidence of disease progression, Expanded Disability Status Scale ≥6.5, age 18-55 years, disease duration 2-20 years, without any alternative approved therapy). The safety primary outcome was reached, with no severe adverse reactions related to hfNPCs at 2-year follow-up, clearly demonstrating that hfNPC therapy in PMS is feasible, safe and tolerable. Exploratory secondary analyses showed a lower rate of brain atrophy in patients receiving the highest dosage of hfNPCs and increased cerebrospinal fluid levels of anti-inflammatory and neuroprotective molecules. Although preliminary, these results support the rationale and value of future clinical studies with the highest dose of hfNPCs in a larger cohort of patients.
Collapse
Affiliation(s)
- Angela Genchi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Elena Brambilla
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sangalli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Radaelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bacigaluppi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Roberto Furlan
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annapaola Andolfo
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Denise Drago
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Magagnotti
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Maria Scotti
- grid.18887.3e0000000417581884Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Greco
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Vezzulli
- grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Linda Ottoboni
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bonopane
- grid.18887.3e0000000417581884Clinical Trial Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Capilupo
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ruffini
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Belotti
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Stefania Cesana
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Giada Matera
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Letizia Leocani
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Vittorio Martinelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Panina-Bordignon
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Falini
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Uglietti
- grid.414818.00000 0004 1757 8749Department of Gynaecology, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Pia Sormani
- grid.5606.50000 0001 2151 3065Biostatistics Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Giancarlo Comi
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | | | - Maria A. Rocca
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Gaipa
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
14
|
Zayed MA, Sultan S, Alsaab HO, Yousof SM, Alrefaei GI, Alsubhi NH, Alkarim S, Al Ghamdi KS, Bagabir SA, Jana A, Alghamdi BS, Atta HM, Ashraf GM. Stem-Cell-Based Therapy: The Celestial Weapon against Neurological Disorders. Cells 2022; 11:3476. [PMID: 36359871 PMCID: PMC9655836 DOI: 10.3390/cells11213476] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Stem cells are a versatile source for cell therapy. Their use is particularly significant for the treatment of neurological disorders for which no definitive conventional medical treatment is available. Neurological disorders are of diverse etiology and pathogenesis. Alzheimer's disease (AD) is caused by abnormal protein deposits, leading to progressive dementia. Parkinson's disease (PD) is due to the specific degeneration of the dopaminergic neurons causing motor and sensory impairment. Huntington's disease (HD) includes a transmittable gene mutation, and any treatment should involve gene modulation of the transplanted cells. Multiple sclerosis (MS) is an autoimmune disorder affecting multiple neurons sporadically but induces progressive neuronal dysfunction. Amyotrophic lateral sclerosis (ALS) impacts upper and lower motor neurons, leading to progressive muscle degeneration. This shows the need to try to tailor different types of cells to repair the specific defect characteristic of each disease. In recent years, several types of stem cells were used in different animal models, including transgenic animals of various neurologic disorders. Based on some of the successful animal studies, some clinical trials were designed and approved. Some studies were successful, others were terminated and, still, a few are ongoing. In this manuscript, we aim to review the current information on both the experimental and clinical trials of stem cell therapy in neurological disorders of various disease mechanisms. The different types of cells used, their mode of transplantation and the molecular and physiologic effects are discussed. Recommendations for future use and hopes are highlighted.
Collapse
Affiliation(s)
- Mohamed A. Zayed
- Physiology Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Physiology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Samar Sultan
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Shimaa Mohammad Yousof
- Physiology Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ghadeer I. Alrefaei
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Nouf H. Alsubhi
- Department of Biological Sciences, College of Science & Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Saleh Alkarim
- Embryonic and Cancer Stem Cell Research Group, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biology Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Embryonic Stem Cells Research Unit, Biology Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kholoud S. Al Ghamdi
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sali Abubaker Bagabir
- Genetic Unit, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Campus-11, Patia, Bhubaneswar 751024, Odisha, India
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hazem M. Atta
- Clinical Biochemistry Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| |
Collapse
|
15
|
Abstract
PURPOSE OF THE REVIEW Despite the significant progress in the development of disease-modifying treatments for multiple sclerosis (MS), repair of existing damage is still poorly addressed. Current research focuses on stem cell-based therapies as a suitable alternative or complement to current drug therapies. RECENT FINDINGS Myelin damage is a hallmark of multiple sclerosis, and novel approaches leading to remyelination represent a promising tool to prevent neurodegeneration of the underlying axon. With increasing evidence of diminishing remyelination capacity of the MS brain with ageing and disease progression, exogenous cell transplantation is a promising therapeutic approach for restoration of oligodendrocyte precursor cell pool reserve and myelin regeneration. SUMMARY The present review summarizes recent developments of remyelinating therapies in multiple sclerosis, focusing on exogenous cell-based strategies and discussing related scientific, practical, and ethical concerns.
Collapse
|
16
|
Fortune AJ, Fletcher JL, Blackburn NB, Young KM. Using MS induced pluripotent stem cells to investigate MS aetiology. Mult Scler Relat Disord 2022; 63:103839. [DOI: 10.1016/j.msard.2022.103839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/01/2022] [Accepted: 04/29/2022] [Indexed: 12/15/2022]
|
17
|
Salwierak-Głośna K, Piątek P, Domowicz M, Świderek-Matysiak M. Effect of Multiple Sclerosis Cerebrospinal Fluid and Oligodendroglia Cell Line Environment on Human Wharton's Jelly Mesenchymal Stem Cells Secretome. Int J Mol Sci 2022; 23:ijms23042177. [PMID: 35216294 PMCID: PMC8878514 DOI: 10.3390/ijms23042177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a neurological disorder of autoimmune aetiology. Experimental therapies with the use of mesenchymal stem cells (MSCs) have emerged as a response to the unmet need for new treatment options. The unique immunomodulatory features of stem cells obtained from Wharton’s jelly (WJ-MSCs) make them an interesting research and therapeutic model. Most WJ-MSCs transplants for multiple sclerosis use intrathecal administration. We studied the effect of cerebrospinal fluid (CSF) obtained from MS patients on the secretory activity of WJ-MSCs and broaden this observation with WJ-MSCs interactions with human oligodendroglia cell line (OLs). Analysis of the WJ-MSCs secretory activity with use of Bio-Plex Pro™ Human Cytokine confirmed significant and diverse immunomodulatory potential. Our data reveal rich WJ-MSCs secretome with markedly increased levels of IL-6, IL-8, IP-10 and MCP-1 synthesis and a favourable profile of growth factors. The addition of MS CSF to the WJ-MSCs culture caused depletion of most proteins measured, only IL-12, RANTES and GM-CSF levels were increased. Most cytokines and chemokines decreased their concentrations in WJ-MSCs co-cultured with OLs, only eotaxin and RANTES levels were slightly increased. These results emphasize the spectrum of the immunomodulatory properties of WJ-MSCs and show how those effects can be modulated depending on the transplantation milieu.
Collapse
Affiliation(s)
| | - Paweł Piątek
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
- Department of Immunogenetics, Medical University of Lodz, 90-419 Lodz, Poland
| | - Małgorzata Domowicz
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
| | - Mariola Świderek-Matysiak
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
- Correspondence:
| |
Collapse
|
18
|
Theotokis P, Kesidou E, Mitsiadou D, Petratos S, Damianidou O, Boziki M, Chatzidimitriou A, Grigoriadis N. Lumbar spine intrathecal transplantation of neural precursor cells promotes oligodendrocyte proliferation in hot spots of chronic demyelination. Brain Pathol 2021; 32:e13040. [PMID: 34845781 PMCID: PMC9245942 DOI: 10.1111/bpa.13040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a basic and reliable model used to study clinical and pathological hallmarks of multiple sclerosis (MS) in rodents. Several studies suggest neural precursor cells (NPCs) as a significant research tool while reporting that transplanted NPCs are a promising therapeutic approach to treating neurological disorders, such as MS. The main objective was to approach a preclinical, in vivo scenario of oligodendrogenesis with NPCs, targeting the main chronic demyelinated lumbosacral milieu of EAE, via the least invasive delivery method which is lumbar puncture. We utilized MOG35‐55 peptide to induce EAE in C57BL/6 mice and prior to the acute relapse, we intervened with either the traceable GFP+ cellular therapy or saline solution in the intrathecal space of their lumbar spine. A BrdU injection, which enabled us to monitor endogenous proliferation, marked the endpoint 50 days post‐induction (50 dpi). Neuropathology with high‐throughput, triple immunofluorescent, and transmission electron microscopy (TEM) data were extracted and analyzed. The experimental treatment attenuated the chronic phase of EAE (50 dpi; score <1) following an acute, clinical relapse. Myelination and axonal integrity were rescued in the NPC‐treated animals along with suppressed immune populations. The differentiation profile of the exogenous NPCs and endogenous BrdU+ cells was location‐dependent where GFP+‐rich areas drove undifferentiated phenotypes toward the oligodendrocyte lineage. In situ oligodendrocyte enrichment was demonstrated through increased (p < 0.001) gap junction channels of Cx32 and Cx47, reliable markers for proliferative oligodendroglia syncytium. TEM morphometric analysis ultimately manifested an increased g‐ratio in lumbosacral fibers of the recovered animals (p < 0.001). Herein, we suggest that a single, lumbar intrathecal administration of NPCs capacitated a viable cellular load and resulted in clinical and pathological amelioration, stimulating resident OPCs to overcome the remyelination failure in EAE demyelinating locale.
Collapse
Affiliation(s)
- Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Dimitra Mitsiadou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Olympia Damianidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece.,Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| |
Collapse
|
19
|
Jiang S, Wang H, Zhou Q, Li Q, Liu N, Li Z, Chen C, Deng Y. Melatonin Ameliorates Axonal Hypomyelination of Periventricular White Matter by Transforming A1 to A2 Astrocyte via JAK2/STAT3 Pathway in Septic Neonatal Rats. J Inflamm Res 2021; 14:5919-5937. [PMID: 34803390 PMCID: PMC8595063 DOI: 10.2147/jir.s337499] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Background Astrocyte A1/A2 phenotypes may play differential role in the pathogenesis of periventricular white matter (PWM) damage in septic postnatal rats. This study aimed to determine whether melatonin (MEL) would improve the axonal hypomyelination through shifting A1 astrocytes towards A2. Methods One-day-old Sprague-Dawley rats were divided into control, LPS, and LPS+MEL groups. Immunofluorescence was performed to detect C1q, IL-1α, TNF-α, IBA1, GFAP, MAG, C3 and S100A10 immunoreactivity in the PWM of neonatal rats. Electron microscopy was conducted to observe alterations of axonal myelin sheath in the PWM; moreover, myelin protein expression was assessed using in situ hybridization. The effects of MEL on neurological function were evaluated by behavioral tests. In vitro, A1 astrocytes were induced by IL-1α, C1q and TNF-α, and following which the effect of MEL on C3 and S100A10 expression was determined by Western blot and immunofluorescence. Results At 1 and 3 days after LPS injection, IBA1+ microglia in the PWM were significantly increased in cell numbers which generated excess amounts of IL-1α, TNF-α, and C1q. The number of A1 astrocytes was significantly increased at 7-28d after LPS injection. In rats given MEL treatment, the number of A1 astrocytes was significantly decreased, but that of A2 astrocytes, PLP+, MBP+ and MAG+ cells was increased. By electron microscopy, ultrastructural features of axonal hypomyelination were attenuated by MEL. Furthermore, MEL improved neurological dysfunction as evaluated by different neurological tests. In vitro, MEL decreased the C3 significantly, and upregulated expression of S100A10 in primary astrocytes subjected to IL-1α, TNF-α and C1q treatment. Importantly, JAK2/STAT3 signaling pathway was found to be involved in modulation of A1/A2 phenotype transformation. Conclusion MEL effectively alleviates PWMD of septic neonatal rats, which is most likely through modulating astrocyte phenotypic transformation from A1 to A2 via the MT1/JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Shuqi Jiang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Huifang Wang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Qiuping Zhou
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510641, People's Republic of China
| | - Qian Li
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Nan Liu
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510641, People's Republic of China
| | - Zhenggong Li
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Yiyu Deng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
20
|
Aishwarya L, Arun D, Kannan S. Stem cells as a potential therapeutic option for treating neurodegenerative diseases. Curr Stem Cell Res Ther 2021; 17:590-605. [PMID: 35135464 DOI: 10.2174/1574888x16666210810105136] [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: 02/25/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
In future, neurodegenerative diseases will take over cancer's place and become the major cause of death in the world, especially in developed countries. Advancements in the medical field and its facilities have led to an increase in the old age population, and thus contributing to the increase in number of people suffering from neurodegenerative diseases. Economically it is of a great burden to society and the affected family. No current treatment aims to replace, protect, and regenerate lost neurons; instead, it alleviates the symptoms, extends the life span by a few months and creates severe side effects. Moreover, people who are affected are physically dependent for performing their basic activities, which makes their life miserable. There is an urgent need for therapy that could be able to overcome the deficits of conventional therapy for neurodegenerative diseases. Stem cells, the unspecialized cells with the properties of self-renewing and potency to differentiate into various cells types can become a potent therapeutic option for neurodegenerative diseases. Stem cells have been widely used in clinical trials to evaluate their potential in curing different types of ailments. In this review, we discuss the various types of stem cells and their potential use in the treatment of neurodegenerative disease based on published preclinical and clinical studies.
Collapse
Affiliation(s)
- Aishwarya L
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Dharmarajan Arun
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| | - Suresh Kannan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai-600 116. India
| |
Collapse
|
21
|
des Rieux A. Stem cells and their extracellular vesicles as natural and bioinspired carriers for the treatment of neurological disorders. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2021.101460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
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: 24] [Impact Index Per Article: 6.0] [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
|
23
|
Tejeda G, Ciciriello AJ, Dumont CM. Biomaterial Strategies to Bolster Neural Stem Cell-Mediated Repair of the Central Nervous System. Cells Tissues Organs 2021; 211:655-669. [PMID: 34120118 DOI: 10.1159/000515351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/12/2021] [Indexed: 01/25/2023] Open
Abstract
Stem cell therapies have the potential to not only repair, but to regenerate tissue of the central nervous system (CNS). Recent studies demonstrate that transplanted stem cells can differentiate into neurons and integrate with the intact circuitry after traumatic injury. Unfortunately, the positive findings described in rodent models have not been replicated in clinical trials, where the burden to maintain the cell viability necessary for tissue repair becomes more challenging. Low transplant survival remains the greatest barrier to stem cell-mediated repair of the CNS, often with fewer than 1-2% of the transplanted cells remaining after 1 week. Strategic transplantation parameters, such as injection location, cell concentration, and transplant timing achieve only modest improvements in stem cell transplant survival and appear inconsistent across studies. Biomaterials provide researchers with a means to significantly improve stem cell transplant survival through two mechanisms: (1) a vehicle to deliver and protect the stem cells and (2) a substrate to control the cytotoxic injury environment. These biomaterial strategies can alleviate cell death associated with delivery to the injury and can be used to limit cell death after transplantation by limiting cell exposure to cytotoxic signals. Moreover, it is likely that control of the injury environment with biomaterials will lead to a more reliable support for transplanted cell populations. This review will highlight the challenges associated with cell delivery in the CNS and the advances in biomaterial development and deployment for stem cell therapies necessary to bolster stem cell-mediated repair.
Collapse
Affiliation(s)
- Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
24
|
Chen Q, Liu Q, Zhang Y, Li S, Yi S. Leukemia inhibitory factor regulates Schwann cell proliferation and migration and affects peripheral nerve regeneration. Cell Death Dis 2021; 12:417. [PMID: 33888681 PMCID: PMC8062678 DOI: 10.1038/s41419-021-03706-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022]
Abstract
Leukemia inhibitory factor (LIF) is a pleiotropic cytokine that stimulates neuronal development and survival. Our previous study has demonstrated that LIF mRNA is dysregulated in the peripheral nerve segments after nerve injury. Here, we show that LIF protein is abundantly expressed in Schwann cells after rat sciatic nerve injury. Functionally, suppressed or elevated LIF increases or decreases the proliferation rate and migration ability of Schwann cells, respectively. Morphological observations demonstrate that in vivo application of siRNA against LIF after peripheral nerve injury promotes Schwann cell migration and proliferation, axon elongation, and myelin formation. Electrophysiological and behavior assessments disclose that knockdown of LIF benefits the function recovery of injured peripheral nerves. Differentially expressed LIF affects the metabolism of Schwann cells and negatively regulates ERFE (Erythroferrone). Collectively, our observations reveal the essential roles for LIF in regulating the proliferation and migration of Schwann cells and the regeneration of injured peripheral nerves, discover ERFE as a downstream effector of LIF, and extend our understanding of the molecular mechanisms underlying peripheral nerve regeneration.
Collapse
Affiliation(s)
- Qianqian Chen
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China.,State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Qianyan Liu
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Yunsong Zhang
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Shiying Li
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Sheng Yi
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
25
|
Mozafari S, Deboux C, Laterza C, Ehrlich M, Kuhlmann T, Martino G, Baron-Van Evercooren A. Beneficial contribution of induced pluripotent stem cell-progeny to Connexin 47 dynamics during demyelination-remyelination. Glia 2020; 69:1094-1109. [PMID: 33301181 PMCID: PMC7984339 DOI: 10.1002/glia.23950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Oligodendrocytes are extensively coupled to astrocytes, a phenomenon ensuring glial homeostasis and maintenance of central nervous system myelin. Molecular disruption of this communication occurs in demyelinating diseases such as multiple sclerosis. Less is known about the vulnerability and reconstruction of the panglial network during adult demyelination‐remyelination. Here, we took advantage of lysolcithin‐induced demyelination to investigate the expression dynamics of the oligodendrocyte specific connexin 47 (Cx47) and to some extent that of astrocyte Cx43, and whether this dynamic could be modulated by grafted induced pluripotent stem cell (iPSC)‐neural progeny. Our data show that disruption of Cx43‐Cx47 mediated hetero‐cellular gap‐junction intercellular communication following demyelination is larger in size than demyelination. Loss of Cx47 expression is timely rescued during remyelination and accelerated by the grafted neural precursors. Moreover, mouse and human iPSC‐derived oligodendrocytes express Cx47, which co‐labels with astrocyte Cx43, indicating their integration into the panglial network. These data suggest that in rodents, full lesion repair following transplantation occurs by panglial reconstruction in addition to remyelination. Targeting panglial elements by cell therapy or pharmacological compounds may help accelerating or stabilizing re/myelination in myelin disorders.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cyrille Deboux
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cecilia Laterza
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy.,Industrial Engineering Department, University of Padova, Padova, Italy
| | - Marc Ehrlich
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Anne Baron-Van Evercooren
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
26
|
Mozafari S, Starost L, Manot-Saillet B, Garcia-Diaz B, Xu YKT, Roussel D, Levy MJF, Ottoboni L, Kim KP, Schöler HR, Kennedy TE, Antel JP, Martino G, Angulo MC, Kuhlmann T, Baron-Van Evercooren A. Multiple sclerosis iPS-derived oligodendroglia conserve their properties to functionally interact with axons and glia in vivo. SCIENCE ADVANCES 2020; 6:6/49/eabc6983. [PMID: 33277253 PMCID: PMC7821889 DOI: 10.1126/sciadv.abc6983] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 05/04/2023]
Abstract
Remyelination failure in multiple sclerosis (MS) is associated with a migration/differentiation block of oligodendroglia. The reason for this block is highly debated. It could result from disease-related extrinsic or intrinsic regulators in oligodendroglial biology. To avoid confounding immune-mediated extrinsic effect, we used an immune-deficient mouse model to compare induced pluripotent stem cell-derived oligodendroglia from MS and healthy donors following engraftment in the developing CNS. We show that the MS-progeny behaves and differentiates into oligodendrocytes to the same extent as controls. They generate equal amounts of myelin, with bona fide nodes of Ranvier, and promote equal restoration of their host slow conduction. MS-progeny expressed oligodendrocyte- and astrocyte-specific connexins and established functional connections with donor and host glia. Thus, MS oligodendroglia, regardless of major immune manipulators, are intrinsically capable of myelination and making functional axo-glia/glia-glia connections, reinforcing the view that the MS oligodendrocyte differentiation block is not from major intrinsic oligodendroglial deficits.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Laura Starost
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Blandine Manot-Saillet
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Beatriz Garcia-Diaz
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Yu Kang T Xu
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Delphine Roussel
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Marion J F Levy
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Linda Ottoboni
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Timothy E Kennedy
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Maria Cecilia Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Anne Baron-Van Evercooren
- INSERM, U1127, F-75013 Paris, France.
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| |
Collapse
|
27
|
Martínez-Larrosa J, Matute-Blanch C, Montalban X, Comabella M. Modelling multiple sclerosis using induced pluripotent stem cells. J Neuroimmunol 2020; 349:577425. [PMID: 33130461 DOI: 10.1016/j.jneuroim.2020.577425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 01/04/2023]
Abstract
Multiple Sclerosis (MS) is one of the leading causes of non-traumatic neurological disability among young adults. Due to its complex pathology and the lack of reliable disease models, there are no effective therapies for MS to prevent neurodegeneration or promote neuroprotection, and hence stop disease progression. The emergence of induced pluripotent stem cells (iPSC) has allowed the generation of patient-specific neural cell types for disease modelling, drug screening, and cell therapy. In this review, the challenges related with the use of iPSC-derived cells in MS are discussed, with a special focus on the functional studies performed, limitations and future perspectives.
Collapse
Affiliation(s)
- Júlia Martínez-Larrosa
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Clara Matute-Blanch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
28
|
Mozafari S, Baron-Van Evercooren A. Human stem cell-derived oligodendrocytes: From humanized animal models to cell therapy in myelin diseases. Semin Cell Dev Biol 2020; 116:53-61. [PMID: 33082116 DOI: 10.1016/j.semcdb.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are main targets in demyelinating and dysmyelinating diseases of the central nervous system (CNS), but are also involved in accidental, neurodegenerative and psychiatric disorders. The underlying pathology of these diseases is not fully understood and treatments are still lacking. The recent discovery of the induced pluripotent stem cell (iPSC) technology has open the possibility to address the biology of human oligodendroglial cells both in the dish and in vivo via engraftment in animal models, and paves the way for the development of treatment for myelin disorders. In this review, we make a short overview of the different sources human oligodendroglial cells, and animal models available for pre-clinical cell therapy. We discuss the anatomical and functional benefit of grafted iPSC-progenitors over their brain counterparts, their use in disease modeling and the missing gaps that still prevent to study their biology in the most integrated way, and to translate iPSC-stem cell based therapy to the clinic.
Collapse
Affiliation(s)
- Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France.
| |
Collapse
|
29
|
Ciciriello AJ, Smith DR, Munsell MK, Boyd SJ, Shea LD, Dumont CM. Acute Implantation of Aligned Hydrogel Tubes Supports Delayed Spinal Progenitor Implantation. ACS Biomater Sci Eng 2020; 6:5771-5784. [DOI: 10.1021/acsbiomaterials.0c00844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew J. Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| | - Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Sydney J. Boyd
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, Michigan 48109, United States
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| |
Collapse
|
30
|
iPS-Derived Early Oligodendrocyte Progenitor Cells from SPMS Patients Reveal Deficient In Vitro Cell Migration Stimulation. Cells 2020; 9:cells9081803. [PMID: 32751289 PMCID: PMC7463559 DOI: 10.3390/cells9081803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
The most challenging aspect of secondary progressive multiple sclerosis (SPMS) is the lack of efficient regenerative response for remyelination, which is carried out by the endogenous population of adult oligoprogenitor cells (OPCs) after proper activation. OPCs must proliferate and migrate to the lesion and then differentiate into mature oligodendrocytes. To investigate the OPC cellular component in SPMS, we developed induced pluripotent stem cells (iPSCs) from SPMS-affected donors and age-matched controls (CT). We confirmed their efficient and similar OPC differentiation capacity, although we reported SPMS-OPCs were transcriptionally distinguishable from their CT counterparts. Analysis of OPC-generated conditioned media (CM) also evinced differences in protein secretion. We further confirmed SPMS-OPC CM presented a deficient capacity to stimulate OPC in vitro migration that can be compensated by exogenous addition of specific components. Our results provide an SPMS-OPC cellular model and encouraging venues to study potential cell communication deficiencies in the progressive form of multiple sclerosis (MS) for future treatment strategies.
Collapse
|
31
|
Villoslada P, Steinman L. New targets and therapeutics for neuroprotection, remyelination and repair in multiple sclerosis. Expert Opin Investig Drugs 2020; 29:443-459. [DOI: 10.1080/13543784.2020.1757647] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Pablo Villoslada
- Department of Psychiatry and Behavioural Sciences & Department of Neurology and Neurological Sciences, Stanford University, California, CA, USA
| | - Lawrence Steinman
- Department of Psychiatry and Behavioural Sciences & Department of Neurology and Neurological Sciences, Stanford University, California, CA, USA
| |
Collapse
|
32
|
Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res 2020; 1729:146615. [DOI: 10.1016/j.brainres.2019.146615] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
|
33
|
Imamura O, Arai M, Dateki M, Oishi K, Takishima K. Donepezil-induced oligodendrocyte differentiation is mediated through estrogen receptors. J Neurochem 2019; 155:494-507. [PMID: 31778582 DOI: 10.1111/jnc.14927] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/24/2022]
Abstract
Loss of oligodendrocytes, the myelin-forming cells of the central nervous system, and subsequent failure of myelin development result in serious neurological disorders such as multiple sclerosis. Using primary mouse embryonic neural stem cells (NSCs), we previously demonstrated that donepezil, an acetylcholinesterase inhibitor developed for the treatment of Alzheimer's disease, stimulates the differentiation of NSCs into oligodendrocytes and neurons, albeit at the expense of astrogenesis. However, the precise mechanisms underlying donepezil-induced differentiation remain unclear. In this study, we aimed at elucidating the molecular pathways contributing to donepezil-induced differentiation of mouse-induced pluripotent stem cell-derived neural stem cells (miPSC-NSCs). We used cell-based reporter gene arrays to investigate effects of donepezil on differentiation of miPSC-NSCs. Subsequently, we assessed the molecular pathway underlying donepezil action on differentiation of miPSC-NSCs into mature oligodendrocytes. Donepezil increased the transcriptional activity of estrogen response element under differentiating conditions. Moreover, estrogen receptors α (ERα) and β (ERβ) were highly expressed in MBP-positive mature oligodendrocytes. The ER antagonist ICI 182,780 abrogated the number of MBP-positive oligodendrocytes induced by donepezil, but showed no effect on the differentiation of miPSC-NSCs into Tuj1-positive neurons and GFAP-positive astrocytes. Furthermore, the donepezil-induced generation of mature oligodendrocytes from miPSC-NSC was significantly attenuated by antagonists and siRNA targeting ERα and ERβ. In conclusion, we demonstrated, for the first time, that donepezil-induced oligodendrogenesis is mediated through both ER subtypes, ERα and ERβ. Cover Image for this issue: https://doi.org/10.1111/jnc.14771.
Collapse
Affiliation(s)
- Osamu Imamura
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Masaaki Arai
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Minori Dateki
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kazuhiko Oishi
- Department of Pharmacology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Kunio Takishima
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
34
|
Oliveira AG, Gonçalves M, Ferreira H, M Neves N. Growing evidence supporting the use of mesenchymal stem cell therapies in multiple sclerosis: A systematic review. Mult Scler Relat Disord 2019; 38:101860. [PMID: 31765999 DOI: 10.1016/j.msard.2019.101860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/29/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) typically arises in early/middle adulthood and is characterized by a progressive disability of the central nervous system (CNS). Currently approved therapies do not promote tissue repair or stop disease progression. Emerging data demonstrate that stem cells present a great potential in regenerative medicine and, consequently, have also been widely investigated as a potential treatment for MS. Therefore, the aim of this study was to conduct a systematic review to inquire into the safety, tolerability, and efficacy of mesenchymal stem cells (MSCs) therapies in MS. METHODS Three electronic databases (Web of Science, PubMed, and Cochrane) were searched from April until June 2019. Clinical trials or case reports with information related to the effects of MSC therapies in MS patients were considered for this review. RESULTS 10 manuscripts were selected, namely 7 uncontrolled clinical trials, 2 randomized controlled clinical trials, and 1 case report. The overall quality of the studies was considered good. Besides minor adverse events (AEs), it was reported one case of encephalopathy with seizures and two cases of iatrogenic meningitis, which were not related to the treatment, but with the administration route. The analyses of the expanded disability status scale (EDSS) in the uncontrolled clinical trials demonstrated that 48 patients improved, 39 maintained and 16 worsened their clinical condition. Regarding the randomized studies, one did not show statistically significant variations in the mean EDSS score and in the other the mean EDSS score was statistically significantly lower for the experimental group. The case report also showed an improvement in the EDSS score. CONCLUSIONS MSCs transplantation proved to be a safe and tolerable therapy. Their potential therapeutic benefits were also validated. However, larger placebo controlled blinded clinical trials will be required to establish the long term safety and efficacy profile of these therapies for MS. Their translation into the clinical practice can provide a new hope for the patients of this highly debilitating disease.
Collapse
Affiliation(s)
| | - Margarida Gonçalves
- Medicine School, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Helena Ferreira
- 3B's Research Group, I3B's - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Nuno M Neves
- 3B's Research Group, I3B's - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães 4805-017, Portugal.
| |
Collapse
|
35
|
Moura RP, Sarmento B. Therapeutic Approaches toward Multiple Sclerosis: Where Do We Stand and Where Are We Headed? ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Rui Pedro Moura
- CESPU – Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Rua Central de Gandra 1317 4585‐116 Gandra Portugal
| | - Bruno Sarmento
- CESPU – Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Rua Central de Gandra 1317 4585‐116 Gandra Portugal
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do Porto Rua Alfredo Allen 208 4200‐135 Porto Portugal
- INEB – Instituto de Engenharia BiomédicaUniversidade do Porto Rua Alfredo Allen 208 4200‐135 Porto Portugal
| |
Collapse
|
36
|
Convergence of human cellular models and genetics to study neural stem cell signaling to enhance central nervous system regeneration and repair. Semin Cell Dev Biol 2019; 95:84-92. [PMID: 31310810 DOI: 10.1016/j.semcdb.2019.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 05/24/2019] [Accepted: 07/05/2019] [Indexed: 01/19/2023]
Abstract
Human central nervous system (CNS) regeneration is considered the holy grail of neuroscience research, and is one of the most pressing and difficult questions in biology and science. Despite more than 20 years of work in the field of neural stem cells (NSCs), the area remains in its infancy as our understanding of the fundamental mechanisms that can be leveraged to improve CNS regeneration in neurological diseases is still growing. Here, we focus on the recent lessons from lower organism CNS regeneration genetics and how such findings are starting to illuminate our understanding of NSC signaling pathways in humans. These findings will allow us to improve upon our knowledge of endogenous NSC function, the utility of exogenous NSCs, and the limitations of NSCs as therapeutic vehicles for providing relief from devastating human neurological diseases. We also discuss the limitations of activating NSC signaling for CNS repair in humans, especially the potential for tumor formation. Finally, we will review the recent advances in new culture techniques, including patient-derived cells and cerebral organoids to model the genetic regulation of signaling pathways controlling the function of NSCs during injury and disease states.
Collapse
|
37
|
Autologous Hematopoietic Cell Transplantation in Multiple Sclerosis: Changing Paradigms in the Era of Novel Agents. Stem Cells Int 2019; 2019:5840286. [PMID: 31341484 PMCID: PMC6612973 DOI: 10.1155/2019/5840286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/22/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022] Open
Abstract
Autologous hematopoietic stem cell transplantation (AHSCT) is established as a standard of care for diseases ranging from hematological malignancies to other neoplastic pathologies and severe immunological deficiencies. In April 1995, our group performed the first AHSCT in progressive multiple sclerosis (MS). Since then, a plethora of studies have been published with encouraging but controversial results. Major challenges in the field include appropriate patient selection, improvements in AHSCT procedure, and timing of this treatment modality. Beyond AHSCT, several new intravenous or oral agents have been developed and approved over the last 20 years in MS. The emergence of multiple effective therapies for MS has created a challenging scenario for both treating physicians and patients. Novel cell-based therapies other than AHSCT are also currently investigated in MS patients with promising results. Our review is aimed at summarizing state-of-the-art knowledge on basic principles and results of AHSCT in MS and its role compared to novel agents.
Collapse
|
38
|
Imitola J. Regenerative neuroimmunology: The impact of immune and neural stem cell interactions for translation in neurodegeneration and repair. J Neuroimmunol 2019; 331:1-3. [PMID: 31023492 DOI: 10.1016/j.jneuroim.2019.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
39
|
Cuascut FX, Hutton GJ. Stem Cell-Based Therapies for Multiple Sclerosis: Current Perspectives. Biomedicines 2019; 7:biomedicines7020026. [PMID: 30935074 PMCID: PMC6631931 DOI: 10.3390/biomedicines7020026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative autoimmune disease of the central nervous system (CNS). Disease-modifying therapies (DMT) targeting inflammation have been shown to reduce disease activity in patients with relapsing–remitting MS (RRMS). The current therapeutic challenge is to find an effective treatment to halt disease progression and reverse established neural damage. Stem cell-based therapies have emerged to address this dilemma. Several types of stem cells have been considered for clinical use, such as autologous hematopoietic (aHSC), mesenchymal (MSC), neuronal (NSC), human embryonic (hESC), and induced pluripotent (iPSC) stem cells. There is convincing evidence that immunoablation followed by hematopoietic therapy (aHSCT) has a high efficacy for suppressing inflammatory MS activity and improving neurological disability in patients with RRMS. In addition, MSC therapy may be a safe and tolerable treatment, but its clinical value is still under evaluation. Various studies have shown early promising results with other cellular therapies for CNS repair and decreasing inflammation. In this review, we discuss the current knowledge and limitations of different stem cell-based therapies for the treatment of patients with MS.
Collapse
Affiliation(s)
- Fernando X Cuascut
- Baylor College of Medicine, Maxine Mesigner Multiple Sclerosis Center, Houston, TX 77030, USA.
| | - George J Hutton
- Baylor College of Medicine, Maxine Mesigner Multiple Sclerosis Center, Houston, TX 77030, USA.
| |
Collapse
|
40
|
Cellular senescence in progenitor cells contributes to diminished remyelination potential in progressive multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:9030-9039. [PMID: 30910981 PMCID: PMC6500153 DOI: 10.1073/pnas.1818348116] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We identify cellular senescence occurring in neural progenitor cells (NPCs) from primary progressive multiple sclerosis (PPMS). In this study, senescent progenitor cells were identified within demyelinated white matter lesions in progressive MS (PMS) autopsy tissue, and induced pluripotent stem-derived NPCs from patients with PPMS were found to express cellular senescence markers compared with age-matched control NPCs. Reversal of this cellular senescence phenotype, by treatment with rapamycin, restored PPMS NPC-mediated support for oligodendrocyte (OL) maturation. Proteomic and histological analyses identify senescent progenitor cells in PMS as a source of high-mobility group box-1, which limits maturation and promotes transcriptomic changes in OLs. These findings provide evidence that cellular senescence is an active process in PMS that may contribute to limited remyelination in disease. Cellular senescence is a form of adaptive cellular physiology associated with aging. Cellular senescence causes a proinflammatory cellular phenotype that impairs tissue regeneration, has been linked to stress, and is implicated in several human neurodegenerative diseases. We had previously determined that neural progenitor cells (NPCs) derived from induced pluripotent stem cell (iPSC) lines from patients with primary progressive multiple sclerosis (PPMS) failed to promote oligodendrocyte progenitor cell (OPC) maturation, whereas NPCs from age-matched control cell lines did so efficiently. Herein, we report that expression of hallmarks of cellular senescence were identified in SOX2+ progenitor cells within white matter lesions of human progressive MS (PMS) autopsy brain tissues and iPS-derived NPCs from patients with PPMS. Expression of cellular senescence genes in PPMS NPCs was found to be reversible by treatment with rapamycin, which then enhanced PPMS NPC support for oligodendrocyte (OL) differentiation. A proteomic analysis of the PPMS NPC secretome identified high-mobility group box-1 (HMGB1), which was found to be a senescence-associated inhibitor of OL differentiation. Transcriptome analysis of OPCs revealed that senescent NPCs induced expression of epigenetic regulators mediated by extracellular HMGB1. Lastly, we determined that progenitor cells are a source of elevated HMGB1 in human white matter lesions. Based on these data, we conclude that cellular senescence contributes to altered progenitor cell functions in demyelinated lesions in MS. Moreover, these data implicate cellular aging and senescence as a process that contributes to remyelination failure in PMS, which may impact how this disease is modeled and inform development of future myelin regeneration strategies.
Collapse
|
41
|
Davis SM, Collier LA, Goodwin S, Lukins DE, Powell DK, Pennypacker KR. Efficacy of leukemia inhibitory factor as a therapeutic for permanent large vessel stroke differs among aged male and female rats. Brain Res 2018; 1707:62-73. [PMID: 30445025 DOI: 10.1016/j.brainres.2018.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023]
Abstract
Preclinical studies using rodent models of stroke have had difficulty in translating their results to human patients. One possible factor behind this inability is the lack of studies utilizing aged rodents of both sexes. Previously, this lab showed that leukemia inhibitory factor (LIF) promoted recovery after stroke through antioxidant enzyme upregulation. This study examined whether LIF promotes neuroprotection in aged rats of both sexes. LIF did not reduce tissue damage in aged animals, but LIF-treated female rats showed partial motor skill recovery. The LIF receptor (LIFR) showed membrane localization in young male and aged rats of both sexes after stroke. Although LIF increased neuronal LIFR expression in vitro, it did not increase LIFR in the aged brain. Levels of LIFR protein in brain tissue were significantly downregulated between young males and aged males/females at 72 h after stroke. These results demonstrated that low LIFR expression reduces the neuroprotective efficacy of LIF in aged rodents of both sexes. Furthermore, the ability of LIF to promote motor improvement is dependent upon sex in aged rodents.
Collapse
Affiliation(s)
- Stephanie M Davis
- Department of Neurology, University of Kentucky, 741 S. Limestone, Lexington, KY 40536, United States.
| | - Lisa A Collier
- Department of Neurology, University of Kentucky, 741 S. Limestone, Lexington, KY 40536, United States.
| | - Sarah Goodwin
- Department of Neurology, University of Kentucky, 741 S. Limestone, Lexington, KY 40536, United States.
| | - Douglas E Lukins
- Department of Radiology, University of Kentucky, 800 Rose St., Lexington, KY 40536, United States.
| | - David K Powell
- Spinal Cord and Brain Injury Research Center, 741 S. Limestone, Lexington, KY 40536, United States.
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, 741 S. Limestone, Lexington, KY 40536, United States; Department of Neuroscience, University of Kentucky, 800 Rose St., Lexington, KY 40536, United States.
| |
Collapse
|
42
|
Dumont CM, Munsell MK, Carlson MA, Cummings BJ, Anderson AJ, Shea LD. Spinal Progenitor-Laden Bridges Support Earlier Axon Regeneration Following Spinal Cord Injury. Tissue Eng Part A 2018; 24:1588-1602. [PMID: 30215293 DOI: 10.1089/ten.tea.2018.0053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPACT STATEMENT Spinal cord injury (SCI) results in loss of tissue innervation below the injury. Spinal progenitors have a greater ability to repair the damage and can be injected into the injury, but their regenerative potential is hampered by their poor survival after transplantation. Biomaterials can create a cell delivery platform and generate a more hospitable microenvironment for the progenitors within the injury. In this work, polymeric bridges are used to deliver embryonic spinal progenitors to the injury, resulting in increased progenitor survival and subsequent regeneration and functional recovery, thus demonstrating the importance of combined therapeutic approaches for SCI.
Collapse
Affiliation(s)
- Courtney M Dumont
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Mary K Munsell
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Mitchell A Carlson
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Brian J Cummings
- 2 Institute for Memory Impairments and Neurological Disorders (iMIND), University of California , Irvine, California.,3 Sue and Bill Gross Stem Cell Research Center, University of California , Irvine, California.,4 Department of Anatomy and Neurobiology and University of California , Irvine, California.,5 Department of Physical Medicine and Rehabilitation, University of California , Irvine, California
| | - Aileen J Anderson
- 2 Institute for Memory Impairments and Neurological Disorders (iMIND), University of California , Irvine, California.,3 Sue and Bill Gross Stem Cell Research Center, University of California , Irvine, California.,4 Department of Anatomy and Neurobiology and University of California , Irvine, California.,5 Department of Physical Medicine and Rehabilitation, University of California , Irvine, California
| | - Lonnie D Shea
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan.,6 Department of Chemical Engineering, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
43
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
44
|
Metcalfe SM, Bickerton S, Fahmy T. Neurodegenerative Disease: A Perspective on Cell-Based Therapy in the New Era of Cell-Free Nano-Therapy. Curr Pharm Des 2018; 23:776-783. [PMID: 27924726 DOI: 10.2174/1381612822666161206141744] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Neurodegenerative diseases (NDD) result in irreversible loss of neurons. Dementia develops when disease-induced neuronal loss becomes sufficient to impair both memory and cognitive functioning and, globally, dementia is increasing to epidemic proportions as populations age. In the current era of regenerative medicine intense activity is asking, can loss of endogenous neurons be compensated by replacement with exogenously derived cells that have either direct, or indirect, neurogenic capacity? But, more recently, excitement is growing around an emerging alternative to the cell-based approach - here nanotechnology for targeted delivery of growth factor aims to support and expand resident central nervous system (CNS) stem cells for endogenous repair. The concept of a high volume, off-the-shelf nano-therapeutic able to rejuvenate the endogenous neuroglia of the CNS is highly attractive, providing a simple solution to the complex challenges posed by cell-based regenerative medicine. The role of inflammation as an underlying driver of NDD is also considered where anti-inflammatory approaches are candidates for therapy. Indeed, cell-based therapy and/or nanotherapy may protect against inflammation to support both immune quiescence and neuronal survival in the CNS - key targets for treating NDD with the potential to reduce or even stop the cascading pathogenesis and disease progression, possibly promoting some repair where disease is treated early. By design, nanoparticles can be formulated to cross the blood brain barrier (BBB) enabling sustained delivery of neuro-protective agents for sufficient duration to reset neuro-immune homeostasis. Proven safe and efficacious, it is now urgent to deliver nano-medicine (NanoMed) as a scalable approach to treat NDD, where key stakeholders are the patients and the global economy.
Collapse
Affiliation(s)
- Su M Metcalfe
- Cambridge University Hospitals Trust Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Sean Bickerton
- Yale School of Engineering and Applied Science and Yale School of Medicine, 55 Prospect Street, New Haven, CT, 06511, United States
| | - Tarek Fahmy
- Yale School of Engineering and Applied Science and Yale School of Medicine, 55 Prospect Street, New Haven, CT, 06511, United States
| |
Collapse
|
45
|
Selective killing of spinal cord neural stem cells impairs locomotor recovery in a mouse model of spinal cord injury. J Neuroinflammation 2018; 15:58. [PMID: 29475438 PMCID: PMC5824446 DOI: 10.1186/s12974-018-1085-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/01/2018] [Indexed: 01/19/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating condition mainly deriving from a traumatic damage of the spinal cord (SC). Immune cells and endogenous SC-neural stem cells (SC-NSCs) play a critical role in wound healing processes, although both are ineffective to completely restore tissue functioning. The role of SC-NSCs in SCI and, in particular, whether such cells can interplay with the immune response are poorly investigated issues, although mechanisms governing such interactions might open new avenues to develop novel therapeutic approaches. Methods We used two transgenic mouse lines to trace as well as to kill SC-NSCs in mice receiving SCI. We used Nestin CreERT2 mice to trace SC-NSCs descendants in the spinal cord of mice subjected to SCI. While mice carrying the suicide gene thymidine kinase (TK) along with the GFP reporter, under the control of the Nestin promoter regions (NestinTK mice) were used to label and selectively kill SC-NSCs. Results We found that SC-NSCs are capable to self-activate after SCI. In addition, a significant worsening of clinical and pathological features of SCI was observed in the NestinTK mice, upon selective ablation of SC-NSCs before the injury induction. Finally, mice lacking in SC-NSCs and receiving SCI displayed reduced levels of different neurotrophic factors in the SC and significantly higher number of M1-like myeloid cells. Conclusion Our data show that SC-NSCs undergo cell proliferation in response to traumatic spinal cord injury. Mice lacking SC-NSCs display overt microglia activation and exaggerate expression of pro-inflammatory cytokines. The absence of SC-NSCs impaired functional recovery as well as neuronal and oligodendrocyte cell survival. Collectively our data indicate that SC-NSCs can interact with microglia/macrophages modulating their activation/responses and that such interaction is importantly involved in mechanisms leading tissue recovery. Electronic supplementary material The online version of this article (10.1186/s12974-018-1085-9) contains supplementary material, which is available to authorized users.
Collapse
|
46
|
Guarnieri FC, Bellani S, Yekhlef L, Bergamaschi A, Finardi A, Fesce R, Pozzi D, Monzani E, Fornasiero EF, Matteoli M, Martino G, Furlan R, Taverna S, Muzio L, Valtorta F. Synapsin I deletion reduces neuronal damage and ameliorates clinical progression of experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 68:197-210. [PMID: 29066310 DOI: 10.1016/j.bbi.2017.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 11/20/2022] Open
Abstract
The classical view of multiple sclerosis (MS) pathogenesis states that inflammation-mediated demyelination is responsible for neuronal damage and loss. However, recent findings show that impairment of neuronal functions and demyelination can be independent events, suggesting the coexistence of other pathogenic mechanisms. Due to the inflammatory milieu, subtle alterations in synaptic function occur, which are probably at the basis of the early cognitive decline that often precedes the neurodegenerative phases in MS patients. In particular, it has been reported that inflammation enhances excitatory synaptic transmission while it decreases GABAergic transmission in vitro and ex vivo. This evidence points to the idea that an excitation/inhibition imbalance occurs in the inflamed MS brain, even though the exact molecular mechanisms leading to this synaptic dysfunction are as yet not completely clear. Along this line, we observed that acute treatment of primary hippocampal neurons in culture with pro-inflammatory cytokines leads to an increased phosphorylation of synapsin I (SynI) by ERK1/2 kinase and to an increase in the frequency of spontaneous synaptic vesicle release events, which is prevented by SynI deletion. In vivo, the ablation of SynI expression is protective in terms of disease progression and neuronal damage in the experimental autoimmune encephalomyelitis mouse model of MS. Our results point to a possible key role in MS pathogenesis of the neuronal protein SynI, a regulator of excitation/inhibition balance in neuronal networks.
Collapse
Affiliation(s)
- Fabrizia C Guarnieri
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Serena Bellani
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Latefa Yekhlef
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Andrea Bergamaschi
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Annamaria Finardi
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Riccardo Fesce
- Centre of Neuroscience and DISTA, University of Insubria, Via Ravasi 2, 21100 Varese, Italy
| | - Davide Pozzi
- Humanitas Clinical and Research Centre, Via Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Elena Monzani
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Eugenio F Fornasiero
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Centre, Via Manzoni 113, 20089 Rozzano, Milan, Italy; CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milan, Italy
| | - Gianvito Martino
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Roberto Furlan
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Stefano Taverna
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Luca Muzio
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
47
|
Xu L, Long J, Shi C, Zhang N, Lv Y, Feng J, Xuan A, He X, Li Q, Bai Y, Liu S, Long D. Effect of leukocyte inhibitory factor on neuron differentiation from human induced pluripotent stem cell-derived neural precursor cells. Int J Mol Med 2018; 41:2037-2049. [PMID: 29393372 PMCID: PMC5810244 DOI: 10.3892/ijmm.2018.3418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/05/2018] [Indexed: 12/27/2022] Open
Abstract
Direct derivation of human induced pluripotent stem cells into neural precursor cells and differentiation of these into neurons holds great promise in the cell therapy of neuro-degenerative diseases. However, the availability and survival rate of neurons requires improvement. In the present study, it was found that the addition of 5 ng/ml leukocyte inhibitory factor (LIF) during the process of differentiation significantly improved the expression of neuron-specific class III β-tubulin (TUJ1) and microtubule-associated protein 2 (MAP2), as detected by immunofluorescence and western blotting. In addition, LIF improved the cell viability, increased the expression of phosphorylated-protein kinase B (AKT), downregulated the expression of proinflammatory cytokines, including interleukin-1α (IL-1α) and tumor necrosis factor-α (TNF-α), and upregulated the expression of anti-inflammatory cytokines, including interleukin-10 (IL-10) and transforming growth factor-β (TGF-β). After adding the phosphatidylinositol 3-kinase (PI3K)/AKT signaling inhibitor LY294002 or wortmannin to the LIF differentiation group, LIF-induced changes in the protein expression of TUJ1 and MAP2 were reversed, but this effect could not be prevented by rapamycin, a mechanistic target of rapamycin signaling inhibitor. The expression of cytokines associated with inflammation and cell viability was reversed by LY294002 and wortmannin, but not by rapamycin. In conclusion, LIF could improve neuronal differentiation and survival through the activation of PI3K/AKT signaling and the anti-inflammatory effect. The anti-inflammatory effect may be mediated by the activation of PI3K/AKT.
Collapse
Affiliation(s)
- Liping Xu
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Jingyi Long
- Institute of Neuroscience and The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Chun Shi
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Nianping Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Ying Lv
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Junda Feng
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Aiguo Xuan
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiaosong He
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Qingqing Li
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Yinshan Bai
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Shanshan Liu
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Dahong Long
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Following the establishment of a number of successful immunomodulatory treatments for multiple sclerosis, current research focuses on the repair of existing damage. RECENT FINDINGS Promotion of regeneration is particularly important for demyelinated areas with degenerated or functionally impaired axons of the central nervous system's white and gray matter. As the protection and generation of new oligodendrocytes is a key to the re-establishment of functional connections, adult oligodendrogenesis and myelin reconstitution processes are of primary interest. Moreover, understanding, supporting and promoting endogenous repair activities such as mediated by resident oligodendroglial precursor or adult neural stem cells are currently thought to be a promising approach toward the development of novel regenerative therapies. SUMMARY This review summarizes recent developments and findings related to pharmacological myelin repair as well as to the modulation/application of stem cells with the aim to restore defective myelin sheaths.
Collapse
|
49
|
Genc B, Bozan HR, Genc S, Genc K. Stem Cell Therapy for Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:145-174. [PMID: 30039439 DOI: 10.1007/5584_2018_247] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system (CNS). It is characterized by demyelination and neuronal loss that is induced by attack of autoreactive T cells to the myelin sheath and endogenous remyelination failure, eventually leading to functional neurological disability. Although recent evidence suggests that MS relapses are induced by environmental and exogenous triggers such as viral infections in a genetic background, its very complex pathogenesis is not completely understood. Therefore, the efficiency of current immunosuppression-based therapies of MS is too low, and emerging disease-modifying immunomodulatory agents such as fingolimod and dimethyl fumarate cannot stop progressive neurodegenerative process. Thus, the cell replacement therapy approach that aims to overcome neuronal cell loss and remyelination failure and to increase endogenous myelin repair capacity is considered as an alternative treatment option. A wide variety of preclinical studies, using experimental autoimmune encephalomyelitis model of MS, have recently shown that grafted cells with different origins including mesenchymal stem cells (MSCs), neural precursor and stem cells, and induced-pluripotent stem cells have the ability to repair CNS lesions and to recover functional neurological deficits. The results of ongoing autologous hematopoietic stem cell therapy studies, with the advantage of peripheral administration to the patients, have suggested that cell replacement therapy is also a feasible option for immunomodulatory treatment of MS. In this chapter, we overview cell sources and applications of the stem cell therapy for treatment of MS. We also discuss challenges including those associated with administration route, immune responses to grafted cells, integration of these cells to existing neural circuits, and risk of tumor growth. Finally, future prospects of stem cell therapy for MS are addressed.
Collapse
Affiliation(s)
- Bilgesu Genc
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Hemdem Rodi Bozan
- School of Medicine, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey.,Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey.
| |
Collapse
|
50
|
Leferink PS, Heine VM. The Healthy and Diseased Microenvironments Regulate Oligodendrocyte Properties: Implications for Regenerative Medicine. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:39-52. [PMID: 29024633 DOI: 10.1016/j.ajpath.2017.08.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/12/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023]
Abstract
White matter disorders are characterized by deficient myelin or myelin loss, lead to a range of neurologic dysfunctions, and can result in early death. Oligodendrocytes, which are responsible for white matter formation, are the first targets for treatment. However, many studies indicate that failure of white matter repair goes beyond the intrinsic incapacity of oligodendrocytes to (re)generate myelin and that failed interactions with neighboring cells or factors in the diseased microenvironment can underlie white matter defects. Moreover, most of the white matter disorders show specific white matter pathology caused by different disease mechanisms. Herein, we review the factors within the cellular and the extracellular microenvironment regulating oligodendrocyte properties and discuss stem cell tools to identify microenvironmental factors of importance to the development of improved regenerative medicine for patients with white matter disorders.
Collapse
Affiliation(s)
- Prisca S Leferink
- Department of Pediatrics/Child Neurology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|