1
|
Brugarolas P, Sánchez-Rodríguez JE, Tsai HM, Basuli F, Cheng SH, Zhang X, Caprariello AV, Lacroix JJ, Freifelder R, Murali D, DeJesus O, Miller RH, Swenson RE, Chen CT, Herscovitch P, Reich DS, Bezanilla F, Popko B. Development of a PET radioligand for potassium channels to image CNS demyelination. Sci Rep 2018; 8:607. [PMID: 29330383 PMCID: PMC5766510 DOI: 10.1038/s41598-017-18747-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/16/2017] [Indexed: 01/22/2023] Open
Abstract
Central nervous system (CNS) demyelination represents the pathological hallmark of multiple sclerosis (MS) and contributes to other neurological conditions. Quantitative and specific imaging of demyelination would thus provide critical clinical insight. Here, we investigated the possibility of targeting axonal potassium channels to image demyelination by positron emission tomography (PET). These channels, which normally reside beneath the myelin sheath, become exposed upon demyelination and are the target of the MS drug, 4-aminopyridine (4-AP). We demonstrate using autoradiography that 4-AP has higher binding in non-myelinated and demyelinated versus well-myelinated CNS regions, and describe a fluorine-containing derivative, 3-F-4-AP, that has similar pharmacological properties and can be labeled with 18F for PET imaging. Additionally, we demonstrate that [18F]3-F-4-AP can be used to detect demyelination in rodents by PET. Further evaluation in Rhesus macaques shows higher binding in non-myelinated versus myelinated areas and excellent properties for brain imaging. Together, these data indicate that [18F]3-F-4-AP may be a valuable PET tracer for detecting CNS demyelination noninvasively.
Collapse
Affiliation(s)
- Pedro Brugarolas
- Department of Neurology, University of Chicago, Chicago, IL, USA. .,Massachusetts General Hospital, Boston, MA, USA.
| | - Jorge E Sánchez-Rodríguez
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.,Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Hsiu-Ming Tsai
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | - Falguni Basuli
- Imaging Probe Development Center, NIH/NHLBI, Bethesda, MD, USA
| | - Shih-Hsun Cheng
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | - Xiang Zhang
- Imaging Probe Development Center, NIH/NHLBI, Bethesda, MD, USA
| | - Andrew V Caprariello
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA.,University of Calgary, Calgary, Alberta, Canada
| | - Jerome J Lacroix
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.,Western University of Health Sciences, Pomona, CA, USA
| | | | - Dhanabalan Murali
- Department of Medical Physics, University of Wisconsin at Madison, Madison, WI, USA
| | - Onofre DeJesus
- Department of Medical Physics, University of Wisconsin at Madison, Madison, WI, USA
| | - Robert H Miller
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA.,George Washington University, Washington, DC, USA
| | - Rolf E Swenson
- Imaging Probe Development Center, NIH/NHLBI, Bethesda, MD, USA
| | - Chin-Tu Chen
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | | | - Daniel S Reich
- Translational Neuroradiology Section, NIH/NINDS, Bethesda, MD, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Brian Popko
- Department of Neurology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Intracellular signaling pathway regulation of myelination and remyelination in the CNS. Exp Neurol 2016; 283:501-11. [PMID: 26957369 DOI: 10.1016/j.expneurol.2016.03.008] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/23/2022]
Abstract
The restoration of myelin sheaths on demyelinated axons remains a major obstacle in the treatment of multiple sclerosis (MS). Currently approved therapies work by modulating the immune system to reduce the number and rate of lesion formation but are only partially effective since they are not able to restore lost myelin. In the healthy CNS, myelin continues to be generated throughout life and spontaneous remyelination occurs readily in response to insults. In patients with MS, however, remyelination eventually fails, at least in part as a result of a failure of oligodendrocyte precursor cell (OPC) differentiation and the subsequent production of new myelin. A better understanding of the molecular mechanisms and signaling pathways that drive the process of myelin sheath formation is therefore important in order to speed the development of novel therapeutics designed to target remyelination. Here we review data supporting critical roles for three highly conserved intracellular signaling pathways: Wnt/β-catenin, PI3K/AKT/mTOR, and ERK/MAPK in the regulation of OPC differentiation and myelination both during development and in remyelination. Potential points of crosstalk between the three pathways and important areas for future research are also discussed.
Collapse
|
3
|
Combination of electroacupuncture and grafted mesenchymal stem cells overexpressing TrkC improves remyelination and function in demyelinated spinal cord of rats. Sci Rep 2015; 5:9133. [PMID: 25779025 PMCID: PMC5390924 DOI: 10.1038/srep09133] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/16/2015] [Indexed: 12/13/2022] Open
Abstract
This study attempted to graft neurotrophin-3 (NT-3) receptor (TrkC) gene modified mesenchymal stem cells (TrkC-MSCs) into the demyelinated spinal cord and to investigate whether electroacupuncture (EA) treatment could promote NT-3 secretion in the demyelinated spinal cord as well as further enhance grafted TrkC-MSCs to differentiate into oligodendrocytes, remyelination and functional recovery. Ethidium bromide (EB) was microinjected into the spinal cord of rats at T10 to establish a demyelinated model. Six groups of animals were prepared for the experiment: the sham, PBS, MSCs, MSCs+EA, TrkC-MSCs and TrkC-MSCs+EA groups. The results showed that TrkC-MSCs graft combined with EA treatment (TrkC-MSCs+EA group) significantly increased the number of OPCs and oligodendrocyte-like cells differentiated from MSCs. Immunoelectron microscopy showed that the oligodendrocyte-like cells differentiated from TrkC-MSCs formed myelin sheaths. Immunofluorescence histochemistry and Western blot analysis indicated that TrkC-MSCs+EA treatment could promote the myelin basic protein (MBP) expression and Kv1.2 arrangement trending towards the normal level. Furthermore, behavioural test and cortical motor evoked potentials detection demonstrated a significant functional recovery in the TrkC-MSCs+EA group. In conclusion, our results suggest that EA treatment can increase NT-3 expression, promote oligodendrocyte-like cell differentiation from TrkC-MSCs, remyelination and functional improvement of demyelinated spinal cord.
Collapse
|
4
|
Gesundheit B, Ashwood P, Keating A, Naor D, Melamed M, Rosenzweig JP. Therapeutic properties of mesenchymal stem cells for autism spectrum disorders. Med Hypotheses 2014; 84:169-77. [PMID: 25592283 DOI: 10.1016/j.mehy.2014.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
Recent studies of autism spectrum disorders (ASD) highlight hyperactivity of the immune system, irregular neuronal growth and increased size and number of microglia. Though the small sample size in many of these studies limits extrapolation to all individuals with ASD, there is mounting evidence of both immune and nervous system related pathogenesis in at least a subset of patients with ASD. Given the disturbing rise in incidence rates for ASD, and the fact that no pharmacological therapy for ASD has been approved by the Food and Drug Administration (FDA), there is an urgent need for new therapeutic options. Research in the therapeutic effects of mesenchymal stem cells (MSC) for other immunological and neurological conditions has shown promising results in preclinical and even clinical studies. MSC have demonstrated the ability to suppress the immune system and to promote neurogenesis with a promising safety profile. The working hypothesis of this paper is that the potentially synergistic ability of MSC to modulate a hyperactive immune system and its ability to promote neurogenesis make it an attractive potential therapeutic option specifically for ASD. Theoretical mechanisms of action will be suggested, but further research is necessary to support these hypothetical pathways. The choice of tissue source, type of cell, and most appropriate ages for therapeutic intervention remain open questions for further consideration. Concern over poor regulatory control of stem cell studies or treatment, and the unique ethical challenges that each child with ASD presents, demands that future research be conducted with particular caution before widespread use of the proposed therapeutic intervention is implemented.
Collapse
Affiliation(s)
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; Department of Medical Microbiology and Immunology, and the MIND Institute, University of California Davis, USA.
| | - Armand Keating
- Division of Hematology, University of Toronto, Cell Therapy Program, Princess Margaret Hospital, Toronto, Canada.
| | - David Naor
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | - Michal Melamed
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | | |
Collapse
|
5
|
Dimarino AM, Caplan AI, Bonfield TL. Mesenchymal stem cells in tissue repair. Front Immunol 2013; 4:201. [PMID: 24027567 PMCID: PMC3761350 DOI: 10.3389/fimmu.2013.00201] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/04/2013] [Indexed: 12/23/2022] Open
Abstract
The advent of mesenchymal stem cell (MSC)-based therapies for clinical therapeutics has been an exciting and new innovation for the treatment of a variety of diseases associated with inflammation, tissue damage, and subsequent regeneration and repair. Application-based ability to measure MSC potency and fate of the cells post-MSC therapy are the variables that confound the use of MSCs therapeutics in human diseases. An evaluation of MSC function and applications with attention to detail in the preparation as well as quality control and quality assurance are only as good as the assays that are developed. In vivo measures of efficacy and potency require an appreciation of the overall pathophysiology of the model and standardization of outcome measures. The new concepts of how MSC’s participate in the tissue regeneration and wound repair process and further, how this is impacted by estimates of efficacy and potency are important new topics. In this regard, this chapter will review some of the in vitro and in vivo assays for MSC function and activity and their application to the clinical arena.
Collapse
Affiliation(s)
- Amy M Dimarino
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University , Cleveland, OH , USA
| | | | | |
Collapse
|
6
|
Lindsay SL, Johnstone SA, Mountford JC, Sheikh S, Allan DB, Clark L, Barnett SC. Human mesenchymal stem cells isolated from olfactory biopsies but not bone enhance CNS myelination in vitro. Glia 2012; 61:368-82. [PMID: 23281012 DOI: 10.1002/glia.22440] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 10/11/2012] [Indexed: 01/09/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition with limited capacity for repair. Cell transplantation is a potential strategy to promote SCI repair with cells from the olfactory system being promising candidates. Although transplants of human olfactory mucosa (OM) are already ongoing in clinical trials, the repair potential of this tissue remains unclear. Previously, we identified mesenchymal-like stem cells that reside in the lamina propria (LP-MSCs) of rat and human OM. Little is known about these cells or their interactions with glia such as olfactory ensheathing cells (OECs), which would be co-transplanted with MSCs from the OM, or endogenous CNS glia such as oligodendrocytes. We have characterized, purified, and assessed the repair potential of human LP-MSCs by investigating their effect on glial cell biology with specific emphasis on CNS myelination in vitro. Purified LP-MSCs expressed typical bone marrow MSC (BM-MSC) markers, formed spheres, were clonogenic and differentiated into bone and fat. LP-MSC conditioned medium (CM) promoted oligodendrocyte precursor cell (OPC) and OEC proliferation and induced a highly branched morphology. LP-MSC-CM treatment caused OEC process extension. Both LP and BM-MSCs promoted OPC proliferation and differentiation, but only myelinating cultures treated with CM from LP and not BM-MSCs had a significant increase in myelination. Comparison with fibroblasts and contaminating OM fibroblast like-cells showed the promyelination effect was LP-MSC specific. Thus LP-MSCs harvested from human OM biopsies may be an important candidate for cell transplantation by contributing to the repair of SCI.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
7
|
Auletta JJ, Bartholomew AM, Maziarz RT, Deans RJ, Miller RH, Lazarus HM, Cohen JA. The potential of mesenchymal stromal cells as a novel cellular therapy for multiple sclerosis. Immunotherapy 2012; 4:529-47. [PMID: 22642335 PMCID: PMC3381871 DOI: 10.2217/imt.12.41] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease of the CNS for which only partially effective therapies exist. Intense research defining the underlying immune pathophysiology is advancing both the understanding of MS as well as revealing potential targets for disease intervention. Mesenchymal stromal cell (MSC) therapy has the potential to modulate aberrant immune responses causing demyelination and axonal injury associated with MS, as well as to repair and restore damaged CNS tissue and cells. This article reviews the pathophysiology underlying MS, as well as providing a cutting-edge perspective into the field of MSC therapy based upon the experience of authors intrinsically involved in MS and MSC basic and translational science research.
Collapse
Affiliation(s)
- Jeffery J Auletta
- National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
An experimental electro-acupuncture study in treatment of the rat demyelinated spinal cord injury induced by ethidium bromide. Neurosci Res 2011; 70:294-304. [PMID: 21470565 DOI: 10.1016/j.neures.2011.03.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 03/26/2011] [Accepted: 03/28/2011] [Indexed: 01/19/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are one of the potential treating tools for multiple sclerosis (MS). Therefore, the cell number and differentiation of OPCs in a demyelinated spinal cord are crucial for improvement of reparative process. In the present study, we investigated whether "Governor Vessel (GV)" electro-acupuncture (EA) could efficiently promote increase in cell number and differentiation of OPCs into oligodendrocytes, remyelination and functional recovery in the demyelinated spinal cord. The spinal cord of adult Sprague-Dawley rats was microinjected with ethidium bromide (EB) at T10, to establish a demyelinated model. Six groups of animals were performed for the experiment. After 15 days EA treatment, neurotrophin-3 (NT-3) level and number of NG2-positive OPCs were significantly increased. Compared with the sham group, more NG2-positive OPCs were distributed between neurofilament (NF)-positive nerve fibres or closely associated with them in the lesion site and nearby tissue. In rats given longer EA treatment for 30 days, the number of adenomatous polyposis coli (APC)-positive oligodendrocytes was increased. Concomitantly, the number of newly formed myelins was increased. This was coupled by increase in endogenous oligodendrocyte involved in myelin formation. Furthermore, behavioural test and spinal cord evoked potential detection demonstrated a significant functional recovery in the EA+EB day 30 group. Our results suggest EA treatment can promote NT-3 expression, increase the cell number and differentiation of endogenous OPCs, and remyelination in the demyelinated spinal cord as well as the functional improvement of demyelinated spinal cord.
Collapse
|
9
|
Faust PL, Kaye EM, Powers JM. Myelin lesions associated with lysosomal and peroxisomal disorders. Expert Rev Neurother 2010; 10:1449-66. [PMID: 20819015 DOI: 10.1586/ern.10.127] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Abnormalities of myelin are common in lysosomal and peroxisomal disorders. Most display a primary loss of myelin in which the myelin sheath and/or oligodendrocytes are selectively targeted by diverse pathogenetic processes. The most severe and, hence, clinically relevant are heritable diseases predominantly of infants and children, the leukodystrophies: metachromatic, globoid cell (Krabbe disease) and adreno-leukodystrophy. Our still limited understanding of these diseases has derived from multiple sources: originally, neurological-neuropathologic-neurochemical correlative studies of the natural disease in humans or other mammals, which has been enhanced by more sophisticated and contemporary techniques of cell and molecular biology. Transgenic mouse models seem to be the most promising methodology, allowing the examination of the cellular role of lysosomes and peroxisomes for formation and maintenance of both myelin and axons, and providing initial platforms to evaluate therapies. Treatment options are woefully inadequate and in their nascent stages, but still inspire some hope for the future.
Collapse
Affiliation(s)
- Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | | | | |
Collapse
|
10
|
Bonfield TL, Koloze M, Lennon DP, Zuchowski B, Yang SE, Caplan AI. Human mesenchymal stem cells suppress chronic airway inflammation in the murine ovalbumin asthma model. Am J Physiol Lung Cell Mol Physiol 2010; 299:L760-70. [PMID: 20817776 DOI: 10.1152/ajplung.00182.2009] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Allogeneic human mesenchymal stem cells (hMSCs) introduced intravenously can have profound anti-inflammatory activity resulting in suppression of graft vs. host disease as well as regenerative events in the case of stroke, infarct, spinal cord injury, meniscus regeneration, tendinitis, acute renal failure, and heart disease in human and animal models of these diseases. hMSCs produce bioactive factors that provide molecular cuing for: 1) immunosuppression of T cells; 2) antiscarring; 3) angiogenesis; 4) antiapoptosis; and 5) regeneration (i.e., mitotic for host-derived progenitor cells). Studies have shown that hMSCs have profound effects on the immune system and are well-tolerated and therapeutically active in immunocompetent rodent models of multiple sclerosis and stroke. Furthermore, intravenous administration of MSCs results in pulmonary localization. Asthma is a major debilitating pulmonary disease that impacts in excess of 150 million people in the world with uncontrolled asthma potentially leading to death. In addition, the socioeconomic impact of asthma-associated illnesses at the pediatric and adult level are in the millions of dollars in healthcare costs and lost days of work. hMSCs may provide a viable multiaction therapeutic for this inflammatory lung disease by secreting bioactive factors or directing cellular activity. Our studies show the effectiveness and specificity of the hMSCs on decreasing chronic airway inflammation associated with the murine ovalbumin model of asthma. In addition, the results from these studies verify the in vivo immunoeffectiveness of hMSCs in rodents and support the potential therapeutic use of hMSCs for the treatment of airway inflammation associated with chronic asthma.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Dept. of Pediatrics, Case Western Reserve Univ., Cleveland, OH 44106-4948, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Huang H, Chen L, Sanberg P. Cell Therapy From Bench to Bedside Translation in CNS Neurorestoratology Era. CELL MEDICINE 2010; 1:15-46. [PMID: 21359168 DOI: 10.3727/215517910x516673] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent advances in cell biology, neural injury and repair, and the progress towards development of neurorestorative interventions are the basis for increased optimism. Based on the complexity of the processes of demyelination and remyelination, degeneration and regeneration, damage and repair, functional loss and recovery, it would be expected that effective therapeutic approaches will require a combination of strategies encompassing neuroplasticity, immunomodulation, neuroprotection, neurorepair, neuroreplacement, and neuromodulation. Cell-based restorative treatment has become a new trend, and increasing data worldwide have strongly proven that it has a pivotal therapeutic value in CNS disease. Moreover, functional neurorestoration has been achieved to a certain extent in the CNS clinically. Up to now, the cells successfully used in preclinical experiments and/or clinical trial/treatment include fetal/embryonic brain and spinal cord tissue, stem cells (embryonic stem cells, neural stem/progenitor cells, hematopoietic stem cells, adipose-derived adult stem/precursor cells, skin-derived precursor, induced pluripotent stem cells), glial cells (Schwann cells, oligodendrocyte, olfactory ensheathing cells, astrocytes, microglia, tanycytes), neuronal cells (various phenotypic neurons and Purkinje cells), mesenchymal stromal cells originating from bone marrow, umbilical cord, and umbilical cord blood, epithelial cells derived from the layer of retina and amnion, menstrual blood-derived stem cells, Sertoli cells, and active macrophages, etc. Proof-of-concept indicates that we have now entered a new era in neurorestoratology.
Collapse
Affiliation(s)
- Hongyun Huang
- Center for Neurorestoratology, Beijing Rehabilitation Center, Beijing, P.R. China
| | | | | |
Collapse
|
12
|
Bai L, Lennon DP, Eaton V, Maier K, Caplan AI, Miller SD, Miller RH. Human bone marrow-derived mesenchymal stem cells induce Th2-polarized immune response and promote endogenous repair in animal models of multiple sclerosis. Glia 2009; 57:1192-203. [PMID: 19191336 DOI: 10.1002/glia.20841] [Citation(s) in RCA: 393] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cell-based therapies are attractive approaches to promote myelin repair. Recent studies demonstrated a reduction in disease burden in mice with experimental allergic encephalomyelitis (EAE) treated with mouse mesenchymal stem cells (MSCs). Here, we demonstrated human bone marrow-derived MSCs (BM-hMSCs) promote functional recovery in both chronic and relapsing-remitting models of mouse EAE, traced their migration into the injured CNS and assayed their ability to modulate disease progression and the host immune response. Injected BM-hMSCs accumulated in the CNS, reduced the extent of damage and increased oligodendrocyte lineage cells in lesion areas. The increase in oligodendrocytes in lesions may reflect BM-hMSC-induced changes in neural fate determination, since neurospheres from treated animals gave rise to more oligodendrocytes and less astrocytes than nontreated neurospheres. Host immune responses were also influenced by BM-hMSCs. Inflammatory T-cells including interferon gamma producing Th1 cells and IL-17 producing Th17 inflammatory cells and their associated cytokines were reduced along with concomitant increases in IL-4 producing Th2 cells and anti-inflammatory cytokines. Together, these data suggest that the BM-hMSCs represent a viable option for therapeutic approaches.
Collapse
Affiliation(s)
- Lianhua Bai
- Case Western Reserve University, Centers for Stem Cells and Regenerative Medicine, Translational Neuroscience, Department of Neurosciences, Case School of Medicine, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Sher F, van Dam G, Boddeke E, Copray S. Bioluminescence Imaging of Olig2-Neural Stem Cells Reveals Improved Engraftment in a Demyelination Mouse Model. Stem Cells 2009; 27:1582-91. [DOI: 10.1002/stem.76] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Abstract
The etiology of multiple sclerosis (MS) is unknown but it manifests as a chronic inflammatory demyelinating disease in the central nervous system (CNS). During chronic CNS inflammation, nicotinamide adenine dinucleotide (NAD) concentrations are altered by (T helper) Th1-derived cytokines through the coordinated induction of both indoleamine 2,3-dioxygenase (IDO) and the ADP cyclase CD38 in pathogenic microglia and lymphocytes. While IDO activation may keep auto-reactive T cells in check, hyper-activation of IDO can leave neuronal CNS cells starving for extracellular sources of NAD. Existing data indicate that glia may serve critical functions as an essential supplier of NAD to neurons during times of stress. Administration of pharmacological doses of non-tryptophan NAD precursors ameliorates pathogenesis in animal models of MS. Animal models of MS involve artificially stimulated autoimmune attack of myelin by experimental autoimmune encephalomyelitis (EAE) or by viral-mediated demyelination using Thieler's murine encephalomyelitis virus (TMEV). The Wld(S) mouse dramatically resists razor axotomy mediated axonal degeneration. This resistance is due to increased efficiency of NAD biosynthesis that delays stress-induced depletion of axonal NAD and ATP. Although the Wld(S) genotype protects against EAE pathogenesis, TMEV-mediated pathogenesis is exacerbated. In this review, we contrast the role of NAD in EAE versus TMEV demyelinating pathogenesis to increase our understanding of the pharmacotherapeutic potential of NAD signal transduction pathways. We speculate on the importance of increased SIRT1 activity in both PARP-1 inhibition and the potentially integral role of neuronal CD200 interactions through glial CD200R with induction of IDO in MS pathogenesis. A comprehensive review of immunomodulatory control of NAD biosynthesis and degradation in MS pathogenesis is presented. Distinctive pharmacological approaches designed for NAD-complementation or targeting NAD-centric proteins (SIRT1, SIRT2, PARP-1, GPR109a, and CD38) are outlined towards determining which approach may work best in the context of clinical application.
Collapse
Affiliation(s)
- W Todd Penberthy
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, Ohio 45237, USA.
| | | |
Collapse
|
15
|
Cellular remyelinating therapy in multiple sclerosis. J Neurol Sci 2009; 276:1-5. [DOI: 10.1016/j.jns.2008.08.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 08/14/2008] [Accepted: 08/19/2008] [Indexed: 11/19/2022]
|
16
|
Abstract
Remyelination involves reinvesting demyelinated axons with new myelin sheaths. In stark contrast to the situation that follows loss of neurons or axonal damage, remyelination in the CNS can be a highly effective regenerative process. It is mediated by a population of precursor cells called oligodendrocyte precursor cells (OPCs), which are widely distributed throughout the adult CNS. However, despite its efficiency in experimental models and in some clinical diseases, remyelination is often inadequate in demyelinating diseases such as multiple sclerosis (MS), the most common demyelinating disease and a cause of neurological disability in young adults. The failure of remyelination has profound consequences for the health of axons, the progressive and irreversible loss of which accounts for the progressive nature of these diseases. The mechanisms of remyelination therefore provide critical clues for regeneration biologists that help them to determine why remyelination fails in MS and in other demyelinating diseases and how it might be enhanced therapeutically.
Collapse
|
17
|
Oligodendrocyte differentiation and implantation: new insights for remyelinating cell therapy. Curr Opin Neurol 2008; 21:607-14. [PMID: 18769257 DOI: 10.1097/wco.0b013e32830f1e50] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Recent research on oligodendrocyte development has yielded new insights on the involvement of morphogens and differentiation factors in oligodendrogenesis. This knowledge has improved strategies to control neural stem cell-derived oligodendrocyte differentiation and functional maturation in vitro. In this review, we highlight the current knowledge on oligodendrocyte differentiation and discuss the novel possibilities of neural stem cell-derived oligodendrocytes for graft-based remyelination therapy, for example, for multiple sclerosis. RECENT FINDINGS Detailed insight into the cellular and molecular processes of embryonic and adult oligodendrogenesis has extended considerably in the past 2 years. Application of extrinsic factors and manipulation of intrinsic factors in neural stem cells have yielded convincing oligodendrocyte differentiation strategies. In addition, the recent groundbreaking developments regarding induced pluripotent stem cells generated from easily accessible somatic cells seem to offer an almost inexhaustible source for transplantable, autologous neural stem cells. Moreover, new approaches to optimize the implantation site for oligodendrocyte survival and functionality have improved the feasibility of stem cell-based oligodendrocyte replacement therapy. SUMMARY Loss of myelin in demyelinating diseases is only partly restored by endogenous oligodendrocyte precursor cells. Application of optimally functional, neural stem cell-derived oligodendrocyte precursors at the lesion site has become a realistic therapeutic approach to promote the remyelination process.
Collapse
|
18
|
Immune effects of mesenchymal stem cells: Implications for Charcot–Marie–Tooth disease. Cell Immunol 2008; 253:11-5. [DOI: 10.1016/j.cellimm.2008.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Revised: 05/28/2008] [Accepted: 06/04/2008] [Indexed: 12/12/2022]
|