1
|
Xu Z, He S, Begum MM, Han X. Myelin Lipid Alterations in Neurodegenerative Diseases: Landscape and Pathogenic Implications. Antioxid Redox Signal 2024; 41:1073-1099. [PMID: 39575748 PMCID: PMC11971557 DOI: 10.1089/ars.2024.0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024]
Abstract
Significance: Lipids, which constitute the highest portion (over 50%) of brain dry mass, are crucial for brain integrity, energy homeostasis, and signaling regulation. Emerging evidence revealed that lipid profile alterations and abnormal lipid metabolism occur during normal aging and in different forms of neurodegenerative diseases. Moreover, increasing genome-wide association studies have validated new targets on lipid-associated pathways involved in disease development. Myelin, the protective sheath surrounding axons, is crucial for efficient neural signaling transduction. As the primary site enriched with lipids, impairments of myelin are increasingly recognized as playing significant and complex roles in various neurodegenerative diseases, beyond simply being secondary effects of neuronal loss. Recent Advances: With advances in the lipidomics field, myelin lipid alterations and their roles in contributing to or reflecting the progression of diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and others, have recently caught great attention. Critical Issues: This review summarizes recent findings of myelin lipid alterations in the five most common neurodegenerative diseases and discusses their implications in disease pathogenesis. Future Directions: By highlighting myelin lipid abnormalities in neurodegenerative diseases, this review aims to encourage further research focused on lipids and the development of new lipid-oriented therapeutic approaches in this area. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Mst Marium Begum
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
- Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
2
|
Rogujski P, Lukomska B, Janowski M, Stanaszek L. Glial-restricted progenitor cells: a cure for diseased brain? Biol Res 2024; 57:8. [PMID: 38475854 DOI: 10.1186/s40659-024-00486-1] [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: 03/03/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The central nervous system (CNS) is home to neuronal and glial cells. Traditionally, glia was disregarded as just the structural support across the brain and spinal cord, in striking contrast to neurons, always considered critical players in CNS functioning. In modern times this outdated dogma is continuously repelled by new evidence unravelling the importance of glia in neuronal maintenance and function. Therefore, glia replacement has been considered a potentially powerful therapeutic strategy. Glial progenitors are at the center of this hope, as they are the source of new glial cells. Indeed, sophisticated experimental therapies and exciting clinical trials shed light on the utility of exogenous glia in disease treatment. Therefore, this review article will elaborate on glial-restricted progenitor cells (GRPs), their origin and characteristics, available sources, and adaptation to current therapeutic approaches aimed at various CNS diseases, with particular attention paid to myelin-related disorders with a focus on recent progress and emerging concepts. The landscape of GRP clinical applications is also comprehensively presented, and future perspectives on promising, GRP-based therapeutic strategies for brain and spinal cord diseases are described in detail.
Collapse
Affiliation(s)
- Piotr Rogujski
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| |
Collapse
|
3
|
Franklin RJM, Bodini B, Goldman SA. Remyelination in the Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041371. [PMID: 38316552 PMCID: PMC10910446 DOI: 10.1101/cshperspect.a041371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
4
|
Goldman SA, Franklin RJM, Osorio J. Stem and progenitor cell-based therapy of myelin disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:283-295. [PMID: 39341659 DOI: 10.1016/b978-0-323-90120-8.00015-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Much of clinical neurology is concerned with diseases of-or involving-the brain's subcortical white matter. Common to these disorders is the loss of myelin, reflecting the elimination or dysfunction of oligodendrocytes and fibrous astrocytes. As such, the introduction of glial progenitor cells, which can give rise to new oligodendrocytes and astrocytes alike, may be a feasible strategy for treating a broad variety of conditions in which white matter loss is causally involved. This review first covers the sourcing and production of human glial progenitor cells, and the preclinical evidence for their efficacy in achieving myelin restoration in vivo. It then discusses both pediatric and adult disease targets for which transplanted glial progenitors may prove of therapeutic value, those challenges that remain in the clinical application of a glial cell replacement strategy, and the clinical endpoints by which the efficacy of this approach may be assessed.
Collapse
Affiliation(s)
- Steven A Goldman
- Sana Biotechnology, Cambridge, MA, United States; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, United States; University of Copenhagen Faculty of Medicine, Copenhagen, Denmark.
| | | | - Joana Osorio
- Sana Biotechnology, Cambridge, MA, United States; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
5
|
Wang X, Zang J, Yang Y, Lu S, Guan Q, Ye D, Wang Z, Zhou H, Li K, Wang Q, Wu Y, Luan Z. Transplanted Human Oligodendrocyte Progenitor Cells Restore Neurobehavioral Deficits in a Rat Model of Preterm White Matter Injury. Front Neurol 2021; 12:749244. [PMID: 34858313 PMCID: PMC8631304 DOI: 10.3389/fneur.2021.749244] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Preterm white matter injury (PWMI) is a common brain injury and a leading cause of life-long neurological deficits in premature infants; however, no effective treatment is available yet. This study aimed to investigate the fate and effectiveness of transplanted human oligodendrocyte progenitor cells (hOPCs) in a rat model of PWMI. Methods: Hypoxia-ischemia was induced in rats at postnatal day 3, and hOPCs (6 × 105 cells/5 μL) were intracerebroventricularly transplanted at postnatal day 7. Neurobehavior was assessed 12 weeks post-transplant using the CatWalk test and Morris water maze test. Histological analyses, as well as immunohistochemical and transmission electron microscopy, were performed after transcardial perfusion. Results: Transplanted hOPCs survived for 13 weeks in PWMI brains. They were widely distributed in the injured white matter, and migrated along the corpus callosum to the contralateral hemisphere. Notably, 82.77 ± 3.27% of transplanted cells differentiated into mature oligodendrocytes, which produced myelin around the axons. Transplantation of hOPCs increased the fluorescence intensity of myelin basic protein and the thickness of myelin sheaths as observed in immunostaining and transmission electron microscopy, while it reduced white matter atrophy at the level of gross morphology. With regard to neurobehavior, the CatWalk test revealed improved locomotor function and inter-paw coordination after transplantation, and the cognitive functions of hOPC-transplanted rats were restored as revealed by the Morris water maze test. Conclusions: Myelin restoration through the transplantation of hOPCs led to neurobehavioral improvements in PWMI rats, suggesting that transplanting hOPCs may provide an effective and promising therapeutic strategy in children with PWMI.
Collapse
Affiliation(s)
- Xiaohua Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China.,Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Zang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yinxiang Yang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Siliang Lu
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Guan
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dou Ye
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhaoyan Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Haipeng Zhou
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Ke Li
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Zuo Luan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Edelmann MJ, Maegawa GHB. CNS-Targeting Therapies for Lysosomal Storage Diseases: Current Advances and Challenges. Front Mol Biosci 2020; 7:559804. [PMID: 33304924 PMCID: PMC7693645 DOI: 10.3389/fmolb.2020.559804] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022] Open
Abstract
During the past decades, several therapeutic approaches have been developed and made rapidly available for many patients afflicted with lysosomal storage disorders (LSDs), inborn organelle disorders with broad clinical manifestations secondary to the progressive accumulation of undegraded macromolecules within lysosomes. These conditions are individually rare, but, collectively, their incidence ranges from 1 in 2,315 to 7,700 live-births. Most LSDs are manifested by neurological symptoms or signs, including developmental delay, seizures, acroparesthesia, motor weakness, and extrapyramidal signs. The chronic and later-onset clinical forms are at one end of the continuum spectrum and are characterized by a subtle and slow progression of neurological symptoms. Due to its inherent physiological properties, unfortunately, the blood-brain barrier (BBB) constitutes a significant obstacle for current and upcoming therapies to achieve the central nervous system (CNS) and treat neurological problems so prevalent in these conditions. To circumvent this limitation, several strategies have been developed to make the therapeutic agent achieve the CNS. This narrative will provide an overview of current therapeutic strategies under development to permeate the BBB, and address and unmet need for treatment of the progressive neurological manifestations, which are so prevalent in these inherited lysosomal disorders.
Collapse
Affiliation(s)
- Mariola J Edelmann
- Department of Microbiology and Cell Science, The University of Florida's Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Gustavo H B Maegawa
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
Favret JM, Weinstock NI, Feltri ML, Shin D. Pre-clinical Mouse Models of Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:57. [PMID: 32351971 PMCID: PMC7174556 DOI: 10.3389/fmolb.2020.00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
There are over 50 lysosomal hydrolase deficiencies, many of which cause neurodegeneration, cognitive decline and death. In recent years, a number of broad innovative therapies have been proposed and investigated for lysosomal storage diseases (LSDs), such as enzyme replacement, substrate reduction, pharmacologic chaperones, stem cell transplantation, and various forms of gene therapy. Murine models that accurately reflect the phenotypes observed in human LSDs are critical for the development, assessment and implementation of novel translational therapies. The goal of this review is to summarize the neurodegenerative murine LSD models available that recapitulate human disease, and the pre-clinical studies previously conducted. We also describe some limitations and difficulties in working with mouse models of neurodegenerative LSDs.
Collapse
Affiliation(s)
| | | | | | - Daesung Shin
- Hunter James Kelly Research Institute, Department of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
8
|
Yue Y, Zhang L, Qu Y, Mu DZ. [Neuroprotective effects of oligodendrocyte precursor cells on white matter damage in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:326-331. [PMID: 29658460 PMCID: PMC7390025 DOI: 10.7499/j.issn.1008-8830.2018.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/27/2018] [Indexed: 06/08/2023]
Abstract
White matter damage, characterized by demyelination due to the damage of oligodendrocyte precursor cells (OPCs), is the most common type of brain damage in preterm infants. Survivors are often subject to long-term neurodevelopmental sequelae because of the lack of effective treatment. In recent years, it has been found that cell transplantation has the potential for the treatment of white matter damage. OPCs are frequently used cells in cell transplantation therapy. With abilities of migration and myelinization, OPCs are the best seed cells for the treatment of white matter damage. Several studies have found that OPCs may not only replace impaired cells to reconstruct the structure and function of white matter, but also inhibit neuronal apoptosis, promote the proliferation of endogenous neural stem cells, and enhance the repairment of the blood-brain barrier. However, the clinical application of OPC transplantation therapy faces many challenges, such as the effectiveness, risk of tumorigenesis and immune rejection. With reference to these studies, this article reviewed the development of myelination, the obtainment of OPCs, the therapeutic mechanism as well as application research, and analyzed the current challenges of OPC transplantation, in order to provide a new direction for clinical treatment of white matter damage in preterm infants.
Collapse
Affiliation(s)
- Yan Yue
- Department of Pediatrics, West China Second University Hospital/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China.
| | | | | | | |
Collapse
|
9
|
Wu CJ, Wang ZY, Yang YX, Luan Z. [Long-term effect of oligodendrocyte precursor cell transplantation on a rat model of white matter injury in the preterm infant]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:1003-1007. [PMID: 28899472 PMCID: PMC7403059 DOI: 10.7499/j.issn.1008-8830.2017.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 08/02/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the long-term effect of oligodendrocyte precursor cell (OPC) transplantation on a rat model of white matter injury (WMI) in the preterm infant. METHODS A total of 80 Sprague-Dawley rats aged 3 days were randomly divided into sham-operation group, model control group, 5-day ventricular/white matter transplantation group, 9-day ventricular/white matter transplantation group, 14-day ventricular/white matter transplantation group (n=10 each). All groups except the sham-operation group were treated with right common carotid artery ligation and hypoxia for 80 minutes to establish a rat model of WMI in the preterm infant. OPCs were prepared from the human fetal brain tissue (10-12 gestational weeks). At 5, 9, and 14 days after modeling, 3×105 OPCs were injected into the right lateral ventricle or white matter in each transplantation group, and myelin sheath and neurological function were evaluated under an electron microscope at ages of 60 and 90 days. RESULTS Electron microscopy showed that at an age of 60 days, each transplantation group had a slight improvement in myelin sheath injury compared with the model control group; at an age of 90 days, each transplantation group had significantly thickened myelin sheath and reduced structural damage compared with the model control group, and the 14-day transplantation groups had the most significant changes. There were no significant differences in the degree of myelin sheath injury between the ventricular and white matter transplantation groups at different time points. At an age of 60 or 90 days, the transplantation groups had a significantly higher modified neurological severity score (mNSS) than the sham-operation group and a significantly lower mNSS than the model control group (P<0.05). CONCLUSIONS OPC transplantation may have a long-term effect in the treatment of WMI in the preterm infant, and delayed transplantation may enhance its therapeutic effect.
Collapse
Affiliation(s)
- Cheng-Jun Wu
- Third Clinical Medical College, Southern Medical University, Guangzhou 510515, China.
| | | | | | | |
Collapse
|
10
|
Abstract
Diseases of glia, including astrocytes and oligodendrocytes, are among the most prevalent and disabling, yet least appreciated, conditions in neurology. In recent years, it has become clear that besides the overtly glial disorders of oligodendrocyte loss and myelin failure, such as the leukodystrophies and inflammatory demyelinations, a number of neurodegenerative and psychiatric disorders may also be causally linked to glial dysfunction and derive from astrocytic as well as oligodendrocytic pathology. The relative contribution of glial dysfunction to many of these disorders may be so great as to allow their treatment by the delivery of allogeneic glial progenitor cells, the precursors to both astroglia and myelin-producing oligodendrocytes. Given the development of new methods for producing and isolating these cells from pluripotent stem cells, both the myelin disorders and appropriate glial-based neurodegenerative conditions may now be compelling targets for cell-based therapy. As such, glial cell-based therapies may offer potential benefit to a broader range of diseases than ever before contemplated, including disorders such as Huntington's disease and the motor neuron degeneration of amyotrophic lateral sclerosis, which have traditionally been considered neuronal in nature.
Collapse
|
11
|
Osorio MJ, Goldman SA. Glial progenitor cell-based treatment of the childhood leukodystrophies. Exp Neurol 2016; 283:476-88. [PMID: 27170209 DOI: 10.1016/j.expneurol.2016.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 04/19/2016] [Accepted: 05/05/2016] [Indexed: 12/19/2022]
Abstract
The childhood leukodystrophies comprise a group of hereditary disorders characterized by the absence, malformation or destruction of myelin. These disorders share common clinical, radiological and pathological features, despite their diverse molecular and genetic etiologies. Oligodendrocytes and astrocytes are the major affected cell populations, and are either structurally impaired or metabolically compromised through cell-intrinsic pathology, or are the victims of mis-accumulated toxic byproducts of metabolic derangement. In either case, glial cell replacement using implanted tissue or pluripotent stem cell-derived human neural or glial progenitor cells may comprise a promising strategy for both structural remyelination and metabolic rescue. A broad variety of pediatric white matter disorders, including the primary hypomyelinating disorders, the lysosomal storage disorders, and the broader group of non-lysosomal metabolic leukodystrophies, may all be appropriate candidates for glial progenitor cell-based treatment. Nonetheless, a variety of specific challenges remain before this therapeutic strategy can be applied to children. These include timely diagnosis, before irreparable neuronal injury has ensued; understanding the natural history of the targeted disease; defining the optimal cell phenotype for each disorder; achieving safe and scalable cellular compositions; designing age-appropriate controlled clinical trials; and for autologous therapy of genetic disorders, achieving the safe genetic editing of pluripotent stem cells. Yet these challenges notwithstanding, the promise of glial progenitor cell-based treatment of the childhood myelin disorders offers hope to the many victims of this otherwise largely untreatable class of disease.
Collapse
Affiliation(s)
- M Joana Osorio
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, United States; Center for Basic and Translational Neuroscience, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 2200, Denmark.
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, United States; Center for Basic and Translational Neuroscience, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 2200, Denmark.
| |
Collapse
|
12
|
Cell Therapy for Pediatric Disorders of Glia. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
13
|
Biffi A. Gene therapy for lysosomal storage disorders: a good start. Hum Mol Genet 2015; 25:R65-75. [PMID: 26604151 DOI: 10.1093/hmg/ddv457] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are a heterogeneous group of inherited diseases with a collective frequency of ∼1 in 7000 births, resulting from the deficiency in one or more enzymes or transporters that normally reside within the lysosomes. Pathology results from the progressive accumulation of uncleaved lipids, glycoproteins and/or glycosaminoglycans in the lysosomes and secondary damages that affect the brain, viscera, bones and connective tissues. Most treatment modalities developed for LSD, including gene therapy (GT), are based on the lysosome-specific cross-correction mechanism, by which close proximity of normal cells leads to the correction of the biochemical consequences of enzymatic deficiency within the neighboring cells. Here, GT efforts addressing these disorders are reviewed with an up-to-date discussion of their impact on the LSD disease phenotype in animal models and patients.
Collapse
Affiliation(s)
- Alessandra Biffi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
14
|
Arylsulfatase A Overexpressing Human iPSC-derived Neural Cells Reduce CNS Sulfatide Storage in a Mouse Model of Metachromatic Leukodystrophy. Mol Ther 2015; 23:1519-31. [PMID: 26061647 DOI: 10.1038/mt.2015.106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 05/31/2015] [Indexed: 12/16/2022] Open
Abstract
Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disorder resulting from a functional deficiency of arylsulfatase A (ARSA), an enzyme that catalyzes desulfation of 3-O-sulfogalactosylceramide (sulfatide). Lack of active ARSA leads to the accumulation of sulfatide in oligodendrocytes, Schwann cells and some neurons and triggers progressive demyelination, the neuropathological hallmark of MLD. Several therapeutic approaches have been explored, including enzyme replacement, autologous hematopoietic stem cell-based gene therapy, intracerebral gene therapy or cell-based gene delivery into the central nervous system (CNS). However, long-term treatment of the blood-brain-barrier protected CNS remains challenging. Here we used MLD patient-derived induced pluripotent stem cells (iPSCs) to generate long-term self-renewing neuroepithelial stem cells and astroglial progenitors for cell-based ARSA replacement. Following transplantation of ARSA-overexpressing precursors into ARSA-deficient mice we observed a significant reduction of sulfatide storage up to a distance of 300 µm from grafted cells. Our data indicate that neural precursors generated via reprogramming from MLD patients can be engineered to ameliorate sulfatide accumulation and may thus serve as autologous cell-based vehicle for continuous ARSA supply in MLD-affected brain tissue.
Collapse
|
15
|
Schwann cells induce Proliferation and Migration of Oligodendrocyte Precursor Cells Through Secretion of PDGF-AA and FGF-2. J Mol Neurosci 2015; 56:999-1008. [DOI: 10.1007/s12031-015-0570-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/17/2015] [Indexed: 11/26/2022]
|
16
|
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, although this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granular neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet, remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this review, we will review the biology of remyelination, including the cells and signals involved; describe when remyelination occurs and when and why it fails and the consequences of its failure; and discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642 University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
17
|
Xu L, Ryu J, Hiel H, Menon A, Aggarwal A, Rha E, Mahairaki V, Cummings BJ, Koliatsos VE. Transplantation of human oligodendrocyte progenitor cells in an animal model of diffuse traumatic axonal injury: survival and differentiation. Stem Cell Res Ther 2015; 6:93. [PMID: 25971252 PMCID: PMC4453242 DOI: 10.1186/s13287-015-0087-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/13/2015] [Accepted: 05/01/2015] [Indexed: 12/14/2022] Open
Abstract
Introduction Diffuse axonal injury is an extremely common type of traumatic brain injury encountered in motor vehicle crashes, sports injuries, and in combat. Although many cases of diffuse axonal injury result in chronic disability, there are no current treatments for this condition. Its basic lesion, traumatic axonal injury, has been aggressively modeled in primate and rodent animal models. The inexorable axonal and perikaryal degeneration and dysmyelination often encountered in traumatic axonal injury calls for regenerative therapies, including therapies based on stem cells and precursors. Here we explore the proof of concept that treatments based on transplants of human oligodendrocyte progenitor cells can replace or remodel myelin and, eventually, contribute to axonal regeneration in traumatic axonal injury. Methods We derived human oligodendrocyte progenitor cells from the human embryonic stem cell line H9, purified and characterized them. We then transplanted these human oligodendrocyte progenitor cells into the deep sensorimotor cortex next to the corpus callosum of nude rats subjected to traumatic axonal injury based on the impact acceleration model of Marmarou. We explored the time course and spatial distribution of differentiation and structural integration of these cells in rat forebrain. Results At the time of transplantation, over 90 % of human oligodendrocyte progenitor cells expressed A2B5, PDGFR, NG2, O4, Olig2 and Sox10, a profile consistent with their progenitor or early oligodendrocyte status. After transplantation, these cells survived well and migrated massively via the corpus callosum in both injured and uninjured brains. Human oligodendrocyte progenitor cells displayed a striking preference for white matter tracts and were contained almost exclusively in the corpus callosum and external capsule, the striatopallidal striae, and cortical layer 6. Over 3 months, human oligodendrocyte progenitor cells progressively matured into myelin basic protein(+) and adenomatous polyposis coli protein(+) oligodendrocytes. The injured environment in the corpus callosum of impact acceleration subjects tended to favor maturation of human oligodendrocyte progenitor cells. Electron microscopy revealed that mature transplant-derived oligodendrocytes ensheathed host axons with spiral wraps intimately associated with myelin sheaths. Conclusions Our findings suggest that, instead of differentiating locally, human oligodendrocyte progenitor cells migrate massively along white matter tracts and differentiate extensively into ensheathing oligodendrocytes. These features make them appealing candidates for cellular therapies of diffuse axonal injury aiming at myelin remodeling and axonal protection or regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0087-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leyan Xu
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Jiwon Ryu
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Hakim Hiel
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Adarsh Menon
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Ayushi Aggarwal
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Elizabeth Rha
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Vasiliki Mahairaki
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Brian J Cummings
- Departments of Physical and Medical Rehabilitation, Neurological Surgery, and Anatomy and Neurobiology, Sue and Bill Gross Stem Cell Research Center, Institute for Memory Impairments and Neurological Disorders, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Vassilis E Koliatsos
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Pituch KC, Moyano AL, Lopez-Rosas A, Marottoli FM, Li G, Hu C, van Breemen R, Månsson JE, Givogri MI. Dysfunction of platelet-derived growth factor receptor α (PDGFRα) represses the production of oligodendrocytes from arylsulfatase A-deficient multipotential neural precursor cells. J Biol Chem 2015; 290:7040-53. [PMID: 25605750 DOI: 10.1074/jbc.m115.636498] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The membrane-bound receptor for platelet-derived growth factor A (PDGFRα) is crucial for controlling the production of oligodendrocytes (OLs) for myelination, but regulation of its activity during OL differentiation is largely unknown. We have examined the effect of increased sulfated content of galactosylceramides (sulfatides) on the regulation of PDGFRα in multipotential neural precursors (NPs) that are deficient in arylsulfatase A (ASA) activity. This enzyme is responsible for the lysosomal hydrolysis of sulfatides. We show that sulfatide accumulation significantly impacts the formation of OLs via deregulation of PDGFRα function. PDGFRα is less associated with detergent-resistant membranes in ASA-deficient cells and showed a significant decrease in AKT phosphorylation. Rescue experiments with ASA showed a normalization of the ratio of long versus short sulfatides, restored PDGFRα levels, corrected its localization to detergent-resistant membranes, increased AKT phosphorylation, and normalized the production of OLs in ASA-deficient NPs. Moreover, our studies identified a novel mechanism that regulates the secretion of PDGFRα in NPs, in glial cells, and in the brain cortex via exosomal shedding. Our study provides a first step in understanding the role of sulfatides in regulating PDGFRα levels in OLs and its impact in myelination.
Collapse
Affiliation(s)
- Katarzyna C Pituch
- From the Department of Anatomy and Cell Biology, College of Medicine, and
| | - Ana L Moyano
- From the Department of Anatomy and Cell Biology, College of Medicine, and
| | - Aurora Lopez-Rosas
- From the Department of Anatomy and Cell Biology, College of Medicine, and
| | | | - Guannan Li
- the Department of Medical Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago, Illinois 60612 and
| | - Chenqi Hu
- the Department of Medical Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago, Illinois 60612 and
| | - Richard van Breemen
- the Department of Medical Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago, Illinois 60612 and
| | - Jan E Månsson
- the Department of Clinical Chemistry, Sahlgren Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Maria I Givogri
- From the Department of Anatomy and Cell Biology, College of Medicine, and
| |
Collapse
|
19
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 11/21/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin - the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a 'multifunctional nanoplatform' that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive 'magnetic cell targeting' to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- />School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
20
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053 DOI: 10.1186/2052-8426-2-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin – the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a ‘multifunctional nanoplatform’ that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive ‘magnetic cell targeting’ to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
21
|
Sypecka J, Sarnowska A. The neuroprotective effect exerted by oligodendroglial progenitors on ischemically impaired hippocampal cells. Mol Neurobiol 2013; 49:685-701. [PMID: 24085562 PMCID: PMC3950613 DOI: 10.1007/s12035-013-8549-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/27/2013] [Indexed: 01/13/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) are the focus of intense research for the purpose of cell replacement therapies in acquired or inherited neurodegenerative disorders, accompanied by ongoing hypo/demyelination. Recently, it has been postulated that these glia-committed cells exhibit certain properties of neural stem cells. Advances in stem cell biology have shown that their therapeutic effect could be attributed to their ability to secret numerous active compounds which modify the local microenvironment making it more susceptible to restorative processes. To verify this hypothesis, we set up an ex vivo co-culture system of OPCs isolated from neonatal rat brain with organotypic hippocampal slices (OHC) injured by oxygen-glucose deprivation (OGD). The presence of OPCs in such co-cultures resulted in a significant neuroprotective effect manifesting itself as a decrease in cell death rate and as an extension of newly formed cells in ischemically impaired hippocampal slices. A microarray analysis of broad spectrum of trophic factors and cytokines expressed by OPCs was performed for the purpose of finding the factor(s) contributing to the observed effect. Three of them—BDNF, IL-10 and SCF—were selected for the subsequent functional assays. Our data revealed that BDNF released by OPCs is the potent factor that stimulates cell proliferation and survival in OHC subjected to OGD injury. At the same time, it was observed that IL-10 attenuates inflammatory processes by promoting the formation of the cells associated with the immunological response. Those neuroprotective qualities of oligodendroglia-biased progenitors significantly contribute to anticipating a successful cell replacement therapy.
Collapse
Affiliation(s)
- Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, Pawinskiego str.,, 02-106, Warsaw, Poland,
| | | |
Collapse
|
22
|
Patil SA, Maegawa GHB. Developing therapeutic approaches for metachromatic leukodystrophy. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:729-45. [PMID: 23966770 PMCID: PMC3743609 DOI: 10.2147/dddt.s15467] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal disorder caused by the deficiency of arylsulfatase A (ASA), resulting in impaired degradation of sulfatide, an essential sphingolipid of myelin. The clinical manifestations of MLD are characterized by progressive demyelination and subsequent neurological symptoms resulting in severe debilitation. The availability of therapeutic options for treating MLD is limited but expanding with a number of early stage clinical trials already in progress. In the development of therapeutic approaches for MLD, scientists have been facing a number of challenges including blood–brain barrier (BBB) penetration, safety issues concerning therapies targeting the central nervous system, uncertainty regarding the ideal timing for intervention in the disease course, and the lack of more in-depth understanding of the molecular pathogenesis of MLD. Here, we discuss the current status of the different approaches to developing therapies for MLD. Hematopoietic stem cell transplantation has been used to treat MLD patients, utilizing both umbilical cord blood and bone marrow sources. Intrathecal enzyme replacement therapy and gene therapies, administered locally into the brain or by generating genetically modified hematopoietic stem cells, are emerging as novel strategies. In pre-clinical studies, different cell delivery systems including microencapsulated cells or selectively neural cells have shown encouraging results. Small molecules that are more likely to cross the BBB can be used as enzyme enhancers of diverse ASA mutants, either as pharmacological chaperones, or proteostasis regulators. Specific small molecules may also be used to reduce the biosynthesis of sulfatides, or target different affected downstream pathways secondary to the primary ASA deficiency. Given the progressive neurodegenerative aspects of MLD, also seen in other lysosomal diseases, current and future therapeutic strategies will be complementary, whether used in combination or separately at specific stages of the disease course, to produce better outcomes for patients afflicted with this devastating inherited disorder.
Collapse
Affiliation(s)
- Shilpa A Patil
- McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
23
|
Goldman SA, Nedergaard M, Windrem MS. Glial progenitor cell-based treatment and modeling of neurological disease. Science 2012; 338:491-5. [PMID: 23112326 PMCID: PMC3548656 DOI: 10.1126/science.1218071] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The diseases of myelin are among the most prevalent and disabling conditions in neurology. These diseases include both the vascular and inflammatory demyelinating disorders of adulthood, as well as the childhood leukodystrophies and cerebral palsy. These fundamentally glial disorders may be amenable to treatment by glial progenitor cells (GPCs), which give rise to astroglia and myelin-producing oligodendrocytes. Given the development of new methods for generating and isolating human GPCs, the myelin disorders may now be compelling targets for cell-based therapy. In addition, the efficient engraftment and expansion of human GPCs in murine hosts has led to the development of human glial chimeric mouse brains, which provides new opportunities for studying the species-specific roles of human glia in cognition, as well as in disease pathogenesis.
Collapse
Affiliation(s)
- Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
24
|
Miranda CO, Brites P, Mendes Sousa M, Teixeira CA. Advances and pitfalls of cell therapy in metabolic leukodystrophies. Cell Transplant 2012; 22:189-204. [PMID: 23006656 DOI: 10.3727/096368912x656117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Leukodystrophies are a group of disorders characterized by myelin dysfunction, either at the level of myelin formation or maintenance, that affect the central nervous system (CNS) and also in some cases, to a lesser extent, the peripheral nervous system (PNS). Although these genetic-based disorders are generally rare, all together they have a significant impact in the society, with an estimated overall incidence of 1 in 7,663 live births. Currently, there is no cure for leukodystrophies, and the development of effective treatments remains challenging. Not only leukodystrophies generally progress very fast, but also most are multifocal needing the simultaneous targeting at multiple sites. Moreover, as the CNS is affected, the blood-brain barrier (BBB) limits the efficacy of treatment. Recently, interest on cell therapy has increased, and the leukodystrophies for which metabolic correction is needed have become first-choice candidates for cell-based clinical trials. In this review, we present and discuss the available cell transplantation therapies in metabolic leukodystrophies including fucosidosis, X-linked adrenoleukodystrophy, metachromatic leukodystrophy, Canavan disease, and Krabbe's disease. We will discuss the latest advances of cell therapy and its pitfalls in this group of disorders, taking into account, among others, the limitations imposed by reduced cell migration in multifocal conditions, the need to achieve corrective enzyme threshold levels, and the growing awareness that not only myelin but also the associated axonopathy needs to be targeted in some leukodystrophies.
Collapse
|
25
|
Batzios SP, Zafeiriou DI. Developing treatment options for metachromatic leukodystrophy. Mol Genet Metab 2012; 105:56-63. [PMID: 22078456 DOI: 10.1016/j.ymgme.2011.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/10/2011] [Accepted: 10/10/2011] [Indexed: 12/25/2022]
Abstract
Metachromatic leukodystrophy (MLD) represents a devastating lysosomal storage disease characterized by intralysosomal accumulation of the sphingolipid sulfatide in various tissues. Three types of the disease are currently distinguished: the late-infantile, which is the most commonly observed, the juvenile and the adult type. Demyelination represents the main histopathological feature of the disorder, leading to neurological impairment with no curative treatment currently available. Nevertheless, the increased scientific interest on the disease has led to the experimental use of innovative therapeutic approaches in animal models, aiming to provide an effective therapeutic regimen for human patients, as well. This paper provides an overview of developing treatment options among patients with MLD. Apart from hematopoietic stem cell transplantation, already in use for decades, other recent data discussed includes umbilical cord blood and stem cell transplantation, enzyme replacement therapy, gene therapy and autologous hematopoietic transplantation of genetically modified stem cells. Gene therapy with oligodedroglial, neural progenitor, embryonic and microencapsulated recombinant cells represents add-on treatment options still on experimental level.
Collapse
Affiliation(s)
- Spyros P Batzios
- 1st Department of Paediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
26
|
Martino G, Pluchino S, Bonfanti L, Schwartz M. Brain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cells. Physiol Rev 2011; 91:1281-304. [PMID: 22013212 PMCID: PMC3552310 DOI: 10.1152/physrev.00032.2010] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Regenerative processes occurring under physiological (maintenance) and pathological (reparative) conditions are a fundamental part of life and vary greatly among different species, individuals, and tissues. Physiological regeneration occurs naturally as a consequence of normal cell erosion, or as an inevitable outcome of any biological process aiming at the restoration of homeostasis. Reparative regeneration occurs as a consequence of tissue damage. Although the central nervous system (CNS) has been considered for years as a "perennial" tissue, it has recently become clear that both physiological and reparative regeneration occur also within the CNS to sustain tissue homeostasis and repair. Proliferation and differentiation of neural stem/progenitor cells (NPCs) residing within the healthy CNS, or surviving injury, are considered crucial in sustaining these processes. Thus a large number of experimental stem cell-based transplantation systems for CNS repair have recently been established. The results suggest that transplanted NPCs promote tissue repair not only via cell replacement but also through their local contribution to changes in the diseased tissue milieu. This review focuses on the remarkable plasticity of endogenous and exogenous (transplanted) NPCs in promoting repair. Special attention will be given to the cross-talk existing between NPCs and CNS-resident microglia as well as CNS-infiltrating immune cells from the circulation, as a crucial event sustaining NPC-mediated neuroprotection. Finally, we will propose the concept of the context-dependent potency of transplanted NPCs (therapeutic plasticity) to exert multiple therapeutic actions, such as cell replacement, neurotrophic support, and immunomodulation, in CNS repair.
Collapse
Affiliation(s)
- Gianvito Martino
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.
| | | | | | | |
Collapse
|
27
|
Crocker SJ, Bajpai R, Moore CS, Frausto RF, Brown GD, Pagarigan RR, Whitton JL, Terskikh AV. Intravenous administration of human embryonic stem cell-derived neural precursor cells attenuates cuprizone-induced central nervous system (CNS) demyelination. Neuropathol Appl Neurobiol 2011; 37:643-53. [PMID: 21276029 PMCID: PMC3252213 DOI: 10.1111/j.1365-2990.2011.01165.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Previous studies have demonstrated the therapeutic potential for human embryonic stem cell-derived neural precursor cells (hES-NPCs) in autoimmune and genetic animal models of demyelinating diseases. Herein, we tested whether intravenous (i.v.) administration of hES-NPCs would impact central nervous system (CNS) demyelination in a cuprizone model of demyelination. METHODS C57Bl/6 mice were fed cuprizone (0.2%) for 2 weeks and then separated into two groups that either received an i.v. injection of hES-NPCs or i.v. administration of media without these cells. After an additional 2 weeks of dietary cuprizone treatment, CNS tissues were analysed for detection of transplanted cells and differences in myelination in the region of the corpus callosum (CC). RESULTS Cuprizone-induced demyelination in the CC was significantly reduced in mice treated with hES-NPCs compared with cuprizone-treated controls that did not receive stem cells. hES-NPCs were identified within the brain tissues of treated mice and revealed migration of transplanted cells into the CNS. A limited number of human cells were found to express the mature oligodendrocyte marker, O1, or the astrocyte marker, glial fibrillary acidic protein. Reduced apoptosis and attenuated microglial and astrocytic responses were also observed in the CC of hES-NPC-treated mice. CONCLUSIONS These findings indicated that systemically administered hES-NPCs migrated from circulation into a demyelinated lesion within the CNS and effectively reduced demyelination. Observed reductions in astrocyte and microglial responses, and the benefit of hES-NPC treatment in this model of myelin injury was not obviously accountable to tissue replacement by exogenously administered cells.
Collapse
Affiliation(s)
- S J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The childhood leukodystrophies are characterized by neonatal or childhood deficiencies in myelin production or maintenance; these may be due to hereditary defects in genes for myelin maintenance, as in Pelizaeus-Merzbacher disease, or to enzymatic deficiencies resulting in substrate misaccumulation or misprocessing, as in the lysosomal storage disorders. Regardless of their respective etiologies, these disorders are essentially all manifested by a profound deterioration in neurological function with age. A congenital deficit in forebrain myelination is also noted in children with the periventricular leukomalacia of cerebral palsy, which yields a more static morbidity. In light of the wide range of disorders to which congenital hypomyelination or postnatal demyelination may contribute, and the relative homogeneity of oligodendrocytes and their progenitors, the leukodystrophies may be especially attractive targets for cell-based therapeutic strategies. As a result, glial progenitor cells, which can give rise to new myelinogenic oligodendrocytes, have become of great interest as potential vectors for the restoration of myelin to the dysmyelinated brain and spinal cord. In addition, by distributing throughout the neuraxis after perinatal graft, and giving rise to astrocytes as well as oligodendrocytes, glial progenitor cells may be of great utility in rectifying the dysmyelination-associated enzymatic deficiencies of the lysosomal storage disorders.
Collapse
Affiliation(s)
- Steven A Goldman
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
29
|
Tracy ET, Zhang CY, Gentry T, Shoulars KW, Kurtzberg J. Isolation and expansion of oligodendrocyte progenitor cells from cryopreserved human umbilical cord blood. Cytotherapy 2011; 13:722-9. [PMID: 21341973 PMCID: PMC3152318 DOI: 10.3109/14653249.2011.553592] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AIMS Oligodendrocyte precursor cells (OPC) hold promise as a cellular therapy for demyelinating diseases. The feasibility of using OPC-based therapies in humans depends upon a reliable, readily available source. We have previously described the isolation, expansion and characterization of oligodendrocyte-like cells from fresh human umbilical cord blood (UCB). We now describe the isolation and expansion of OPC from thawed, cryopreserved UCB. METHODS We thawed cryopreserved UCB units employing a standard clinical protocol, then isolated and plated mononuclear cells under previously established culture conditions. All OPC cultures were trypsinized at 21 days, counted, then characterized by flow cytometry after fixation, permeablization and labeling with the following antibodies: anti-oligodendrocyte marker 4 (O4), anti-oligodendrocyte marker 1 (O1) and anti-myelin basic protein (MBP). OPC were also placed in co-culture with shiverer mouse neuronal cells then stained in situ for beta tubulin III (BT3) and MBP as a functional assay of myelination. RESULTS The average OPC yield per cryopreserved UCB unit was 64% of that seen with fresh UCB. On flow cytometric analysis, 74% of thawed UCB units yielded cells with an O4-expression level of at least 20% of total events, compared with 95% of fresh UCB units. We observed myelination of shiverer neurons in our functional assay, which could be used as a potency assay for release of OPC cells in phase I human clinical trials. CONCLUSIONS Our results demonstrate that OPC can be derived reliably from thawed, cryopreserved UCB units, and support the feasibility of using these cells in human clinical trials.
Collapse
Affiliation(s)
- Elisabeth T Tracy
- Pediatric Blood and Marrow Transplant Program, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
30
|
White AB, Galbiati F, Givogri MI, Lopez Rosas A, Qiu X, van Breemen R, Bongarzone ER. Persistence of psychosine in brain lipid rafts is a limiting factor in the therapeutic recovery of a mouse model for Krabbe disease. J Neurosci Res 2011; 89:352-64. [PMID: 21259322 PMCID: PMC3064524 DOI: 10.1002/jnr.22564] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 10/18/2010] [Accepted: 10/30/2010] [Indexed: 11/10/2022]
Abstract
Sphingolipids are intrinsic components of membrane lipid rafts. The abnormal accumulation of these molecules may introduce architectural and functional changes in these domains, leading to cellular dysfunction. Galactosylsphingosine (psychosine) is a pathogenic lipid raft-associated molecule whose accumulation leads to brain deterioration and irreversible neurological handicap in the incurable leukodystrophy Krabbe disease (KD). The relevance of clearing excessive levels of pathogenic psychosine from lipid rafts in therapy for KD has not been investigated. The work presented here demonstrates that psychosine inhibits raft-mediated endocytosis in neural cells. In addition, although in vitro enzyme reconstitution is sufficient for the reversal of related endocytic defects in affected neural cells, traditional in vivo enzyme therapies in the mouse model of KD appear to be insufficient for complete removal of pathogenic levels of raft-associated psychosine. This work describes a mechanism that may contribute to limiting the in vivo efficacy of traditional therapies for KD.
Collapse
Affiliation(s)
- AB White
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| | - F Galbiati
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| | - MI Givogri
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| | - A Lopez Rosas
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| | - X Qiu
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois, Chicago, Illinois
| | - R van Breemen
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois, Chicago, Illinois
| | - ER Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois
| |
Collapse
|
31
|
Pathology and current treatment of neurodegenerative sphingolipidoses. Neuromolecular Med 2010; 12:362-82. [PMID: 20730629 DOI: 10.1007/s12017-010-8133-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 08/10/2010] [Indexed: 01/09/2023]
Abstract
Sphingolipidoses constitute a large subgroup of lysosomal storage disorders (LSDs). Many of them are associated with a progressive neurodegeneration. As is the case for LSDs in general, most sphingolipidoses are caused by deficiencies in lysosomal hydrolases. However, accumulation of sphingolipids can also result from deficiencies in proteins involved in the transport or posttranslational modification of lysosomal enzymes, transport of lipids, or lysosomal membrane proteins required for transport of lysosomal degradation end products. The accumulation of sphingolipids in the lysosome together with secondary changes in the concentration and localization of other lipids may cause trafficking defects of membrane lipids and proteins, affect calcium homeostasis, induce the unfolded protein response, activate apoptotic cascades, and affect various signal transduction pathways. To what extent, however, these changes contribute to the pathogenesis of the diseases is not fully understood. Currently, there is no cure for sphingolipidoses. Therapies like enzyme replacement, pharmacological chaperone, and substrate reduction therapy, which have been shown to be efficient in non-neuronopathic LSDs, are currently evaluated in clinical trials of neuronopathic sphingolipidoses. In the future, neural stem cell therapy and gene therapy may become an option for these disorders.
Collapse
|
32
|
Zhao G, Karageorgos L, Hutchinson RG, Hopwood JJ, Hemsley K. Genetic manipulation of murine embryonic stem cells with enhanced green fluorescence protein and sulfatase-modifying factor I genes. Cytotherapy 2010; 12:400-7. [DOI: 10.3109/14653241003695026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
33
|
Munoz-Sanjuan I. Glial progenitor cell transplantation and the generation of chimeric animal models with human brain cells: implications for novel therapeutics. Expert Opin Ther Pat 2009; 19:1639-46. [PMID: 19939186 DOI: 10.1517/13543770903443105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The potential of exogenous stem cell or progenitor cell transplantation as a novel therapeutic strategy to address unmet medical needs is a vast and important area of investigation. A recent US patent has been issued to Goldman from the University of Rochester based on pioneering studies with human fetal and adult-derived glial progenitor cells (GPCs), covering the generation of chimeric mouse/human animals. OBJECTIVE/METHOD In this patent and associated manuscript, extensive chimerism due to grafting of human GPCs is associated with remyelination and functional rescue of mice congenitally deficient in oligodendrocyte survival and myelination, due to a deletion in the myelin basic protein gene (the shiverer mouse). This review highlights the implications of generating human/mouse chimeric animals for the study of human brain physiology, preclinical studies and the clinical application of progenitor cells towards the development of novel therapeutics for the treatment of demyelinating disorders. CONCLUSION The use of GPCs offers promise for remyelination disorders, and the ability of these cells to repopulate the entire rodent nervous system should allow for the investigation of the physiological properties of human glial derivatives in an in vivo context, enhancing the understanding of mechanisms with a primary effect through the modulation of human glial cell biology.
Collapse
|
34
|
Sawada T, Tanaka A, Higaki K, Takamura A, Nanba E, Seto T, Maeda M, Yamaguchi E, Matsuda J, Yamano T. Intracerebral cell transplantation therapy for murine GM1 gangliosidosis. Brain Dev 2009; 31:717-24. [PMID: 19118961 DOI: 10.1016/j.braindev.2008.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 10/15/2008] [Accepted: 11/01/2008] [Indexed: 11/25/2022]
Abstract
We performed a cell transplantation study to treat the brain involvement in lysosomal storage diseases. We used acid beta-galactosidase knock-out mice (BKO) from C57BL/6 as recipients. To minimize immune responses, we used cells derived from transgenic mice of C57BL/6 overexpressing the normal human beta-galactosidase. Fetal brain cells (FBC), bone marrow-derived mesenchymal stem cells (MSC), and mixed FBC and MSC cells were prepared and injected into the ventricle of newborn BKO mouse brain. The mice were examined at 1, 2, 4, and 8 weeks and 6 months after injection. In each experiment, the injected cells migrated into the whole brain effectively and survived for at least 8 weeks. Decrease in ganglioside GM1 level was also observed. FBC could survive for 6 months in recipient brain. However, the number of transplanted FBC decreased. In the brains of MSC- or mixed cell-treated mice, no grafted cells could be found at 6 months. To achieve sufficient long-term effects on the brain, a method of steering the immune response away from cytotoxic responses or of inducing tolerance to the products of therapeutic genes must be developed.
Collapse
Affiliation(s)
- Tomo Sawada
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sypecka J, Sarnowska A, Domanska-Janik K. Crucial role of the local micro-environment in fate decision of neonatal rat NG2 progenitors. Cell Prolif 2009; 42:661-71. [PMID: 19614677 DOI: 10.1111/j.1365-2184.2009.00618.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES The fate choice of neural progenitor cells could be dictated by local cellular environment of the adult CNS. The aim of our study was to investigate the effect of hippocampal tissue on differentiation and maturation of oligodendrocyte NG2 precursor cells. MATERIALS AND METHODS Hippocampal slice culture was established from the brains of 7-day-old rats. NG2 precursor cells, obtained from a 12-day-old mixed primary culture of neonatal rat cerebral hemispheres, were labelled with chloromethyl-fluorescein-diacetete and seeded on the hippocampal slices. After 7-14 days in co-culture, cells were stained with neural markers. RESULTS NG2 cells differentiated predominantly into oligodendrocytes, presenting various stages of maturation: progenitors (NG2), pre-oligodendrocytes (O4) and finally mature GalC-positive cells. However, except for a few cells with astrocyte-specific S100b staining, a considerable number of these cells differentiated into neurons: TUJ(+) and even MAP-2(+) cells were frequently observed. Moreover, a certain population of these cells preserved proliferative properties of primary precursor cells, as revealed by Ki67 expression. CONCLUSIONS The neuronal micro-environment provided by the culture of hippocampal slices is potent for induction of neurogenesis from oligodendrocyte NG2(+)/PDGFRalpha(+)/CNP(+) progenitor cells and promotes their differentiation not only into macroglia but also into neurons. It also sustains their proliferative capacity. The results indicate the crucial role of the local cellular environment in fate decision of primary NG2(+) multipotent neural progenitor cells, which may affect their behaviour after transplantation into the central nervous system.
Collapse
Affiliation(s)
- J Sypecka
- NeuroRepair Department, Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
| | | | | |
Collapse
|
36
|
Psychosine accumulates in membrane microdomains in the brain of krabbe patients, disrupting the raft architecture. J Neurosci 2009; 29:6068-77. [PMID: 19439584 DOI: 10.1523/jneurosci.5597-08.2009] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Lipid rafts (LRs) are membrane realms characterized by high concentrations of cholesterol and sphingolipids. Often, they are portrayed as scaffolds on which many different signaling molecules can assemble their cascades. The idea of rafts as scaffolds is garnering significant attention as the consequences of LR disruption have been shown to be manifest in multiple signaling pathways. In this study, LRs in the brain of the twitcher (TWI) mouse, a bona-fide model for infant variants of human globoid cell leukodystrophy or Krabbe disease, were investigated. This mouse has deficient activity of GALC (beta-galactosylceramidase) that leads to a progressive accumulation of some galactosyl-sphingolipids in the brain. We hypothesized that the accumulation of psychosine (galactosyl-sphingosine) in the TWI CNS may result in the disruption of rafts in different cell populations such as neurons and oligodendrocytes, both cellular targets during disease. In this communication, we demonstrate that psychosine specifically accumulates in LRs in the TWI brain and sciatic nerve and in samples from brains of human Krabbe patients. It is also shown that this accumulation is accompanied by an increase in cholesterol in these domains and changes in the distribution of the LR markers flotillin-2 and caveolin-1. Finally, we show evidence that this phenomenon may provide a mechanism by which psychosine can exert its known inhibitory effect on protein kinase C. This study provides a previously undescribed biophysical aspect for the mechanism of pathogenesis in Krabbe disease.
Collapse
|
37
|
Abstract
Recent advances in stem cell biology have raised expectations that both diseases of, and injuries to, the central nervous system may be ameliorated by cell transplantation. In particular, cell therapy has been studied for inducing efficient remyelination in disorders of myelin, including both the largely pediatric disorders of myelin formation and maintenance and the acquired demyelinations of both children and adults. Potential cell-based treatments of two major groups of disorders include both delivery of myelinogenic replacements and mobilization of residual oligodendrocyte progenitor cells as a means of stimulating endogenous repair; the choice of modality is then predicated upon the disease target. In this review we consider the potential application of cell-based therapeutic strategies to disorders of myelin, highlighting the promises as well as the problems and potential perils of this treatment approach.
Collapse
Affiliation(s)
- Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
38
|
Goldman SA, Schanz S, Windrem MS. Stem cell-based strategies for treating pediatric disorders of myelin. Hum Mol Genet 2008; 17:R76-83. [PMID: 18632701 DOI: 10.1093/hmg/ddn052] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The pediatric leukodystrophies comprise a category of disease manifested by neonatal or childhood deficiencies in myelin production or maintenance; these may be due to hereditary defects in one or more genes critical to the initiation of myelination, as in Pelizaeus-Merzbacher Disease, or to enzymatic deficiencies with aberrant substrate accumulation-related dysfunction, as in the lysosomal storage disorders. Despite differences in both phenotype and natural history, these disorders are all essentially manifested by a profound deterioration in neurological function with age. A congenital deficit in forebrain myelination is also noted in children with the periventricular leukomalacia of cerebral palsy, another major source of neurological morbidity. In light of the wide range of disorders to which congenital hypomyelination and/or postnatal demyelination may contribute, and the relative homogeneity of central oligodendrocytes and their progenitors, the pediatric leukodystrophies may be especially attractive targets for cell-based therapeutic strategies. As a result, glial progenitor cells (GPCs), which can give rise to new myelinogenic oligodendrocytes, have become of great interest as potential therapeutic vectors for the restoration of myelin to the hypomyelinated or dysmyelinated childhood CNS. In addition, by distributing themselves throughout the deficient host neuraxis after perinatal allograft, and giving rise to astrocytes as well as oligodendrocytes, glial progenitors appear to be of potential great utility in rectifying enzymatic deficiencies. In this review, we focus on current efforts to develop the use of isolated human GPCs as transplantable agents both for mediating enzymatic restoration to the enzyme-deficient brain and for therapeutic myelination in the disorders of congenital hypomyelination.
Collapse
Affiliation(s)
- Steven A Goldman
- Division of Cell and Gene Therapy and Center for Translational Neuromedicine, Department of Neurology and Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
39
|
The Role and Metabolism of Sulfatide in the Nervous System. Mol Neurobiol 2008; 37:93-103. [DOI: 10.1007/s12035-008-8022-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/09/2008] [Indexed: 12/16/2022]
|
40
|
Abstract
UNLABELLED Metachromatic leukodystrophy is a lysosomal storage disease caused by the deficiency of arylsulphatase A (ASA). This leads to storage of the membrane lipid sulphatide, which is abundant in myelin. A pathological hallmark of the disease is demyelination, causing various and ultimately lethal neurological symptoms. Today more than 110 mutations in the ASA gene have been identified, of which only three are frequent. Patients homozygous for alleles, which do not allow for the synthesis of functional ASA always suffer from the severe form of the disease, whereas alleles allowing the expression of residual enzyme activity are associated with the later onset juvenile or adult forms of metachromatic leukodystrophy. In addition, there are other as yet unknown genetic or epigenetic factors modifying the phenotype substantially. ASA-deficient mice have been generated as a model of metachromatic leukodystrophy. These mice store sulphatide and show progressive neurological symptoms, but do not demyelinate. This animal model was recently improved using a transgenic approach, which generated mice in which sulphatide synthesis in myelin-producing cells is enhanced. This new animal model reflects the pathological characteristics of the human disease. ASA-deficient mice have been used in various therapeutic trials involving enzyme replacement, haematopoietic stem-cell-based gene therapy and direct injections of ASA-expressing viral vectors into the brain. These animal studies have paved the way for future clinical studies of enzyme replacement and gene therapy. CONCLUSION For many years this devastating disorder was considered untreatable and the outlook for patients was poor. Within a comparatively short period of time since the ASA gene was cloned in 1989, genetic and biochemical studies and data generated from newly developed animal models have led to the first clinical trials. It is hoped that these developments will prove beneficial for patients.
Collapse
Affiliation(s)
- Volkmar Gieselmann
- Institut für Physiologische Chemie, Rheinische-Friedrich-Wilhems Universität Bonn, Bonn, Germany.
| |
Collapse
|
41
|
Glaser T, Schmandt T, Brüstle O. Generation and potential biomedical applications of embryonic stem cell-derived glial precursors. J Neurol Sci 2008; 265:47-58. [DOI: 10.1016/j.jns.2007.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 09/03/2007] [Accepted: 09/07/2007] [Indexed: 01/19/2023]
|
42
|
Autonomic denervation of lymphoid organs leads to epigenetic immune atrophy in a mouse model of Krabbe disease. J Neurosci 2008; 27:13730-8. [PMID: 18077684 DOI: 10.1523/jneurosci.3379-07.2007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Lysosomal beta-galactosylceramidase deficiency results in demyelination and inflammation in the nervous system causing the neurological Krabbe disease. In the Twitcher mouse model of this disease, we found that neurological symptoms parallel progressive and severe lymphopenia. Although lymphopoiesis is normal before disease onset, primary and secondary lymphoid organs progressively degenerate afterward. This occurs despite preserved erythropoiesis and leads to severe peripheral lymphopenia caused by reduced numbers of T cell precursors and mature lymphocytes. Hematopoietic cell replacement experiments support the existence of an epigenetic factor in mutant mice reconcilable with a progressive loss of autonomic axons that hampers thymic functionality. We propose that degeneration of autonomic nerves leads to the irreversible thymic atrophy and loss of immune-competence. Our study describes a new aspect of Krabbe disease, placing patients at risk of immune-related pathologies, and identifies a novel target for therapeutic interventions.
Collapse
|
43
|
Eichler F, Van Haren K. Immune response in leukodystrophies. Pediatr Neurol 2007; 37:235-44. [PMID: 17903666 DOI: 10.1016/j.pediatrneurol.2007.06.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 05/02/2007] [Accepted: 06/27/2007] [Indexed: 01/13/2023]
Abstract
Although the genetics and biochemistry of leukodystrophies have been extensively explored, the immune response in these disorders has received relatively little attention. Both the disease course and its response to treatment may be highly dependent on the immune system. In this review, we compare three common leukodystrophies, each with a different immune response: (1) X-linked adrenoleukodystrophy, which demonstrates a severe, lymphocytic inflammatory response; (2) metachromatic leukodystrophy, which yields a histiocytic response; and (3) vanishing white-matter disease, in which no inflammation is typically seen. We highlight the biochemical, pathologic, and clinical differences, while focusing on the immune response in each disease. We also review the response of leukodystrophies to immunomodulatory therapies and interventions such as hematopoietic stem-cell transplantation. Future studies may delineate specific inflammatory markers as possible candidates for therapeutic intervention.
Collapse
Affiliation(s)
- Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
44
|
Vawda R, Woodbury J, Covey M, Levison SW, Mehmet H. Stem cell therapies for perinatal brain injuries. Semin Fetal Neonatal Med 2007; 12:259-72. [PMID: 17553762 DOI: 10.1016/j.siny.2007.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This chapter reviews four groups of paediatric brain injury. The pathophysiology of these injuries is discussed to establish which cells are damaged and therefore which cells represent targets for cell replacement. Next, we review potential sources of cellular replacements, including embryonic stem cells, fetal and neonatal neural stem cells and a variety of mesenchymal stem cells. The advantages and disadvantages of each source are discussed. We review published studies to illustrate where stem cell therapies have been evaluated for therapeutic gain and discuss the hurdles that will need to be overcome to achieve therapeutic benefit. Overall, we conclude that children with paediatric brain injuries or inherited genetic disorders that affect the brain are worthy candidates for stem cell therapeutics.
Collapse
Affiliation(s)
- Reaz Vawda
- RY80Y-215, Merck Research Laboratories, Rahway, NJ 07065, USA
| | | | | | | | | |
Collapse
|
45
|
Biffi A, Naldini L. Novel candidate disease for gene therapy: metachromatic leukodystrophy. Expert Opin Biol Ther 2007; 7:1193-205. [PMID: 17696818 DOI: 10.1517/14712598.7.8.1193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metachromatic leukodystrophy (MLD) is a rare, fatal, inherited, autosomal recessive, lysosomal storage disorder, characterized by severe and progressive demyelination affecting the central and peripheral nervous systems. Despite some initial expectations in hematopoietic stem cell transplantation, and despite the ameliorated supportive therapy, MLD remains a life-threatening disease, with an extremely poor quality of life and a severe prognosis for all affected patients. Prospectively, in children affected by MLD, who have no other therapeutic option and an extremely poor prognosis, the potential risks associated with the use of a novel technology, such as gene therapy, might be well balanced by the potential benefit of a positive outcome. Thus, MLD might be considered an optimal candidate disease for testing innovative and potentially efficacious therapeutic approaches. Some of the gene therapy approaches discussed here, such as hematopoietic stem cells gene therapy, are likely to enter clinical testing in the near future.
Collapse
Affiliation(s)
- Alessandra Biffi
- San Raffaele Telethon Institute for Gene Therapy and Vita Salute University, H. San Raffaele Scientific Institute, Milan, Italy. a.biffi @hsr.it
| | | |
Collapse
|
46
|
Zawadzka M, Franklin RJM. Myelin regeneration in demyelinating disorders: new developments in biology and clinical pathology. Curr Opin Neurol 2007; 20:294-8. [PMID: 17495623 DOI: 10.1097/wco.0b013e32813aee7f] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The following article reviews recent advances in remyelination biology and its presentation in demyelinating diseases such as multiple sclerosis. It is based primarily on articles published during 2006. RECENT FINDINGS The principal themes are the identity and properties of remyelinating cells; the role of inflammation in remyelination; and the complexity of and redundancy within the signalling environment regulating remyelination. SUMMARY Central nervous system remyelination is mainly mediated by oligodendrocyte precursor cells, although subventricular zone-derived cells contribute to the repair of periventricular lesions. Oligodendrocyte precursor cells may differentiate into astrocytes and Schwann cells following injury and exhibit more stem cell-like features than previously recognized. A complex matrix of environmental factors, including cytokines, chemokines and growth factors, act upon oligodendrocyte precursor cells, causing their activation and eventual differentiation into remyelinating oligodendrocytes. Inflammatory cells contribute by providing components of the signalling matrix and by the phagocytic removal of myelin debris. Many factors within the signalling environment have redundant functions - a feature of regeneration with implications for developing remyelination therapies. Advances in remyelination biology have been accompanied by more detailed analyses of remyelination in multiple sclerosis and important translational developments, including the ability to identify myelin by positron emission tomography.
Collapse
Affiliation(s)
- Malgorzata Zawadzka
- Department of Veterinary Medicine and Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
47
|
Keyoung HM, Goldman SA. Glial progenitor-based repair of demyelinating neurological diseases. Neurosurg Clin N Am 2007; 18:93-104, x. [PMID: 17244557 DOI: 10.1016/j.nec.2006.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Demyelinating diseases of the brain and spinal cord affect more than one-quarter million of Americans, with numbers reaching more than two million across the world. These patients experience not only the vascular, traumatic, and inflammatory demyelinations of adulthood but the congenital and childhood dysmyelinating syndromes of the pediatric leukodystrophies. Several disease-modifying strategies have been developed that slow disease progression, especially in the inflammatory demyelinations and in multiple sclerosis in particular. Yet, currently available disease modifiers typically influence the immune system and are neither intended to nor competent to reverse the structural neurologic damage attending acquired demyelination. Fortunately, however, the disorders of myelin lend themselves well to attempts at structural repair, because central oligodendrocytes are the primary, and often sole, victims of the underlying disease process. Given the relative availability and homogeneity of human oligodendrocyte progenitor cells, the disorders of myelin formation and maintenance may be especially compelling targets for cell-based neurologic therapy.
Collapse
Affiliation(s)
- H Michael Keyoung
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, M779, PO Box 0470, San Francisco, CA 94143-0470, USA.
| | | |
Collapse
|
48
|
Sevin C, Aubourg P, Cartier N. Enzyme, cell and gene-based therapies for metachromatic leukodystrophy. J Inherit Metab Dis 2007; 30:175-83. [PMID: 17347913 DOI: 10.1007/s10545-007-0540-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 01/29/2007] [Accepted: 01/30/2007] [Indexed: 12/11/2022]
Abstract
Metachromatic leukodystrophy (MLD) is a demyelinating storage disease caused by deficiency of the lysosomal enzyme arylsulfatase A (ARSA). Lack of ARSA activity leads to the accumulation of galactosylceramide-3-O-sulfate (sulfatide) in the central and peripheral nervous systems. Based on the age at onset, the disease is usually classified into three forms: the late-infantile form, which manifests in the second year of life; the juvenile variants (onset between 4 and 12 years), which are subdivided into early-juvenile (EJ, onset before 6 years) and late-juvenile (LJ, onset after 6 years); and the adult form (onset after 12 years of age). Currently, there is no efficient therapy for the late-infantile form of MLD (50% of the patients), death occurring within a few years after onset of neurological symptoms. Allogeneic haematopoietic cell transplantation (HCT), when performed at a very early stage of the disease, may improve selected patients with juvenile or adult forms of MLD. As with other lysosomal storage diseases, the physiopathology of MLD is poorly understood. Demyelination is the main pathological finding, but substantial storage of sulfatides in neurons also occurs, and may contribute to the clinical phenotype. The physiopathological process leading to neuronal and glial cell degeneration and apoptosis involves accumulation of undegraded sulfatides but also secondary abnormalities (storage/mislocalization of unrelated lipids, inflammatory processes). This review summarizes the recent advances in the understanding of the physiopathology of MLD and the new therapeutic perspectives currently under preclinical investigation, including enzyme replacement therapy, gene therapy and cell therapy.
Collapse
Affiliation(s)
- C Sevin
- University René-Descartes Paris 5, INSERM U745, Paris, France
| | | | | |
Collapse
|
49
|
Lee JP, Jeyakumar M, Gonzalez R, Takahashi H, Lee PJ, Baek RC, Clark D, Rose H, Fu G, Clarke J, McKercher S, Meerloo J, Muller FJ, Park KI, Butters TD, Dwek RA, Schwartz P, Tong G, Wenger D, Lipton SA, Seyfried TN, Platt FM, Snyder EY. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat Med 2007; 13:439-47. [PMID: 17351625 DOI: 10.1038/nm1548] [Citation(s) in RCA: 239] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Accepted: 01/16/2007] [Indexed: 02/07/2023]
Abstract
Intracranial transplantation of neural stem cells (NSCs) delayed disease onset, preserved motor function, reduced pathology and prolonged survival in a mouse model of Sandhoff disease, a lethal gangliosidosis. Although donor-derived neurons were electrophysiologically active within chimeric regions, the small degree of neuronal replacement alone could not account for the improvement. NSCs also increased brain beta-hexosaminidase levels, reduced ganglioside storage and diminished activated microgliosis. Additionally, when oral glycosphingolipid biosynthesis inhibitors (beta-hexosaminidase substrate inhibitors) were combined with NSC transplantation, substantial synergy resulted. Efficacy extended to human NSCs, both to those isolated directly from the central nervous system (CNS) and to those derived secondarily from embryonic stem cells. Appreciating that NSCs exhibit a broad repertoire of potentially therapeutic actions, of which neuronal replacement is but one, may help in formulating rational multimodal strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jean-Pyo Lee
- Stem Cell & Regeneration Program, Center for Neuroscience and Aging Research, Burnham Institute for Medical Research, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Galbiati F, Clementi G, Superchi D, Givogri MI, Bongarzone ER. Effects of irradiation on the postnatal development of the brain in a genetic mouse model of globoid cell leukodystrophy. Neurochem Res 2007; 32:377-88. [PMID: 17203404 DOI: 10.1007/s11064-006-9247-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Accepted: 07/24/2006] [Indexed: 11/30/2022]
Abstract
Irradiation is one way to condition Twitcher mice--a natural model of globoid cell leukodystrophy (GLD)--prior to receive bone marrow transplantation (BMT). BMT showed to delay but not to completely prevent GLD disease in treated mutants. The reasons why BMT is not completely preventive in Twitchers are unclear but we speculate that irradiation might contribute to worsen the neurological impairments generated by the disease by altering postnatal neurogenesis. To test this hypothesis, we examined proliferation, migration and differentiation of neural precursors in neurogenic areas of the Twitcher brain after exposure of 5 day-old mutant pups to 620 rad, a non-lethal dose that leads to 80-90% of bone-marrow engraftment in classic BMT. Twitchers showed to be sensitive to irradiation, leading to a severe retardation of body growth of irradiated mutants. Irradiated Twitchers had reduced proliferation of neural precursors and increased astrogliosis and microgliosis, with reduced numbers of migratory neuroblasts and significantly less brain myelination. These effects were accompanied by caspase-3 activation and appeared largely irreversible in the lifespan of the Twitcher. Our work confirms that exposure of the neonatal brain to irradiation conditions such as those performed prior to BMT, can lead to long-lasting alterations of postnatal neurogenesis and myelination, which might contribute to worsen the progression of disease in these myelin mutants and to reduce the success of BMT.
Collapse
Affiliation(s)
- Francesca Galbiati
- San Raffaele Scientific Institute, Via Olgettina 58, Milano 20132, Italy
| | | | | | | | | |
Collapse
|