Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Glia ; 60(7): 1117-29, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22499166

ABSTRACT

Human glial precursor cells (hGPs) have potential for remyelinating lesions and are an attractive cell source for cell therapy of multiple sclerosis (MS). To investigate whether transplanted hGPs can affect the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of MS, we evaluated the therapeutic effects of transplanted hGPs together with the in vivo fate of these cells using magnetic resonance imaging (MRI) and bioluminescence imaging (BLI). At 14 days post-EAE induction, mice (n = 19) were intracerebroventricularly (ICV) injected with 5 × 10(5) hGPs that were magnetically labeled with superparamagnetic iron oxide (SPIO) particles as MR contrast agent and transduced with firefly luciferase for BLI of cell survival. Control mice (n = 18) received phosphate buffered saline (PBS) vehicle only. The severity of EAE clinical disability in the hGP-transplanted group was significantly suppressed (P < 0.05) with concomitant inhibition of ConA and MOG-specific T cell proliferation in the spleen. Astrogliosis was reduced and a lower activity of macrophages and/or microglia was observed in the spinal cord (P < 0.05). On MRI, SPIO signal was detected within the lateral ventricle from 1 day post-transplantation and remained there for up to 34 days. BLI indicated that most cells did not survive beyond 5-10 days, consistent with the lack of detectable migration into the brain parenchyma and the histological presence of an abundance of apoptotic cells. Transplanted hGPs could not be detected in the spleen. We conclude that ICV transplantation of short-lived hGPs can have a remote therapeutic effect through immunomodulation from within the ventricle, without cells directly participating in remyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Neural Stem Cells/transplantation , Neuroglia/transplantation , T-Lymphocytes/immunology , Animals , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunologic Factors , Injections, Intraventricular , Mice , Stem Cell Transplantation/methods
2.
Glia ; 59(3): 499-510, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264955

ABSTRACT

Transplantation of glial progenitor cells results in transplant-derived myelination and improved function in rodents with genetic dysmyelination or chemical demyelination. However, glial cell transplantation in adult CNS inflammatory demyelinating models has not been well studied. Here we transplanted human glial-restricted progenitor (hGRP) cells into the spinal cord of adult rats with inflammatory demyelination, and monitored cell fate in chemically immunosuppressed animals. We found that hGRPs migrate extensively, expand within inflammatory spinal cord lesions, do not form tumors, and adopt a mature glial phenotype, albeit at a low rate. Human GRP-transplanted rats, but not controls, exhibited preserved electrophysiological conduction across the spinal cord, though no differences in behavioral improvement were noted between the two groups. Although these hGRPs myelinated extensively after implantation into neonatal shiverer mouse brain, only marginal remyelination was observed in the inflammatory spinal cord demyelination model. The low rate of transplant-derived myelination in adult rat spinal cord may reflect host age, species, transplant environment/location, and/or immune suppression regime differences. We conclude that hGRPs have the capacity to myelinate dysmyelinated neonatal rodent brain and preserve conduction in the inflammatory demyelinated adult rodent spinal cord. The latter benefit is likely dependent on trophic support and suggests further exploration of potential of glial progenitors in animal models of chronic inflammatory demyelination.


Subject(s)
Demyelinating Diseases/surgery , Inflammation Mediators/physiology , Myelitis/surgery , Neuroglia/physiology , Neuroglia/transplantation , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Animals, Newborn , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Female , Graft Survival/physiology , Humans , Mice , Mice, Knockout , Mice, Neurologic Mutants , Myelitis/pathology , Myelitis/physiopathology , Neuroglia/cytology , Neuroglia/pathology , Rats , Rats, Inbred Lew , Recovery of Function/physiology , Stem Cells/cytology , Stem Cells/pathology
3.
Stem Cells Transl Med ; 8(4): 355-365, 2019 04.
Article in English | MEDLINE | ID: mdl-30618148

ABSTRACT

One of the fundamental limitations in assessing potential efficacy in Central Nervous System (CNS) transplantation of stem cells is the capacity for monitoring cell survival and migration noninvasively and longitudinally. Human glial-restricted progenitor (hGRP) cells (Q-Cells) have been investigated for their utility in providing neuroprotection following transplantation into models of amyotrophic lateral sclerosis (ALS) and have been granted a Food and Drug Administration (FDA) Investigational New Drug (IND) for intraspinal transplantation in ALS patients. Furthermore, clinical development of these cells for therapeutic use will rely on the ability to track the cells using noninvasive imaging methodologies as well as the verification that the transplanted GRPs have disease-relevant activity. As a first step in development, we investigated the use of a perfluorocarbon (PFC) dual-modal (19 F magnetic resonance imaging [MRI] and fluorescence) tracer agent to label Q-Cells in culture and following spinal cord transplantation. PFCs have a number of potential benefits that make them appealing for clinical use. They are quantitative, noninvasive, biologically inert, and highly specific. In this study, we developed optimized PFC labeling protocols for Q-Cells and demonstrate that PFCs do not significantly alter the glial identity of Q-Cells. We also show that PFCs do not interfere with the capacity for differentiation into astrocytes either in vitro or following transplantation into the ventral horn of the mouse spinal cord, and can be visualized in vivo by hot spot 19 F MRI. These studies provide a foundation for further preclinical development of PFCs within the context of evaluating Q-Cell transplantation in the brain and spinal cord of future ALS patients using 19 F MRI. Stem Cells Translational Medicine 2019;8:355-365.


Subject(s)
Fluorocarbons/administration & dosage , Neuroglia/cytology , Stem Cells/cytology , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/therapy , Animals , Astrocytes/cytology , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Fluorine-19 Magnetic Resonance Imaging/methods , Humans , Male , Mice , Spinal Cord/cytology , Spinal Cord/diagnostic imaging , Stem Cell Transplantation/methods
4.
BMC Dev Biol ; 8: 102, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18947415

ABSTRACT

BACKGROUND: We have generated gene expression databases for human glial precursors, neuronal precursors, astrocyte precursors and neural stem cells and focused on comparing the profile of glial precursors with that of other populations. RESULTS: A total of 14 samples were analyzed. Each population, previously distinguished from each other by immunocytochemical analysis of cell surface markers, expressed genes related to their key differentiation pathways. For the glial precursor cell population, we identified 458 genes that were uniquely expressed. Expression of a subset of these individual genes was validated by RT-PCR. We also report genes encoding cell surface markers that may be useful for identification and purification of human glial precursor populations. CONCLUSION: We provide gene expression profile for human glial precursors. Our data suggest several signaling pathways that are important for proliferation and differentiation of human glial precursors. Such information may be utilized to further purify glial precursor populations, optimize media formulation, or study the effects of glial differentiation.


Subject(s)
Gene Expression Profiling , Neuroglia/metabolism , Stem Cells/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Differentiation/genetics , Cell Separation , Cells, Cultured , Fetus/cytology , Humans , Neuroglia/physiology , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Exp Neurol ; 291: 74-86, 2017 05.
Article in English | MEDLINE | ID: mdl-28163160

ABSTRACT

The therapeutic effect of glial progenitor transplantation in diseases of dysmyelination is currently attributed to the formation of new myelin. Using magnetic resonance imaging (MRI), we show that the therapeutic outcome in dysmyelinated shiverer mice is dependent on the extent of cell migration but not the presence of mature and compact myelin. Human or mouse glial restricted progenitors (GRPs) were transplanted into rag2-/- shiverer mouse neonates and followed for over one year. Mouse GRPs produced mature myelin as detected with multi-parametric MRI, but showed limited migration without extended animal lifespan. In sharp contrast, human GRPs migrated extensively and significantly increased animal survival, but production of mature myelin did not occur until 46weeks post-grafting. We conclude that human GRPs can extend the survival of transplanted shiverer mice prior to production of mature myelin, while mouse GRPs fail to extend animal survival despite the early presence of mature myelin. This paradox suggests that transplanted GRPs provide therapeutic benefits through biological processes other than the formation of mature myelin capable to foster rapid nerve conduction, challenging the current dogma of the primary role of myelination in regaining function of the central nervous system.


Subject(s)
Demyelinating Diseases/surgery , Myelin Sheath/metabolism , Neuroglia/transplantation , Stem Cell Transplantation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/diagnostic imaging , Cell Differentiation , Cell Movement , Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/pathology , Disease Models, Animal , Gangliosides/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Neuroglia/ultrastructure , Oligodendrocyte Transcription Factor 2 , Spinal Cord/diagnostic imaging , Time Factors , Tubulin/metabolism
6.
J Neurosci ; 23(2): 392-402, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12533599

ABSTRACT

Synaptic differentiation results from an exchange of informational molecules between synaptic partners during development. At the vertebrate neuromuscular junction, agrin is one molecule presented by the presynaptic motor neuron that plays an instructive role in postsynaptic differentiation of the muscle cell, most notably in aggregation of acetylcholine receptors (AChRs). Although agrin is the best-characterized synaptogenic molecule, its mechanism of action remains uncertain, but clearly, it requires the receptor tyrosine kinase MuSK (muscle-specific kinase), the intracellular protein rapsyn, an Src-like kinase, and cytoskeletal components. In addition, the transmembrane protein dystroglycan interacts with the cytoskeleton and is implicated in agrin responsiveness. This alpha-beta heterodimer can bind agrin via its extracellular alpha subunit and associates with the membrane cytoskeleton via its beta subunit. In this study, we demonstrate that overexpression of the beta subunit of dystroglycan in cultured muscle cells inhibits agrin-induced AChR clustering. Deletion analysis and point mutagenesis demonstrate that the inhibition is mediated by an intracellular, juxtamembrane region composed of basic amino acids. Finally, the inhibition mediated by beta-dystroglycan extends to the minimal agrin fragment required for AChR clustering, suggesting that dystroglycan plays an important role in postsynaptic differentiation in response to agrin.


Subject(s)
Agrin/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptor Aggregation/physiology , Receptors, Cholinergic/metabolism , Agrin/pharmacology , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cytoskeletal Proteins/genetics , Dystroglycans , Epitopes , Green Fluorescent Proteins , Luminescent Proteins/genetics , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Mutagenesis, Site-Directed , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Receptor Aggregation/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Structure-Activity Relationship , Transfection
7.
Exp Neurol ; 264: 188-99, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25523812

ABSTRACT

Although Amyotrophic Lateral Sclerosis (ALS) is a motor neuron disease, basic research studies have highlighted that astrocytes contribute to the disease process. Therefore, strategies which replace the diseased astrocyte population with healthy astrocytes may protect against motor neuron degeneration. Our studies have sought to evaluate astrocyte replacement using glial-restricted progenitors (GRPs), which are lineage-restricted precursors capable of differentiating into astrocytes after transplantation. The goal of our current study was to evaluate how transplantation to the diseased ALS spinal cord versus a healthy, wild-type spinal cord may affect human GRP engraftment and selected gene expression. Human GRPs were transplanted into the spinal cord of either an ALS mouse model or wild-type littermate mice. Mice were sacrificed for analysis at either the onset of disease course or at the endstage of disease. The transplanted GRPs were analyzed by immunohistochemistry and NanoString gene profiling which showed no gross differences in the engraftment or gene expression of the cells. Our data indicate that human glial progenitor engraftment and gene expression is independent of the neurodegenerative ALS spinal cord environment. These findings are of interest given that human GRPs are currently in clinical development for spinal cord transplantation into ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/surgery , Gene Expression Regulation/physiology , Neuroglia/physiology , Neuroglia/transplantation , Stem Cell Transplantation/methods , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Antigens, Nuclear/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadaver , Cell Movement , Cell Proliferation/genetics , Disease Models, Animal , Fetus , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Spinal Cord/metabolism , Spinal Cord/pathology , Stem Cells , Superoxide Dismutase/genetics
8.
J Neurotrauma ; 28(4): 579-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21222572

ABSTRACT

Transplantation of neural progenitors remains a promising therapeutic approach to spinal cord injury (SCI), but the anatomical and functional evaluation of their effects is complex, particularly when using human cells. We investigated the outcome of transplanting human glial-restricted progenitors (hGRP) and astrocytes derived from hGRP (hGDA) in spinal cord contusion with respect to cell fate and host response using athymic rats to circumvent xenograft immune issues. Nine days after injury hGRP, hGDA, or medium were injected into the lesion center and rostral and caudal to the lesion, followed by behavioral testing for 8 weeks. Both hGRP and hGDA showed robust graft survival and extensive migration. The total number of cells increased 3.5-fold for hGRP, and twofold for hGDA, indicating graft expansion, but few proliferating cells remained by 8 weeks. Grafted cells differentiated into glia, predominantly astrocytes, and few remained at progenitor state. About 80% of grafted cells around the injury were glial fibrillary acidic protein (GFAP)-positive, gradually decreasing to 40-50% at a distance of 6 mm. Conversely, there were few graft-derived oligodendrocytes at the lesion, but their numbers increased away from the injury to 30-40%. Both cell grafts reduced cyst and scar formation at the injury site compared to controls. Microglia/macrophages were present at and around the lesion area, and axons grew along the spared tissue with no differences among groups. There were no significant improvements in motor function recovery as measured by the Basso, Beattie, and Bresnahan (BBB) scale and grid tests in all experimental groups. Cystometry revealed that hGRP grafts attenuated hyperactive bladder reflexes. Importantly, there was no increased sensory or tactile sensitivity associated with pain, and the hGDA group showed sensory function returning to normal. Although the improved lesion environment was not sufficient for robust functional recovery, the permissive properties and lack of sensory hypersensitivity indicate that human GRP and astrocytes remain promising candidates for therapy after SCI.


Subject(s)
Nerve Regeneration/physiology , Neuroglia/transplantation , Spinal Cord Injuries/therapy , Analysis of Variance , Animals , Cell Count , Cell Differentiation , Female , Immunohistochemistry , Lumbar Vertebrae , Motor Activity/physiology , Neuroglia/metabolism , Random Allocation , Rats , Rats, Nude , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Stem Cell Transplantation
9.
PLoS One ; 6(10): e25968, 2011.
Article in English | MEDLINE | ID: mdl-21998733

ABSTRACT

Cellular abnormalities are not limited to motor neurons in amyotrophic lateral sclerosis (ALS). There are numerous observations of astrocyte dysfunction in both humans with ALS and in SOD1(G93A) rodents, a widely studied ALS model. The present study therapeutically targeted astrocyte replacement in this model via transplantation of human Glial-Restricted Progenitors (hGRPs), lineage-restricted progenitors derived from human fetal neural tissue. Our previous findings demonstrated that transplantation of rodent-derived GRPs into cervical spinal cord ventral gray matter (in order to target therapy to diaphragmatic function) resulted in therapeutic efficacy in the SOD1(G93A) rat. Those findings demonstrated the feasibility and efficacy of transplantation-based astrocyte replacement for ALS, and also show that targeted multi-segmental cell delivery to cervical spinal cord is a promising therapeutic strategy, particularly because of its relevance to addressing respiratory compromise associated with ALS. The present study investigated the safety and in vivo survival, distribution, differentiation, and potential efficacy of hGRPs in the SOD1(G93A) mouse. hGRP transplants robustly survived and migrated in both gray and white matter and differentiated into astrocytes in SOD1(G93A) mice spinal cord, despite ongoing disease progression. However, cervical spinal cord transplants did not result in motor neuron protection or any therapeutic benefits on functional outcome measures. This study provides an in vivo characterization of this glial progenitor cell and provides a foundation for understanding their capacity for survival, integration within host tissues, differentiation into glial subtypes, migration, and lack of toxicity or tumor formation.


Subject(s)
Amyotrophic Lateral Sclerosis/surgery , Cervical Vertebrae/surgery , Neuroglia/cytology , Spinal Cord/surgery , Stem Cell Transplantation/methods , Stem Cells/cytology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Anterior Horn Cells/drug effects , Anterior Horn Cells/pathology , Astrocytes/cytology , Astrocytes/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cervical Vertebrae/drug effects , Cervical Vertebrae/immunology , Cervical Vertebrae/pathology , Cyclosporine/pharmacology , Disease Models, Animal , Female , Humans , Immunosuppression Therapy , Male , Mice , Mutation , Neuroglia/drug effects , Neurons/cytology , Neurons/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Pregnancy , Sirolimus/pharmacology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Stem Cell Transplantation/adverse effects , Stem Cells/drug effects , Superoxide Dismutase-1 , Tacrolimus/pharmacology
10.
Regen Med ; 5(3): 381-94, 2010 May.
Article in English | MEDLINE | ID: mdl-20455649

ABSTRACT

AIM: Glial-restricted progenitor cells (GRPs), a neural cell population that gives rise to astrocytes and oligodendrocytes both in vitro and in vivo, hold great promise as a cellular therapeutic for the treatment of demyelinating and neurodegenerative diseases of the CNS. The manufacturing and characterization protocols of human-derived GRPs (hGRPs; trade name Q-Cells) for use in a clinical setting that adhere to rigorous standards for their isolation, propagation, characterization and storage are presented. MATERIALS & METHODS: hGRPs, defined by their immunoreactivity with A2B5 antibodies, were isolated from fetal cadaver forebrain tissue of mice 17-24 weeks gestational age using Miltenyi paramagnetic bead cell separation technology. GRPs were grown in a defined xenobiotic-free medium for 6 days. At harvest, hGRPs were characterized using immunocytochemical techniques. Long-term cryopreservation and storage conditions, and viability upon freeze-thaw were determined. The phenotypic differentiation potential of hGRPs was determined by implantation experiments into the CNS of shiverer mice. RESULTS: hGRPs were isolated from over 50 neural tissues of either sex during gestational ages of 17-24 weeks. Cells expanded out to 6 days in vitro in a xenobiotic-free medium demonstrated very consistent immunocytochemical profiles. No residual antibody used in the purification process was detected after 6 days of growth in vitro. GRPs could be frozen at up to 24 million cells/ml and were over 70% viable upon freeze-thaw. Thawed hGRPs transplanted into the brain of the dysmyelinated shiverer mouse model were observed to differentiate into both glial fibrillary acidic protein-positive astrocytes and myelin basic protein-positive oligodendrocytes; no human-derived NeuN-positive neuronal cells were observed and no abnormal cell proliferation was observed. CONCLUSION: We demonstrate that hGRPs can be consistently obtained, propagated, cryopreserved and characterized using protocols that can be transferred to a good laboratory practice/good manufacturing practice setting for the manufacture of clinical-grade hGRP cellular therapeutics. Functional data demonstrate that cells manufactured under these conditions are able to differentiate into appropriate cellular phenotypes in an animal model of dysmyelination.


Subject(s)
Cell Transplantation/methods , Neurodegenerative Diseases/metabolism , Neuroglia/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Epitopes/chemistry , Female , Flow Cytometry/methods , Humans , Immunohistochemistry/methods , Male , Mice , Neurodegenerative Diseases/therapy , Prosencephalon/metabolism , Regenerative Medicine/methods
11.
Mol Cell Neurosci ; 20(1): 140-53, 2002 May.
Article in English | MEDLINE | ID: mdl-12056845

ABSTRACT

During synaptogenesis information exchanged locally between synaptic partners results in precise alignment of morphological and molecular specializations. For example, agrin derived from motoneurons induces localized postsynaptic differentiation at the neuromuscular synapse. Similar information molecules are thought to act at other synapses; however, techniques for directly evaluating synaptogenic activities of such molecules are lacking. Here we use agrin-induced differentiation as a model system to validate a novel approach for characterizing synaptogenic molecules. Proteins are patterned with micron scale resolution on glass coverslips by covalent microcontact printing and these substrates are used for cell culture. Postsynaptic molecules accumulate specifically at sites of contact between muscle cells and patterned agrin: a response which is quantifiable. Our results demonstrate that microcontact printing is applicable to the analysis of cellular response to locally immobilized information molecules.


Subject(s)
Agrin/chemistry , Cell Differentiation/physiology , Growth Substances/chemistry , Microchemistry/methods , Neurochemistry/methods , Neuromuscular Junction/growth & development , Printing/methods , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cells, Cultured , Collagen/chemistry , Image Processing, Computer-Assisted , Immunohistochemistry/instrumentation , Immunohistochemistry/methods , Mice , Mice, Knockout , Microchemistry/instrumentation , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Neurochemistry/instrumentation , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Printing/instrumentation , Rats , Receptors, Cholinergic/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL