Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters











Publication year range
1.
Neurochem Res ; 37(6): 1335-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22367451

ABSTRACT

Sandhoff Disease (SD) involves the CNS accumulation of ganglioside GM2 and asialo-GM2 (GA2) due to inherited defects in the ß-subunit gene of ß-hexosaminidase A and B (Hexb gene). Substrate reduction therapy, utilizing imino sugar N-butyldeoxygalactonojirimycin (NB-DGJ), reduces ganglioside biosynthesis and levels of stored GM2 in SD mice. Intracranial transplantation of Neural Stem Cells (NSCs) can provide enzymatic cross correction, to help reduce ganglioside storage and extend life. Here we tested the effect of NSCs and NB-DGJ, alone and together, on brain ß-hexosaminidase activity, GM2, and GA2 content in juvenile SD mice. The SD mice received either cerebral NSC transplantation at post-natal day 0 (p-0), intraperitoneal injection of NB-DGJ (500 mg/kg/day) from p-9 to p-15, or received dual treatments. The brains were analyzed at p-15. ß-galactosidase staining confirmed engraftment of lacZ-expressing NSCs in the cerebral cortex. Compared to untreated and sham-treated SD controls, NSC treatment alone provided a slight increase in Hex activity and significantly decreased GA2 content. However, NSCs had no effect on GM2 content when analyzed at p-15. NB-DGJ alone had no effect on Hex activity, but significantly reduced GM2 and GA2 content. Hex activity was slightly elevated in the NSC + drug-treated mice. GM2 and GA2 content in the dual treated mice were similar to that of the NB-DGJ treated mice. These data indicate that NB-DGJ alone was more effective in targeting storage in juvenile SD mice than were NSCs alone. No additive or synergistic effect between NSC and drug was found in these juvenile SD mice.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Neural Stem Cells/transplantation , Sandhoff Disease/therapy , 1-Deoxynojirimycin/therapeutic use , Animals , G(M2) Ganglioside , Hexosaminidase B/metabolism , Mice , Sandhoff Disease/drug therapy , beta-N-Acetylhexosaminidases/genetics
2.
Article in English | MEDLINE | ID: mdl-16315610

ABSTRACT

Neural stem and progenitor cells express a variety of receptors that enable them to sense and react to signals emanating from physiological and pathophysiological conditions in the brain as well as elsewhere in the body. Many of these receptors and were first described in investigations of the immune system, particularly with respect to hematopoietic stem cells. This emerging view of neurobiology has two major implications. First, many phenomena known from the hematopoietic system may actually be generalizable to stem cells from many organ systems, reflecting the cells' progenitor-mediated regenerative potential. Second, regenerative interfaces may exist between diverse organ systems; populations of cells of neuroectodermal and hematopoietic origin may interact to play a crucial role in normal brain physiology, pathology, and repair. An understanding of the origins of signals and the neural progenitors' responses might lead to the development of effective therapeutic strategies to counterbalance acute and chronic neurodegenerative processes. Such strategies may include modifying and modulating cells with regenerative potential in subtle ways. For example, stem cells might be able to detect pathology-associated signals and be used as "interpreters" to mediate drug and other therapeutic interventions. This review has focused on the role of inflammation in brain repair. We propose that resident astroglia and blood-born cells both contribute to an inflammatory signature that is unique to each kind of neuronal degeneration or injury. These cells play a key role in coordinating the neural progenitor cell response to brain injury by exerting direct and indirect environmentally mediated influence on neural progenitor cells. We suggest that investigations of the neural progenitor-immunologic interface will provide valuable data related to the mechanisms by which endogenous and exogenous neural progenitor cells react to brain pathology, ultimately aiding in the design of more effective therapeutic applications of stem cell biology. Such improvements will include: (1) ascertaining the proper timing for implanting exogenous neural progenitor cells in relation to the administration of anti-inflammatory agents; (2) identifying what types of molecules might be administered during injury to enhance the mobilization and differentiation of endogenous and exogenous neural progenitor cells while also inhibiting the detrimental aspects of the inflammatory reaction; (3) divining clues as to which molecules may be required to change the lesioned environment in order to invite the homing of reparative neural progenitor cells.


Subject(s)
Immune System , Nervous System/pathology , Animals , Brain/pathology , Cell Differentiation , Cell Lineage , Humans , Inflammation , Models, Biological , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Stem Cells/cytology
3.
Cell Transplant ; 14(4): 193-202, 2005.
Article in English | MEDLINE | ID: mdl-15929554

ABSTRACT

Neural stem cells (NSCs) of the central nervous system (CNS) recently have attracted a great deal of interest not only because of their importance in basic research on neural development, but also in terms of their therapeutic potential in neurological diseases, such as Parkinson's disease (PD). To examine if genetically modified NSCs are a suitable source for the cell and gene therapy of PD, an immortalized mouse NSC line, C17.2, was transduced with tyrosine hydroxylase (TH) gene and with GTP cyclohydrolase 1 (GTPCH1) gene, which are important enzymes in dopamine biosynthesis. The expression of TH in transduced C17.2-THGC cells was confirmed by RT-PCR, Western blot analysis, and immunocytochemistry, and expression of GTPCH1 by RT-PCR. The level of L-DOPA released by C17.2-THGC cells, as determined by HPLC assay, was 3793 pmol/10(6) cells, which is 760-fold higher than that produced by C17.2-TH cells, indicating that GTPCH1 expression is important for L-DOPA production by transduced C17.2 cells. Following the implantation of C17.2-THGcC NSCs into the striata of parkinsonian rats, a marked improvement in amphetamine-induced turning behavior was observed in parkinsonian rats grafted with C17.2-THGC cells but not in the control rats grafted with C17.2 cells. These results indicate that genetically modified NSCs grafted into the brain of the parkinsonian rats are capable of survival, migration, and neuronal differentiation. Collectively, these results suggest that NSCs have great potential as a source of cells for cell therapy and an effective vehicle for therapeutic gene transfer in Parkinson's disease.


Subject(s)
GTP Cyclohydrolase/genetics , Genetic Therapy/methods , Neurons/physiology , Parkinson Disease/therapy , Stem Cell Transplantation , Tyrosine 3-Monooxygenase/genetics , Animals , Behavior, Animal , Cell Differentiation , Female , GTP Cyclohydrolase/metabolism , Graft Survival , Humans , Levodopa/metabolism , Neurons/cytology , Rats , Rats, Sprague-Dawley , Stem Cells/physiology , Transduction, Genetic , Tyrosine 3-Monooxygenase/metabolism
4.
Exp Neurol ; 181(2): 115-29, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12781986

ABSTRACT

Neural stem cells (NSCs) offer the potential to replace lost tissue after nervous system injury. This study investigated whether grafts of NSCs (mouse clone C17.2) could also specifically support host axonal regeneration after spinal cord injury and sought to identify mechanisms underlying such growth. In vitro, prior to grafting, C17.2 NSCs were found for the first time to naturally constitutively secrete significant quantities of several neurotrophic factors by specific ELISA, including nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor. When grafted to cystic dorsal column lesions in the cervical spinal cord of adult rats, C17.2 NSCs supported extensive growth of host axons of known sensitivity to these growth factors when examined 2 weeks later. Quantitative real-time RT-PCR confirmed that grafted stem cells expressed neurotrophic factor genes in vivo. In addition, NSCs were genetically modified to produce neurotrophin-3, which significantly expanded NSC effects on host axons. Notably, overexpression of one growth factor had a reciprocal effect on expression of another factor. Thus, stem cells can promote host neural repair in part by secreting growth factors, and their regeneration-promoting activities can be modified by gene delivery.


Subject(s)
Neurons/physiology , Neurotrophin 3/metabolism , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Axons/physiology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Female , Glial Cell Line-Derived Neurotrophic Factor , Graft Survival/physiology , Humans , Mice , Neck , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Growth Factor/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/physiology , Neurons/cytology , Neurons/drug effects , Neurotrophin 3/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/pathology , Stem Cells/cytology , Transduction, Genetic
5.
Gene Ther ; 9(10): 613-24, 2002 May.
Article in English | MEDLINE | ID: mdl-12032707

ABSTRACT

The inherent biology of neural stem cells (NSCs) endows them with capabilities that not only circumvent many of the limitations of other gene transfer vehicles, but that enable a variety of novel therapeutic strategies heretofore regarded as beyond the purview of neural transplantation. Most neurodegenerative diseases are characterized not by discrete, focal abnormalities but rather by extensive, multifocal, or even global neuropathology. Such widely disseminated lesions have not conventionally been regarded as amenable to neural transplantation. However, the ability of NSCs to engraft diffusely and become integral members of structures throughout the host CNS, while also expressing therapeutic molecules, may permit these cells to address that challenge. Intriguingly, while NSCs can be readily engineered to express specified foreign genes, other intrinsic factors appear to emanate spontaneously from NSCs and, in the context of reciprocal donor-host signaling, seem to be capable of neuroprotective and/or neuroregenerative functions. Stem cells additionally have the appealing ability to 'home in' on pathology, even over great distances. Such observations help to advance the idea that NSCs - as a prototype for stem cells from other solid organs - might aid in reconstructing the molecular and cellular milieu of maldeveloped or damaged organs.


Subject(s)
Central Nervous System/cytology , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Models, Neurological , Nerve Regeneration , Trauma, Nervous System/therapy , Adult , Amyloidosis/therapy , Animals , Brain Ischemia/therapy , Brain Neoplasms/therapy , Humans , Intellectual Disability/therapy , Nerve Degeneration/therapy
6.
J Neurosci ; 21(20): 8108-18, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11588183

ABSTRACT

Neural stem cells (NSCs) have been proposed as tools for treating neurodegeneration because of their capacity to give rise to cell types appropriate to the structure in which they are grafted. In the present work, we explore the ability of NSCs to stably express transgenes and locally deliver soluble molecules with neuroprotective activity, such as glial cell line-derived neurotrophic factor (GDNF). NSCs engineered to release GDNF engrafted well in the host striatum, integrated and gave rise to neurons, astrocytes, and oligodendrocytes, and maintained stable high levels of GDNF expression for at least 4 months. The therapeutic potential of intrastriatal GDNF-NSCs grafts was tested in a mouse 6-hydroxydopamine model of Parkinson's disease. We found that GDNF-NSCs prevented the degeneration of dopaminergic neurons in the substantia nigra and reduced behavioral impairment in these animals. Thus, our results demonstrate that NSCs efficiently express therapeutic levels of GDNF in vivo, suggesting a use for NSCs engineered to release neuroprotective molecules in the treatment of neurodegenerative disorders, including Parkinson's disease.


Subject(s)
Nerve Growth Factors , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Parkinson Disease, Secondary/therapy , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Behavior, Animal/drug effects , Cell Differentiation , Cell Movement , Cell Survival/drug effects , Cells, Cultured , Clone Cells/metabolism , Clone Cells/transplantation , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor , Graft Survival/drug effects , Male , Mice , Mice, Nude , Nerve Tissue Proteins/pharmacology , Neurons/cytology , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Substantia Nigra/drug effects , Substantia Nigra/pathology
7.
J Neurosci Res ; 65(6): 549-64, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11550223

ABSTRACT

To test the idea that genetically engineered cells can rescue axotomized neurons, we transplanted fibroblasts and immortalized neural stem cells (NSCs) modified to express neurotrophic factors into the injured spinal cord. The neurotrophin-3 (NT-3) or nerve growth factor (NGF) transgene was introduced into these cells using recombinant retroviral vectors containing an internal ribosome entry site (IRES) sequence and the beta-galactosidase or alkaline phosphatase reporter gene. Bioassay confirmed biological activity of the secreted neurotrophic factors. Clarke's nucleus (CN) axons, which project to the rostral spinal cord and cerebellum, were cut unilaterally in adult rats by T8 hemisection. Rats received transplants of fibroblasts or NSCs genetically modified to express NT-3 or NGF and a reporter gene, only a reporter gene, or no transplant. Two months postoperatively, grafted cells survived at the hemisection site. Grafted fibroblasts and NSCs expressed a reporter gene and immunoreactivity for the NGF or NT-3 transgene. Rats receiving no transplant or a transplant expressing only a reporter gene showed a 30% loss of CN neurons in the L1 segment on the lesioned side. NGF-expressing transplants produced partial rescue compared with hemisection alone. There was no significant neuron loss in rats receiving grafts of either fibroblasts or NSCs engineered to express NT-3. We postulate that NT-3 mediates survival of CN neurons through interaction with trkC receptors, which are expressed on CN neurons. These results support the idea that NT-3 contributes to long-term survival of axotomized CN neurons and show that genetically modified cells rescue axotomized neurons as efficiently as fetal CNS transplants.


Subject(s)
Brain Tissue Transplantation/methods , Fibroblasts/transplantation , Nerve Regeneration/physiology , Neurotrophin 3/genetics , Spinal Cord Injuries/surgery , Stem Cell Transplantation , Transfection/methods , Animals , Axotomy , Cell Line, Transformed , Cell Survival/genetics , Chick Embryo , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/growth & development , Graft Survival/genetics , Immunohistochemistry , Mice , Nerve Growth Factor/genetics , Neurons/cytology , Rats , Rats, Sprague-Dawley , Receptor, trkC/metabolism , Retrograde Degeneration/physiopathology , Retrograde Degeneration/prevention & control , Retrograde Degeneration/surgery , Spinal Cord/cytology , Spinal Cord/surgery , Spinal Cord Injuries/physiopathology , Stem Cells/cytology , Stem Cells/metabolism
8.
Science ; 293(5536): 1820-4, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11474066

ABSTRACT

Many central nervous system regions at all stages of life contain neural stem cells (NSCs). We explored how these disparate NSC pools might emerge. A traceable clone of human NSCs was implanted intraventricularly to allow its integration into cerebral germinal zones of Old World monkey fetuses. The NSCs distributed into two subpopulations: One contributed to corticogenesis by migrating along radial glia to temporally appropriate layers of the cortical plate and differentiating into lamina-appropriate neurons or glia; the other remained undifferentiated and contributed to a secondary germinal zone (the subventricular zone) with occasional members interspersed throughout brain parenchyma. An early neurogenetic program allocates the progeny of NSCs either immediately for organogenesis or to undifferentiated pools for later use in the "postdevelopmental" brain.


Subject(s)
Cell Movement , Neocortex/cytology , Neocortex/embryology , Neurons/cytology , Prosencephalon/cytology , Prosencephalon/embryology , Stem Cells/cytology , Animals , Brain Tissue Transplantation , Cell Differentiation , Cell Lineage , Cell Transplantation , Clone Cells/cytology , Clone Cells/transplantation , Humans , Macaca radiata/embryology , Neurons/transplantation , Stem Cell Transplantation , Transplantation, Heterologous
9.
Novartis Found Symp ; 231: 242-62; discussion 262-9, 302-6, 2000.
Article in English | MEDLINE | ID: mdl-11131542

ABSTRACT

In recent years, it has become evident that the developing and even the adult mammalian CNS contain a population of undifferentiated, multipotent cell precursors, neural stem cells, the plastic properties of which might be of advantage for the design of more effective therapies for many neurological diseases. This article reviews the recent progress in establishing rodent and human clonal neural stem cell lines, their biological properties, and how these cells can be utilized to correct a variety of defects, with prospects for the near future to harness their behaviour for neural stem cell-based treatment of diseases in humans.


Subject(s)
Central Nervous System/surgery , Genetic Therapy/methods , Neurodegenerative Diseases/surgery , Neurodegenerative Diseases/therapy , Neurons/transplantation , Stem Cell Transplantation , Adult , Animals , Cell Line , Humans
10.
Proc Natl Acad Sci U S A ; 97(23): 12846-51, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11070094

ABSTRACT

One of the impediments to the treatment of brain tumors (e.g., gliomas) has been the degree to which they expand, infiltrate surrounding tissue, and migrate widely into normal brain, usually rendering them "elusive" to effective resection, irradiation, chemotherapy, or gene therapy. We demonstrate that neural stem cells (NSCs), when implanted into experimental intracranial gliomas in vivo in adult rodents, distribute themselves quickly and extensively throughout the tumor bed and migrate uniquely in juxtaposition to widely expanding and aggressively advancing tumor cells, while continuing to stably express a foreign gene. The NSCs "surround" the invading tumor border while "chasing down" infiltrating tumor cells. When implanted intracranially at distant sites from the tumor (e.g., into normal tissue, into the contralateral hemisphere, or into the cerebral ventricles), the donor cells migrate through normal tissue targeting the tumor cells (including human glioblastomas). When implanted outside the CNS intravascularly, NSCs will target an intracranial tumor. NSCs can deliver a therapeutically relevant molecule-cytosine deaminase-such that quantifiable reduction in tumor burden results. These data suggest the adjunctive use of inherently migratory NSCs as a delivery vehicle for targeting therapeutic genes and vectors to refractory, migratory, invasive brain tumors. More broadly, they suggest that NSC migration can be extensive, even in the adult brain and along nonstereotypical routes, if pathology (as modeled here by tumor) is present.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Glioblastoma/pathology , Neurons/physiology , Nucleoside Deaminases/genetics , Stem Cells/physiology , Animals , Brain Neoplasms/therapy , Cell Movement/physiology , Cytosine Deaminase , Disease Models, Animal , Female , Genetic Therapy/methods , Glioblastoma/therapy , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Nude , Neurons/cytology , Rats , Rats, Inbred F344 , Stem Cells/cytology , Tropism
11.
J Neurosci Res ; 61(6): 597-604, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10972956

ABSTRACT

The potential of a neural stem cell line to acquire cholinergic characteristics was studied in transplants injected into the septum/diagonal band nuclei of young adult rats and mice. The stem cells integrated within the nuclei and survived for up to 9 months. Three methods were used to identify the grafted cells and to show differentiation into astrocytes and neurons. Enhanced survival of the stem cells occurred in the host brain with a previous lesion of the fimbria-fornix pathway. Differentiated cells acquired neuronal-like features including the expression of neurofilament subunits. In lesioned hosts, subpopulations of the grafted cells acquired a cholinergic neuronal phenotype and expressed choline acetyltransferase and the p75 neurotrophin receptor. Cells that developed into astrocytes were often associated with blood vessels and expressed glial fibrillary acidic protein. The results further exemplify the potential of stem cell lines and the property of site-specific differentiation when this line is transplanted to the cholinergic system of the adult brain.


Subject(s)
Acetylcholine/physiology , Brain Tissue Transplantation , Diagonal Band of Broca/surgery , Neurons/transplantation , Septal Nuclei/surgery , Stem Cell Transplantation , Age Factors , Animals , Astrocytes/cytology , Cell Differentiation/physiology , Cell Line , Choline O-Acetyltransferase/analysis , Denervation , Fluorescent Dyes , Fornix, Brain/cytology , Graft Survival/physiology , Lac Operon , Mice , Neurons/cytology , Neurons/enzymology , Phenotype , Rats , Rats, Inbred Lew , Stem Cells/cytology , Stem Cells/enzymology
13.
Mol Ther ; 1(4): 347-57, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10933953

ABSTRACT

Cellular delivery of a replication-conditional herpes simplex virus type 1 (HSV-1) vector provides a means for gene therapy of invasive tumor cells. LacZ-bearing neural precursor cells, which can migrate and differentiate in the brain, were infected with a ribonucleotide reductase-deficient HSV-1 mutant virus (rRp450) that replicates only in dividing cells. Replication of rRp450 in neural precursor cells was blocked prior to implantation into the tumor by growth arrest in late G1 phase through treatment with mimosine. Viral titers in the medium of mimosine-treated, rRp450-infected neural precursor cells were below detection levels 3 days after infection. In culture, after removal of mimosine and passaging, cells resumed growth and replication of rRp450 so that, 7 days later, virus was present in the medium and cell death was evident. Mimosine-treated neural precursor cells injected into established intracerebral CNS-1 gliomas in nude mice migrated extensively throughout the tumor and into the surrounding parenchyma beyond the tumor over 3 days. Mimosine-treated neural precursor cells, infected with rRp450 and injected into intracerebral CNS-1 tumors, also migrated within the tumor with the appearance of foci of HSV-thymidine kinase-positive (TK+) cells, presumably including tumor cells, distributed throughout the tumor and in the surrounding parenchyma over a similar period. This migratory cell delivery method has the potential to expand the range of delivery of HSV-1 vectors to tumor cells in the brain.


Subject(s)
Brain Neoplasms/therapy , Genetic Vectors , Glioma/therapy , Herpesvirus 1, Human/genetics , Neurons/virology , Stem Cells/virology , Animals , Brain Neoplasms/pathology , Cell Movement , Ganciclovir/pharmacology , Genes, Viral , Genetic Therapy/methods , Glioma/pathology , Herpes Simplex Virus Protein Vmw65/genetics , Herpesvirus 1, Human/physiology , Mice , Mice, Nude , Mimosine/pharmacology , Mutation , Neurons/cytology , Neurons/drug effects , Ribonucleotide Reductases/genetics , Stem Cells/cytology , Stem Cells/drug effects , Thymidine Kinase/genetics , Virus Replication/drug effects
14.
Exp Neurol ; 161(1): 67-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683274

ABSTRACT

The ready availability of unlimited quantities of neural stem cells derived from the human brain holds great interest for basic and applied neuroscience, including therapeutic cell replacement and gene transfer following transplantation. We report here the combination of epigenetic and genetic procedures for perpetuating human neural stem cell lines. Thus we tested various culture conditions and genes for those that optimally allow for the continuous, rapid expansion and passaging of human neural stem cells. Among them, v-myc (the p110 gag-myc fusion protein derived from the avian retroviral genome) seems to be the most effective gene; we have also identified a strict requirement for the presence of mitogens (FGF-2 and EGF) in the growth medium, in effect constituting a conditional perpetuality or immortalization. A monoclonal, nestin-positive, human neural stem cell line (HNSC.100) perpetuated in this way divides every 40 h and stops dividing upon mitogen removal, undergoing spontaneous morphological differentiation and upregulating markers of the three fundamental lineages in the CNS (neurons, astrocytes, and oligodendrocytes). HNSC.100 cells therefore retain basic features of epigenetically expanded human neural stem cells. Clonal analysis confirmed the stability, multipotency, and self-renewability of the cell line. Finally, HNSC.100 can be transfected and transduced using a variety of procedures and genes encoding proteins for marking purposes and of therapeutic interest (e.g., human tyrosine hydroxylase I).


Subject(s)
Cell Culture Techniques/methods , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Nerve Tissue Proteins , Neurons/cytology , Stem Cells/cytology , Blotting, Southern , Brain/cytology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Transformed , Central Nervous System/cytology , Gene Expression Regulation, Viral , Genes, gag/physiology , Genes, myc/physiology , Genetic Therapy , Glial Fibrillary Acidic Protein/analysis , Humans , Intermediate Filament Proteins/analysis , Microtubule-Associated Proteins/analysis , Nerve Degeneration , Nerve Regeneration , Nestin , Neurons/chemistry , Neurons/drug effects , Phenotype , Proliferating Cell Nuclear Antigen/analysis , Retroviridae/genetics , Stem Cells/chemistry , Stem Cells/drug effects , Transfection , Tubulin/analysis , Vimentin/analysis
15.
Cell Transplant ; 9(5): 657-67, 2000.
Article in English | MEDLINE | ID: mdl-11144962

ABSTRACT

Cell transplantation into host brain requires a reliable cell marker to trace lineage and location of grafted cells in host tissue. The lacZ gene encodes the bacterial (E. coli) enzyme beta-galactosidase (beta-gal) and is commonly visualized as a blue intracellular precipitate following its incubation with a substrate, "X gal," in an oxidation reaction. LacZ is the "reporter gene" most commonly employed to follow gene expression in neural tissue or to track the fate of transplanted exogenous cells. If the reaction is not performed carefully-with adequate optimization and individualization of various parameters (e.g.. pH, concentration of reagents, addition of chelators, composition of fixatives) and the establishment of various controls--then misleading nonspecific background X-gal positivity can result, leading to the misidentification of cells. Some of this background results from endogenous nonbacterial beta-gal activity in discrete populations of neurons in the mammalian brain; some results from an excessive oxidation reaction. Surprisingly, few articles have empha sized how to recognize and to eliminate these potential confounding artifacts in order to maximize the utility and credibility of this histochemical technique as a cell marker. We briefly review the phenomenon in general, discuss a specific case that illustrates how an insufficiently scrutinized X-gal positivity can be a pitfall in cell transplantation studies, and then provide recommendations for optimizing the specificity and reliability of this histochemical reaction for discerning E. coli beta-gal activity.


Subject(s)
Chromogenic Compounds/metabolism , Galactosides/metabolism , Histocytochemistry/methods , Indoles/metabolism , Neurons/transplantation , beta-Galactosidase/genetics , Animals , Artifacts , Brain/metabolism , Escherichia coli/genetics , Fixatives , Gene Expression , Genes, Reporter , Hydrogen-Ion Concentration , Mice , Neurons/metabolism , Rats , beta-Galactosidase/metabolism
16.
Gene Ther ; 6(11): 1851-66, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10602381

ABSTRACT

Neural stem cell lines represent a homogeneous source of cells for genetic, developmental, and gene transfer and repair studies in the nervous system. Since both gene transfer of neurotrophic factors and cell replacement strategies are of immediate interest for therapeutical purposes, we have generated BDNF-secreting neural stem cell lines and investigated to what extent different endogenous levels of BDNF expression affect in vitro survival, proliferation and differentiation of these cells. Also, we have investigated the in vivo effects of such BDNF gene transfer procedure in the rat neostriatum. Hippocampus- and cerebellum-derived cell lines reacted differently to manipulations aimed at varying their levels of BDNF production. Over-expression of BDNF enhanced survival of both cell types, in a serum-deprivation assay. Conversely, and ruling out unspecific effects, expression of an antisense version of BDNF resulted in compromised survival of cerebellum-derived cells, and in a lethal phenotype in hippocampal progenitors. These data indicate that endogenous BDNF level strongly influences the in vitro survival of these cells. These effects are more pronounced for hippocampus- than for cerebellum-derived progenitors. Hippocampus-derived BDNF overproducers showed no major change in their capacity to differentiate towards a neuronal phenotype in vitro. In contrast, cerebellar progenitors overproducing BDNF did not differentiate into neurons, whereas cells expressing the antisense BDNF construct generated cells with morphological features of neurons and expressing immunological neuronal markers. Taken together, these results provide evidence that BDNF controls both the in vitro survival and differentiation of neural stem cells. After in vivo transplantation of BDNF-overproducing cells to the rat neostriatum, these survived better than the control ones, and induced the expected neurotrophic effects on cholinergic neurons. However, long-term (3 months) administration of BDNF resulted in detrimental effects, at this location. These findings may be of importance for the understanding of brain development, for the design of therapeutic neuro-regenerative strategies, and for cell replacement and gene therapy studies.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebellum/metabolism , Gene Transfer Techniques , Hippocampus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebellum/cytology , Female , Hippocampus/cytology , Immunohistochemistry , Neurons/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells
17.
Virology ; 263(1): 23-9, 1999 Oct 10.
Article in English | MEDLINE | ID: mdl-10544079

ABSTRACT

Following intraperitoneal (IP) inoculation of neonatal mice, the polytropic recombinant murine leukemia virus (MuLV), Fr98, induces a severe brain disease characterized by ataxia, seizures and death. In contrast, no apparent clinical neurological disease is seen after IP infection with Fr54, a polytropic MuLV differing from Fr98 in its envelope gene sequences. In the brain both Fr98 and Fr54 infect primarily capillary endothelial cells and microglia. However, the level of microglial infection by Fr98 is twofold higher than by Fr54, which might account for the difference in neurovirulence. In the present study, in order to test directly whether an increase in the number of microglia infected by Fr54 would be sufficient to induce clinical disease, we attempted to increase the level of Fr54 in the brain by changing the route of infection. After intraventricular inoculation with Fr54-infected neural stem cells (clone C17.2), a well-established vehicle for delivery of viruses and genes to the brain, mice became ataxic and died 4 weeks postinfection. In these mice induction of brain disease was correlated with a higher level of viral antigen in the cerebrum and an increase in the number of infected microglial cells in all brain regions examined compared with mice inoculated IP. In contrast, mice inoculated with neural stem cells infected with an ecotropic nonneurovirulent murine leukemia virus, FB29, developed no clinical disease in spite of evidence for widespread infection of microglia in brain. Since the main differences between Fr54 and FB29 are in the SU (gp70) region of the envelope gene, this region is most likely to account for the differences in induction of CNS disease seen in the current experiments.


Subject(s)
Brain Diseases/virology , Brain/virology , Leukemia Virus, Murine/pathogenicity , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Brain/pathology , Brain Diseases/pathology , Capsid/metabolism , Leukemia Virus, Murine/isolation & purification , Mice , Mice, Inbred Strains , Microglia/virology , Neurons/cytology , Neurons/virology , Recombinant Proteins , Retroviridae Infections/pathology , Stem Cells/cytology , Stem Cells/virology , Tumor Virus Infections/pathology , Viral Envelope Proteins/metabolism , Viral Load , Virulence
18.
J Neurosci ; 19(20): 8954-65, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516314

ABSTRACT

Activation of the Sonic hedgehog (Shh) signal transduction pathway is essential for normal pattern formation and cellular differentiation in the developing CNS. However, it is also thought to be etiological in primitive neuroectodermal tumors. We adapted GAL4/UAS methodology to ectopically express full-length Shh in the dorsal neural tube of transgenic mouse embryos commencing at 10 d postcoitum (dpc), beyond the period of primary dorsal-ventral pattern formation and floorplate induction. Expression of Shh was maintained until birth, permitting us to investigate effects of ongoing exposure to Shh on CNS precursors in vivo. Proliferative rates of spinal cord precursors were twice that of wild-type littermates at 12.5 dpc. In contrast, at late fetal stages (18.5 dpc), cells that were Shh-responsive but postmitotic were present in persistent structures reminiscent of the ventricular zone germinal matrix. This tissue remained blocked in an undifferentiated state. These results indicate that cellular competence restricts the proliferative response to Shh in vivo and provide evidence that proliferation and differentiation can be regulated separately in precursor cells of the spinal cord. Thus, Hedgehog signaling may contribute to CNS tumorigenesis by directly enhancing proliferation and preventing neural differentiation in selected precursor cells.


Subject(s)
Central Nervous System/cytology , Proteins/physiology , Stem Cells/cytology , Trans-Activators , Zebrafish Proteins , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cerebral Ventricles/embryology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/physiology , Embryonic and Fetal Development , Gene Expression , Gene Targeting , Hedgehog Proteins , Mice , Mice, Transgenic/genetics , Neurons/pathology , Oligodendroglia/pathology , Proteins/genetics , Proto-Oncogene Proteins/genetics , Spinal Cord/embryology , Wnt Proteins
19.
J Neurotrauma ; 16(8): 675-87, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10511240

ABSTRACT

Multipotent neural progenitors and stem cells may integrate appropriately into the developing and degenerating central nervous system. They may also be effective in the replacement of genes, cells, and nondiffusible factors in either a widespread or a more circumscribed manner, depending on the therapeutic demands of the clinical situation. In addition, they may be uniquely responsive to some types of neurodegenerative conditions. We believe that these various appealing capabilities are the normal expression of basic biologic properties and attributes of a stem cell. The therapeutic utility of some of those properties is illustrated in this review of ongoing work in our laboratory, particularly with regard to spinal dysfunction. In these examples, we believe we have tapped into a mechanism that underlies a remarkable degree of natural plasticity programmed into the nervous system at the cellular level, and we have now exploited those properties for therapeutic ends.


Subject(s)
Brain Injuries/surgery , Spinal Cord Injuries/surgery , Stem Cell Transplantation , Animals , Brain Tissue Transplantation , Clone Cells , Fetal Tissue Transplantation , Mice
20.
Exp Neurol ; 158(1): 9-26, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10448414

ABSTRACT

Neural stem cells have been shown to participate in the repair of experimental CNS disorders. To examine their potential in spinal cord repair, we used retroviral vectors to genetically modify a clone of neural stem cells, C17, to overproduce neurotrophin-3 (NT-3). The cells were infected with a retrovirus construct containing the NT-3.IRES.lacZ/neo sequence and cloned by limiting dilution and selection for lacZ expression. We studied the characteristics of the modified neural stem cells in vitro and after transplantation into the intact spinal cord of immunosuppressed adult rats. Our results show that: (i) most of the genetically modified cells express both NT-3 and lacZ genes with a high coexpression ratio in vitro and after transplantation; and (ii) large numbers of the xenografted cells survive in the spinal cord of adult rats for at least 2 months, differentiate into neuronal and glial phenotypes, and migrate for long distances. We conclude that genetically modified neural stem cells, acting as a source of neurotrophic factors, have the potential to participate in spinal cord repair.


Subject(s)
Nerve Growth Factors/biosynthesis , Neurons/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Animals , Cell Differentiation , Cell Survival/physiology , Cells, Cultured , Clone Cells/physiology , Female , Gene Expression/genetics , Genetic Markers , Injections, Spinal , Neurons/metabolism , Neurotrophin 3 , Rats , Rats, Sprague-Dawley , Recombination, Genetic/genetics , Retroviridae/genetics , Stem Cells/metabolism , Transfection/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL