201
|
Abstract
Retinal stem cells (RSCs) are multipotent central nervous system (CNS) precursors that give rise to the retina during the course of development. RSCs are present in the embryonic eyecup of all vertebrate species and remain active in lower vertebrates throughout life. Mammals, however, exhibit little RSC activity in adulthood and thus little capacity for retinal growth or regeneration. Because CNS precursors can now be isolated from immature and mature mammals and expanded ex vivo, it is possible to study these cells in culture as well as following transplantation to the diseased retina. Such experiments have revealed a wealth of unanticipated findings, both in terms of the instructive cues present in the mature mammalian retina as well as the ability of grafted CNS precursors to respond to them. This review examines current knowledge regarding RSCs, together with other CNS precursors, from the perspective of investigators who wish to isolate, propagate, genetically modify, and transplant these cells as a regenerative strategy with application to retinal disease.
Collapse
Affiliation(s)
- Henry Klassen
- Stem Cell Research, Children's Hospital of Orange County, Orange, CA 92868, USA
| | | | | |
Collapse
|
202
|
Nonaka M, Yoshikawa M, Nishimura F, Yokota H, Kimura H, Hirabayashi H, Nakase H, Ishizaka S, Wanaka A, Sakaki T. Intraventricular transplantation of embryonic stem cell-derived neural stem cells in intracerebral hemorrhage rats. Neurol Res 2004; 26:265-72. [PMID: 15142318 DOI: 10.1179/016164104225014049] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In the present study, we attempted to explore cell transplantation therapy for intracerebral hemorrhage (ICH) using embryonic stem (ES) cells. Collagenase-induced ICH rats were used as model animals. Mouse ES cells were differentiated into nestin-positive neural stem cells in vitro by alltrans retinoic acid (ATRA). ATRA-treated ES cells (10(5)) were transplanted into the lateral ventricle in the hemisphere contralateral to the hemorrhage 7 days after collagenase infusion. Twenty-eight days after transplantation, ES-derived neurons and astrocytes were observed around the hematoma cavities of the brain in all of the ten rats receiving grafts. Graft-derived neurons were found in the subependymal area of the lateral ventricle as cellular nodules. Although one of the ten rats receiving grafts showed uncontrolled growth of astroglia derived from the ES cells, intraventricular transplantation of ATRA-treated ES cells is an effective delivery system of neuronal lineage-committed progenitor cells toward the site of ICH.
Collapse
Affiliation(s)
- Masahiro Nonaka
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Jung CG, Hida H, Nakahira K, Ikenaka K, Kim HJ, Nishino H. Pleiotrophin mRNA is highly expressed in neural stem (progenitor) cells of mouse ventral mesencephalon and the product promotes production of dopaminergic neurons from embryonic stem cell-derived nestin-positive cells. FASEB J 2004; 18:1237-9. [PMID: 15180956 DOI: 10.1096/fj.03-0927fje] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neural stem cells are promising candidates for donor cells in neural transplantation. However, the mechanism by which neural stem cells differentiate into neurons is not well understood. In the present study, a serial analysis of gene expression (SAGE) was carried out to generate a gene file of neural stem (progenitor) cells from the mouse ventral mesencephalon. Among the 15,815 tags investigated, the mRNA of the housekeeping genes (elongation factor 1-alpha, ATPase subunit 6, GAPDH, actin), laminin receptor 1, HSP 70, pleiotrophin, and nestin were highly expressed. Because pleiotrophin (PTN) exhibits mitogenic and trophic effects on neural development and exhibits trophic effects on survival of dopaminergic (DAergic) neurons, we investigated the role of PTN in neurogenesis, especially to DAergic neurons. Here, we show that PTN increased the production of tyrosine hydroxylase (TH)-positive neurons from embryonic stem (ES) cell-derived nestin-positive cells. The expression of Nurr1 mRNA was enhanced by PTN. L-dopa in the culture medium was increased by PTN. This effect was as strong as with sonic hedgehog. Data suggest that PTN mRNA is highly expressed in neural stem (progenitor) cells of mouse ventral mesencephalon, and PTN promotes the production of DAergic neurons from ES cell-derived nestin-positive cells.
Collapse
MESH Headings
- Animals
- Biomarkers
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Carrier Proteins/pharmacology
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Cytokines/biosynthesis
- Cytokines/genetics
- Cytokines/pharmacology
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Dopamine/analysis
- Expressed Sequence Tags
- Gene Expression Profiling
- Gene Expression Regulation, Developmental/drug effects
- Intermediate Filament Proteins/analysis
- Levodopa/biosynthesis
- Mesencephalon/cytology
- Mesencephalon/embryology
- Mice
- Mice, Inbred ICR
- Nerve Tissue Proteins/analysis
- Nestin
- Neurons/chemistry
- Neurons/cytology
- Nuclear Receptor Subfamily 4, Group A, Member 2
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Stem Cells/drug effects
- Stem Cells/metabolism
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Tyrosine 3-Monooxygenase/analysis
Collapse
Affiliation(s)
- Cha-Gyun Jung
- Department of Neuro-Physiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Ku, Nagoya 467-8601, Japan.
| | | | | | | | | | | |
Collapse
|
204
|
Martino G. How the brain repairs itself: new therapeutic strategies in inflammatory and degenerative CNS disorders. Lancet Neurol 2004; 3:372-8. [PMID: 15157853 DOI: 10.1016/s1474-4422(04)00771-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In the early 20th century, seminal work by Tello and Cajal showed that the CNS has the ability to regenerate itself after injury. In the most recent years, this pivotal observation has been rejuvenated by detailed in vitro and in vivo evidence supporting the idea of an innate self-maintenance programme to sustain brain homoeostasis and repair. These observations support the idea that chronic inflammatory and degenerative disorders of the brain might result from defective repair mechanisms rather than uncontrollable pathogenetic events. Investigation of the molecular and cellular events sustaining intrinsic brain-repair mechanisms and a better understanding of why they fail over time in chronic disorders might, therefore, provide an attractive conceptual framework within which to develop new and efficacious therapies for neurological diseases.
Collapse
Affiliation(s)
- Gianvito Martino
- Neuroimmunology Unit (DIBIT) and Department of Neurology, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
205
|
Horiguchi S, Takahashi J, Kishi Y, Morizane A, Okamoto Y, Koyanagi M, Tsuji M, Tashiro K, Honjo T, Fujii S, Hashimoto N. Neural precursor cells derived from human embryonic brain retain regional specificity. J Neurosci Res 2004; 75:817-24. [PMID: 14994342 DOI: 10.1002/jnr.20046] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recent studies have revealed that neural precursor cells can be expanded not only from the subventricular zone and hippocampus but also from other regions of the human embryonic brain. To determine the regional differences of these precursor cells, we divided the brain of a 9-week-old human embryo into four parts, i.e., telencephalon, diencephalon, mesencephalon, and rhombencephalon. All cultures of the tissues yielded neurospheres, and these spheres gave rise to neurons, astrocytes, and oligodendrocytes. An analysis of clonal populations revealed that these precursor cells were multipotent, and two region-specific differences in neural precursor cells were revealed: 1) The precursor cells from the rostral part of the brain tended to proliferate faster than those from the caudal part, and 2) the precursor cells from the diencephalon and mesencephalon gave rise to more tyrosine hydoxylase (TH)-positive neurons than those from the telencephalon and rhombencephalon. When 50-day-cultured spheres were caused to differentiate, the percentage of TH-positive cells per total cell population was 1.2% for diencephalic and mesencephalic precursors, whereas it was 0.4% for telencephalic and rhombencephalic ones. Furthermore, the TH-positive cells from diencephalic and mesencephalic precursors were large, multipolar, and gamma-aminobutyric acid (GABA)-negative, which suggested that these cells were midbrain dopaminergic neurons. In contrast, TH-positive cells from telencephalic and rhombencephalic precursors were small, bipolar, and GABA-positive. These results suggest that human neural precursor cells might have the potential to differentiate into a variety of cells but retain regional specificity.
Collapse
Affiliation(s)
- Satoshi Horiguchi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Glioma Tropic Neural Stem Cells Consist of Astrocytic Precursors and Their Migratory Capacity Is Mediated by CXCR4. Neoplasia 2004. [DOI: 10.1593/neo.03427] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
207
|
LEPORE ANGELOC, HAN STEVENS, TYLER-POLSZ CARLAJ, CAI JINGLI, RAO MAHENDRAS, FISCHER ITZHAK. Differential fate of multipotent and lineage-restricted neural precursors following transplantation into the adult CNS. NEURON GLIA BIOLOGY 2004; 1:113-26. [PMID: 16520830 PMCID: PMC1389711 DOI: 10.1017/s1740925x04000213] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multiple classes of precursor cells have been isolated and characterized from the developing spinal cord including multipotent neuroepithelial (NEP) stem cells and lineage-restricted precursors for neurons (NRPs) and glia (GRPs). We have compared the survival, differentiation and integration of multipotent NEP cells with lineage-restricted NRPs and GRPs using cells isolated from transgenic rats that express the human placental alkaline phosphatase gene. Our results demonstrate that grafted NEP cells survive poorly, with no cells observed 3 days after transplant in the adult hippocampus, striatum and spinal cord, indicating that most CNS regions are not compatible with transplants of multipotent cells derived from fetal CNS. By contrast, at 3 weeks and 5 weeks post-engraftment, lineage-restricted precursors showed selective migration along white-matter tracts and robust survival in all three CNS regions. The grafted precursors expressed the mature neuronal markers NeuN and MAP2, the astrocytic marker GFAP, the oligodendrocytic markers RIP, NG2 and Sox-10, and the synaptic marker synaptophysin. Similar behavior was observed when these precursors were transplanted into the injured spinal cord. Predifferentiated, multipotent NEP cells also survive and integrate, which indicates that lineage-restricted CNS precursors are well suited for transplantation into the adult CNS and provide a promising cellular replacement candidate.
Collapse
Affiliation(s)
- ANGELO C. LEPORE
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - STEVEN S.W. HAN
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - CARLA J. TYLER-POLSZ
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - JINGLI CAI
- Laboratory of Neuroscience, NIA, Baltimore, MD, USA
| | | | - ITZHAK FISCHER
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
208
|
Liang P, Zhao S, Kawamoto K, Jin L, Liu E. Neuronal and glial differentiation following culture of the human embryonic cortical stem cells. Hum Cell 2004; 16:151-6. [PMID: 15005246 DOI: 10.1111/j.1749-0774.2003.tb00147.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To set up long-term in vitro culture system of the human neural stem cells (hNSC) and to study their biological properties. METHODS Human fetuses aged about 20 weeks following spontaneous abortion were adopted. A serum-free medium containing basic fibroblast growth factor and epidermal growth factor was used to make the hNSCs divide continuously in the culture. The growth curve of continually passaged cells was examined. The effects of long-term culture on the cell cycle, cell differentiation were analyzed. The cell cycles of these cells were analyzed using flow cytometry. RESULTS The cells from the human embryonic cortical tissue could be maintained and propagated in the presence of growth factors. Neurospheres were generated continually. Only one month after the primary culture, the precursors could be effectively discarded. The cells could be cultured for ten months. The cells had an exponential, consistent growth. The cell cycle analysis indicated that the hNSCs maintained remarkable proliferation. Upon differentiation, the hNSCs gave rise to mature cells. The multi-lineage potential of differentiation after different passages remained unchanged. CONCLUSION The hNSCs isolated from the human embryonic tissues retained their biological features after long-term culture in vitro.
Collapse
Affiliation(s)
- Peng Liang
- Department of Neurosurgery, First Medical College of Harbin Medical University, Harbin 150001, PR China.
| | | | | | | | | |
Collapse
|
209
|
Le Belle JE, Caldwell MA, Svendsen CN. Improving the survival of human CNS precursor-derived neurons after transplantation. J Neurosci Res 2004; 76:174-83. [PMID: 15048915 DOI: 10.1002/jnr.20035] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have examined the effects of predifferentiation and energy substrate deprivation on long-term expanded human neural precursor cells (HNPCs). The pre-differentiation of HNPC cultures produced large numbers of neurons (>60%) and mature glial cells capable of generating glycogen stores that protected the neuronal population from experimental metabolic stress. When predifferentiated HNPCs were transplanted into intact adult rat hippocampus, fewer cells survived compared to undifferentiated HNPC transplants. This cell death was completely attenuated, however, when predifferentiated HNPC cultures were pretreated to boost glial energy stores and resulted in greatly increased neuronal survival in vivo. The transplanted cells primarily engrafted within the granular layer of the dentate gyrus, where a large proportion of the predifferentiated HNPCs co-expressed neuronal markers whereas most HNPCs outside of the neuronal layer did not, indicating that the predifferentiated cells remained capable of responding to local cues in the adult brain. Undifferentiated HNPCs migrated more widely in the brain after grafting than did the predifferentiated cells, which generally remained within the hippocampus.
Collapse
Affiliation(s)
- J E Le Belle
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom.
| | | | | |
Collapse
|
210
|
Sayles M, Jain M, Barker RA. The cellular repair of the brain in Parkinson's disease—past, present and future. Transpl Immunol 2004; 12:321-42. [PMID: 15157925 DOI: 10.1016/j.trim.2003.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Damage to the central nervous system was once considered irreparable. However, there is now growing optimism that neural transplant therapies may one day enable complete circuit reconstruction and thus functional benefit for patients with neurodegenerative conditions such as Parkinson's disease (PD), and perhaps even those with more widespread damage such as stroke patients. Indeed, since the late 1980s hundreds of patients with Parkinson's disease have received allografts of dopamine-rich embryonic human neural tissue. The grafted tissue has been shown to survive and ameliorate many of the symptoms of the disease, both in the clinical setting and in animal models of the disease. However, practical problems associated with tissue procurement and storage, and ethical concerns over using aborted human fetal tissue have fuelled a search for alternative sources of suitable material for grafting. In particular, stem cells and xenogeneic embryonic dopamine-rich neural tissue are being explored, both of which bring their own practical and ethical dilemmas. Here we review the progress made in neural transplantation, both in the laboratory and in the clinic with particular attention to the development of stem cell and xenogeneic tissue based therapy.
Collapse
Affiliation(s)
- Mark Sayles
- Cambridge Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK
| | | | | |
Collapse
|
211
|
Burnstein RM, Foltynie T, He X, Menon DK, Svendsen CN, Caldwell MA. Differentiation and migration of long term expanded human neural progenitors in a partial lesion model of Parkinson’s disease. Int J Biochem Cell Biol 2004; 36:702-13. [PMID: 15010333 DOI: 10.1016/j.biocel.2003.11.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Revised: 10/27/2003] [Accepted: 11/03/2003] [Indexed: 10/26/2022]
Abstract
Human neural progenitor cells (HNPCs) can be expanded in large numbers for significant periods of time to provide a reliable source of neural cells for transplantation in neurodegenerative disorders such as Parkinson's disease (PD). In the present study, HNPCs isolated from embryonic cortex were expanded as neurospheres in cell culture for 10 months. Just prior to transplantation, a proportion of the HNPCs were treated in a "predifferentiation" protocol in combination with the neurotropic factor NT4, in order to yield significant numbers of neurons. For transplantation, either undifferentiated HNPCs, or predifferentiated HNPCs were transplanted into the substantia nigra of a rat model of Parkinson's disease. At 12 weeks, there was good survival with proliferation of transplanted HNPCs occurring after transplantation but ceasing before the animals were sacrificed. Transplants of predifferentiated cells contained a higher proportion of neurons. The presence of a lesion in the striatum had a significant influence on the migration of transplanted cells from the substantia nigra into the striatum. There was no significant behavioural recovery or effect of transplanted HNPCs on the loss of dopaminergic cells from the host brain. In conclusion, HNPCs may provide a source of cells for use in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Rowan M Burnstein
- Cambridge Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 2PY, UK
| | | | | | | | | | | |
Collapse
|
212
|
Kokuzawa J, Yoshimura S, Kitajima H, Shinoda J, Kaku Y, Iwama T, Morishita R, Shimazaki T, Okano H, Kunisada T, Sakai N. Hepatocyte growth factor promotes proliferation and neuronal differentiation of neural stem cells from mouse embryos. Mol Cell Neurosci 2004; 24:190-7. [PMID: 14550779 DOI: 10.1016/s1044-7431(03)00160-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hepatocyte growth factor (HGF), originally cloned as a hepatocyte mitogen, has recently been reported to exhibit neurotrophic activity in addition to being expressed in different parts of the nervous system. At present, the effects of HGF on neural stem cells (NSCs) are not known. In this study, we first report the promoting effect of HGF on the proliferation of neurospheres and neuronal differentiation of NSCs. Medium containing only HGF was capable of inducing neurosphere formation. Addition of HGF to medium containing fibroblast growth factor 2 or epidermal growth factor increased both the size and number of newly formed neurospheres. More neurons were also obtained when HGF was added in differentiation medium. In contrast, neurosphere numbers were reduced after repeated subculture by mechanical dissociation, suggesting that HGF-formed neurospheres comprised predominantly progenitor cells committed to neuronal or glial lines. Together, these results suggest that HGF promotes proliferation of neurospheres and neuronal differentiation of NSCs derived from mouse embyos.
Collapse
Affiliation(s)
- Jouji Kokuzawa
- Department of Neurosurgery, Division of Neuroscience, Gifu University School of Medicine, 40 Tsukasa-machi, Gifu 500-8705, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Jin K, Mao XO, Batteur S, Sun Y, Greenberg DA. Induction of neuronal markers in bone marrow cells: differential effects of growth factors and patterns of intracellular expression. Exp Neurol 2004; 184:78-89. [PMID: 14637082 DOI: 10.1016/s0014-4886(03)00133-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bone marrow cells (BMC) can be induced to express neuronal phenotypic features in vitro, but the extent to which they can transdifferentiate to mature, functional neurons is uncertain. We examined the effects of different growth factors and combinations thereof on the expression of neuronal marker proteins in cultures of BMC enriched in marrow stromal cells. Patterns of neuronal marker expression varied depending on the growth factor or factors to which BMC cultures were exposed. Cultures treated for up to 5 weeks with epidermal growth factor, fibroblast growth factor-2, retinoic acid, and nerve growth factor displayed neuron-like cellular processes and expressed neuronal markers, including the neuronal nuclear antigen NeuN, microtubule-associated protein 2, tau, synaptophysin, alpha(1A) and alpha(1B) calcium channel subunits, NR2A glutamate receptor subunits, and gamma-aminobutyric acid. However, the intracellular distribution of these markers was distinct from their usual distribution in mature neurons. We conclude that a variety of growth factors can drive BMC toward a neuronal phenotype or phenotypes, but that morphological neuronal features and the ectopic expression of neuronal proteins and neurotransmitters may not equate with the ability to execute normal neuronal functions.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | |
Collapse
|
214
|
Yang M, Donaldson AE, Marshall CE, Shen J, Iacovitti L. Studies on the differentiation of dopaminergic traits in human neural progenitor cells in vitro and in vivo. Cell Transplant 2004; 13:535-47. [PMID: 15565866 PMCID: PMC1949040 DOI: 10.3727/000000004783983729] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The development of cell replacement therapies for the treatment of neurodegenerative disorders such as Parkinson's disease (PD) may depend upon the successful differentiation of human neural stem/progenitor cells into dopamine (DA) neurons. We show here that primary human neural progenitors (HNPs) can be expanded and maintained in culture both as neurospheres (NSPs) and attached monolayers where they develop into neurons and glia. When transplanted into the 6-hydroxydopamine-lesioned rat striatum, undifferentiated NSPs survive longer (60% graft survival at 8-16 weeks vs. 30% graft survival at 8-13 weeks) and migrate farther than their attached counterparts. While both NSP and attached cells continue to express neuronal traits after transplantation, the spontaneous expression of differentiated transmitter-related traits is not observed in either cell type. However, following predifferentiation in culture using a previously described cocktail of reagents, approximately 25% of HNPs can permanently express the DA enzyme tyrosine hydroxylase (TH), even following replating and removal of the DA differentiation cocktail. When these predifferentiated HNPs are transplanted into the brain, however, TH staining is not observed, either because expression is lost or TH-expressing cells preferentially die. Consistent with the latter view is a decrease in total cell survival and migration, and an enhanced glial response in these grafts. In contrast, we found that the overall survival of HNPs is improved when cells engraft near blood vessels or CSF compartments or when they are placed into an intact unlesioned brain, suggesting that there are factors, as yet unidentified, that can better support the development of engrafted HNPs.
Collapse
Affiliation(s)
- Ming Yang
- Farber Institute for Neurosciences, Department of Neurology, Thomas Jefferson University Medical College, 900 Walnut Street, Philadelphia, PA 19107
| | - Angela E. Donaldson
- Farber Institute for Neurosciences, Department of Neurology, Thomas Jefferson University Medical College, 900 Walnut Street, Philadelphia, PA 19107
| | - Cheryl E. Marshall
- Farber Institute for Neurosciences, Department of Neurology, Thomas Jefferson University Medical College, 900 Walnut Street, Philadelphia, PA 19107
| | - James Shen
- ScienCell Research Laboratories, 4050 Sorrento Valley Boulevard, San Diego, CA 92121
| | - Lorraine Iacovitti
- Farber Institute for Neurosciences, Department of Neurology, Thomas Jefferson University Medical College, 900 Walnut Street, Philadelphia, PA 19107
| |
Collapse
|
215
|
Palomo T, Archer T, Beninger RJ, Kostrzewa RM. Gene-environment interplay in neurogenesis and neurodegeneration. Neurotox Res 2004; 6:415-34. [PMID: 15639777 DOI: 10.1007/bf03033279] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Factors associated with predisposition and vulnerability to neurodegenerative disorders may be described usefully within the context of gene-environment interplay. There are many identified genetic determinants for so-called genetic disorders, and it is possible to duplicate many elements of recognized human neurodegenerative disorders in either knock-in or knock-out mice. However, there are similarly, many identifiable environmental influences on outcomes of the genetic defects; and the course of a progressive neurodegenerative disorder can be greatly modified by environmental elements. Constituent cellular defense mechanisms responsive to the challenge of increased reactive oxygen species represent only one crossroad whereby environment can influence genetic predisposition. In this paper we highlight some of the major neurodegenerative disorders and discuss possible links of gene-environment interplay. The process of adult neurogenesis in brain is also presented as an additional element that influences gene-environment interplay. And the so-called priming processes (i.e., production of receptor supersensitization by repeated drug dosing), is introduced as yet another process that influences how genes and environment ultimately and co-dependently govern behavioral ontogeny and outcome. In studies attributing the influence of genetic alteration on behavioral phenotypy, it is essential to carefully control environmental influences.
Collapse
Affiliation(s)
- Tomás Palomo
- Servicio Psiquiátrico, Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
| | | | | | | |
Collapse
|
216
|
Aarum J, Sandberg K, Haeberlein SLB, Persson MAA. Migration and differentiation of neural precursor cells can be directed by microglia. Proc Natl Acad Sci U S A 2003; 100:15983-8. [PMID: 14668448 PMCID: PMC307679 DOI: 10.1073/pnas.2237050100] [Citation(s) in RCA: 353] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2003] [Indexed: 11/18/2022] Open
Abstract
Recent reports have supported the existence of neural stem cells in the adult mammalian CNS. Important features of such cells are self-renewal and multipotency, i.e., they can give rise to neurons, astrocytes, and oligodendrocytes and thus in principle replace lost cells in the CNS. Observations in several animal models of CNS diseases have shown that by unknown mechanisms endogenous as well as exogenous precursor cells preferentially migrate to damaged areas. Microglia are immunoreactive cells of nonneural lineage resident in the CNS. After injury to the CNS, microglia are rapidly activated and found concentrated at the sites of injury. In the present article we show, in two different assays, that soluble factors released from mouse microglial cells direct the migration of neural CNS precursor cells. We also provide evidence that microglia have the capacity to influence the differentiation of both adult and embryonic neural precursor cells toward a neuronal phenotype. Given that an invariant feature of pathological processes in CNS is the activation of microglia, these results indicate an important and unique role for microglia in directing the replacement of damaged or lost cells in the CNS.
Collapse
Affiliation(s)
- Johan Aarum
- Karolinska Institutet, Department of Medicine and Center for Molecular Medicine, Karolinska Hospital, 171 76 Stockholm, Sweden
| | | | | | | |
Collapse
|
217
|
Jin HK, Schuchman EH. Ex vivo gene therapy using bone marrow-derived cells: combined effects of intracerebral and intravenous transplantation in a mouse model of niemann–pick disease. Mol Ther 2003; 8:876-85. [PMID: 14664789 DOI: 10.1016/j.ymthe.2003.07.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Normal murine bone marrow cells were transduced with a retroviral vector to overexpress and release human acid sphingomyelinase (ASM). The transduced cells were then transplanted intravenously into 3-day-old, irradiated ASM-deficient mice, a model of human Niemann-Pick disease (NPD). At 4 weeks, engrafted mice received intracerebral injections of mesenchymal stem cells obtained from the original, transduced bone marrow. By 16 weeks, most of the treated NPD mice had near-normal levels of ASM activity in their tissues, including the brain; dramatically improved histology; and marked reductions in sphingomyelin. Cerebellar function also was normal, and the number of Purkinje cells was > 80% of normal. Remarkably, in certain regions of the cerebellum many of the surviving Purkinje cells expressed human ASM RNA, suggesting that either they were donor-derived or that the transplanted bone marrow cells had fused with existing Purkinje cells. However, despite these positive results, by 24 weeks the ASM activities were dramatically reduced and cerebellar function began to decline, coincident with the detection of anti-human ASM antibodies in the plasma. We conclude that this gene therapy procedure might be useful in Type A NPD and other neurological lysosomal storage disorders, particularly since it is an approach that could be beneficial for both the neurological and the visceral organ features of these diseases.
Collapse
Affiliation(s)
- Hee-Kyung Jin
- Department of Human Genetics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
218
|
Limke TL, Rao MS. Neural Stem Cell Therapy in the Aging Brain: Pitfalls and Possibilities. ACTA ACUST UNITED AC 2003; 12:615-23. [PMID: 14977471 DOI: 10.1089/15258160360732641] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
As aging progresses, there is a decline in the brain's capacity to produce new neurons in the two neurogenic regions, the subventricular zone surrounding the lateral ventricles and the subgranular layer of the hippocampal dentate gyrus. The underlying cause of the declining neurogenesis is unknown, but is presumably related to age-related changes that occur during normal aging of the brain. It is exacerbated by age-related neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Stem cell-based therapy to replace lost and/or damaged cells in the aging brain is currently the focus of intense research. The two most promising approaches involve transplantation of exogenous tissue and promoting proliferation of endogenous cells. However, age-related changes in the brain environment, including elevated oxidative stress and accumulation of protein and lipid by-products, present several unique challenges that must be addressed before cell-based therapy can be used as a viable option. Although progress has been made toward replacement of lost cells and recovery of lost function, there are fundamental issues that need to be addressed for stem cell therapy to be successful in the aging brain. In this review, we focus on recent progresses made toward understand the biology of neural stem cells in the aging brain, as well as progress toward using stem cells to replace cells lost during disease.
Collapse
Affiliation(s)
- Tobi L Limke
- Laboratory of Neurosciences, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | | |
Collapse
|
219
|
Abstract
Injury or degeneration of the vertebrate central nervous system often disrupts neuronal circuitry that is built by projection neurons during early embryonic life. Repair of neural network through regeneration of these early-born projection neurons in adult life often fails since stem cells residing in the adult brain are generally programmed to give rise to late-born interneurons. Thus, exogenous cells are needed to rebuild the neural circuitry. Nevertheless, cell replacement in the brain remains a challenging goal because of the lack of safe and effective donor cells, as well as difficulty in remodeling the nonneurogenic adult CNS environment. Here I will concentrate on the donor side and discuss how recent advancement in stem cell technology offers hope for transplant therapy, with a focus on the potentials and hurdles of human embryonic stem cells as a sustainable source.
Collapse
Affiliation(s)
- Su-Chun Zhang
- Department of Anatomy, School of Medicine, Waisman Center, WiCell Institute, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
220
|
Fodor WL. Tissue engineering and cell based therapies, from the bench to the clinic: the potential to replace, repair and regenerate. Reprod Biol Endocrinol 2003; 1:102. [PMID: 14614775 PMCID: PMC293418 DOI: 10.1186/1477-7827-1-102] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2003] [Accepted: 11/13/2003] [Indexed: 01/22/2023] Open
Abstract
The field of Regenerative Biology as it applies to Regenerative Medicine is an increasingly expanding area of research with hopes of providing therapeutic treatments for diseases and/or injuries that conventional medicines and even new biologic drug therapies cannot effectively treat. Extensive research in the area of Regenerative Medicine is focused on the development of cells, tissues and organs for the purpose of restoring function through transplantation. The general belief is that replacement, repair and restoration of function is best accomplished by cells, tissues or organs that can perform the appropriate physiologic/metabolic duties better than any mechanical device, recombinant protein therapeutic or chemical compound. Several strategies are currently being investigated and include, cell therapies derived from autologous primary cell isolates, cell therapies derived from established cell lines, cell therapies derived from a variety of stem cells, including bone marrow/mesenchymal stem cells, cord blood stem cells, embryonic stem cells, as well as cells tissues and organs from genetically modified animals. This mini-review is not meant to be exhaustive, but aims to highlight clinical applications for the four areas of research listed above and will address a few key advances and a few of the hurdles yet to be overcome as the technology and science improve the likelihood that Regenerative Medicine will become clinically routine.
Collapse
Affiliation(s)
- William L Fodor
- Center for Regenerative Biology and Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06250-4243, USA.
| |
Collapse
|
221
|
Abstract
Studies in animal models have suggested a role for stem cells in repair and regeneration of the nervous system. Human equivalents of stem and precursor cells have been isolated and their efficacy is being evaluated in rodent and primate models. Difficulties exist in translating results of these preclinical models to therapy in humans. Evolutionary differences among rodents, primates, and humans; fundamental differences in the anatomy and physiology; differences in immune responses in xenotransplant models; the paucity of good transplant models of chronic disease; and allelic variability in the cells themselves make any study evaluating the efficacy of cells in transplant models difficult to interpret. As no better alternatives to testing in animals exist, we suggest that at this early stage a considered step-by-step approach to testing and comparison of different transplant strategies in isolation will prepare us better for clinical trials than simple evaluation of functional outcomes in various models of disease. We emphasize that we do not recommend delaying or abandoning clinical trials; rather, we suggest that one anticipate failures and design experiments and data collection such that we learn from these failures to ensure future success in as rapid a time frame as possible.
Collapse
Affiliation(s)
- Irene Ginis
- Gerontology Research Center, Stem Cell Biology Unit/Laboratory of Neuroscience, National Institute on Aging, National Institutes of Health, 5600 Nathan Shock Drive, Room 4E02, Baltimore, MD 21224, USA
| | | |
Collapse
|
222
|
Mochizuki H, Mizuno Y. Gene therapy for Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2003:205-13. [PMID: 12946058 DOI: 10.1007/978-3-7091-0643-3_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
We review recent progress in gene therapy utilizing experimental parkinsonian models including our data. Investigation of ex vivo gene therapy for Parkinson's disease (PD) is to provide L-dopa by transplantation of genetically modified cells into the striatum. Recently, neuronal progenitor cells (NPC) are recognized as the most appropriate target population for such genetic and cellular therapy of PD. We have developed modified pseudo-typed retrovirus production system. Using this gene transfer system, it is easy and efficient to introduce the gene into NPC because high titer virus vector is easily obtained. For the in vivo gene therapy, adeno-associated virus (AAV) vector is best virus vector because it is easy to introduce gene into neurons without inflammatory reaction. We established in vivo models of the inhibition of the caspase-cascade by overexpression of apoptotic protease activating factor-1-dominant negative inhibitor (Apaf-1-DN) using AAV vector. We showed that Apaf-1-DN delivery using an AAV vector system could prevent nigrostriatal degeneration in MPTP mice, suggesting that it might be an anti-mitochondrial apoptotic gene therapy for PD.
Collapse
Affiliation(s)
- H Mochizuki
- Department of Neurology, Juntendo University School Medicine, Tokyo, Japan.
| | | |
Collapse
|
223
|
Storch A, Lester HA, Boehm BO, Schwarz J. Functional characterization of dopaminergic neurons derived from rodent mesencephalic progenitor cells. J Chem Neuroanat 2003; 26:133-42. [PMID: 14599663 DOI: 10.1016/s0891-0618(03)00067-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neural progenitor cells existing in the developing and adult brain retain the capacity to self renew and to produce the major cell types of the brain opening new avenues for restorative therapy of neuropsychiatric disorders. These cells can be grown in vitro while retaining the potential to differentiate into nervous tissue. A primary target for neurorestoration is Parkinson's disease, characterized by a continuous loss of the dopaminergic neurons in the substantia nigra pars compacta leading to dopamine depletion in the striatum and subsequent clinical symptoms including bradykinesia, rigidity and tremor. We established a protocol for long-term expansion and dopaminergic differentiation of rodent and human mesencephalic neural progenitor cells. Here we perform functional studies using both biochemical and electrophysiological techniques on dopaminergic neurons derived from rodent mesencephalic progenitor cells labeled with tyrosine hydroxylase (TH) gene promotor-driven expression of enhanced green fluorescence protein (EGFP). Thus, we demonstrate that these cells produce and release dopamine, express voltage-gated potassium and sodium currents, and fire action potentials. Furthermore, we detect a slowly activating hyperpolarization-activated inward cation current (I(h)), which is specific for dopaminergic neurons among present midbrain neurons. Our results demonstrate that differentiated mesencephalic progenitors exhibit some major morphological and functional characteristics of dopaminergic neurons. Therefore, these neural progenitor cells might serve as a useful source of dopaminergic neurons for studying the development and degeneration of these cells and may further serve as a continuous, on-demand source of cells for therapeutic transplantation in Parkinson's disease.
Collapse
Affiliation(s)
- Alexander Storch
- Department of Neurology, University of Ulm, Oberer Eselsberg 45, 89081 Ulm, Germany.
| | | | | | | |
Collapse
|
224
|
Yang M, Donaldson AE, Jiang Y, Iacovitti L. Factors influencing the differentiation of dopaminergic traits in transplanted neural stem cells. Cell Mol Neurobiol 2003; 23:851-64. [PMID: 14514036 PMCID: PMC1949421 DOI: 10.1023/a:1025017423102] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
1. Our previous studies demonstrated that when neural stem cells (NSCs) of the C17.2 clonal line are transplanted into the intact or 6-hydroxydopamine (6-OHDA) lesioned rat striatum, in most, but not all grafts, cells spontaneously express the dopamine (DA) biosynthetic enzymes, tyrosine hydroxylase (TH), and aromatic L-amino acid decarboxylase (Yang, M., Stull, N. D., Snyder. E. Y., Berk, M. A., and Iacovitti, L. (2002). Exp. Neurol.). 2. These results suggested that there were certain conditions which were more conducive to the development of DA traits in NSCs and possibly other neurotransmitter phenotypes. 3. In the present study, we modified a number of variables in vitro (i.e. passage number, confluence) and/or in vivo (degree, type, and site of injury) before assessing the survival, migration. and differentiation of engrafted NSCs. 4. We found that low confluence cultures were comprised exclusively of flattened polygonal cells, which when transplanted, migrated widely in the brain but did not express TH. 5. In contrast, high confluence cultures contained both polygonal cells and an overlying bed of fusiform cells. 6. When these NSCs were maintained for 12-20 passages and then transplanted, virtually all engrafted cells in 65% of the grafts expressed TH but not markers of other neurotransmitter systems. 7. Importantly, all TH+ grafts were accompanied by significant physical damage to the brain while TH- grafts were not, suggesting that local injury-related factors were also important. 8. Of no apparent influence on TH expression, regardless of how cells were grown prior to implantation, was the site of transplantation (cortex or striatum) or the degree of chemical lesion (intact, partial or full). 9. We conclude that transplanted NSCs can express traits specifically associated with DA neurons but only when cells are grown under certain conditions in vitro and then transplanted in proximity to injury-induced factors present in vivo.
Collapse
Affiliation(s)
- Ming Yang
- Department of Neurology, Thomas Jefferson University Medical College, Philadelphia, Pennsylvania USA
| | - Angela E. Donaldson
- Department of Neurology, Thomas Jefferson University Medical College, Philadelphia, Pennsylvania USA
| | - Yubao Jiang
- Department of Neurology, Thomas Jefferson University Medical College, Philadelphia, Pennsylvania USA
| | - Lorraine Iacovitti
- Department of Neurology, Thomas Jefferson University Medical College, Philadelphia, Pennsylvania USA
| |
Collapse
|
225
|
Yamada H, Dezawa M, Shimazu S, Baba M, Sawada H, Kuroiwa Y, Yamamoto I, Kanno H. Transfer of the von Hippel-Lindau gene to neuronal progenitor cells in treatment for Parkinson's disease. Ann Neurol 2003; 54:352-9. [PMID: 12953267 DOI: 10.1002/ana.10672] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuronal progenitor cells (NPCs) may provide dopaminergic neurons for the treatment of Parkinson's disease (PD). However, transplantation of NPCs into the striatum by current methods has had limited success. It is possible to reverse the symptoms of PD in model rats but difficult to reverse them in humans because the number of dopaminergic neurons generated from NPCs is low. We transduced the von Hippel-Lindau (VHL) gene into NPCs isolated from embryonic rat brain. The NPCs with the transduced VHL gene efficiently differentiated into tyrosine hydroxylase-positive neurons in vitro. NPCs with the transduced VHL gene, which were labeled in advance with bromodeoxyuridine, were transplanted into the striatum of a rat model of PD. Numerous bromodeoxyuridine-tyrosine hydroxylase double-labeled cells were seen close to the transplant site, showing that the transplanted cells efficiently generated new dopaminergic neurons within the host striatum. Moreover, all of the animals with NPCs with VHL showed a remarkable decrease in apomorphine-induced rotations. These findings show that NPCs with the VHL gene can efficiently generate dopaminergic neurons and that a sufficient number of dopaminergic neurons can develop from them to reverse the symptoms of PD in humans. VHL gene transduction provides a new therapeutic approach for treatment of PD.
Collapse
Affiliation(s)
- Hitoshi Yamada
- Yokohama Neurology Clinic and School of Medicine, Yokohama City University, 1-211-1 Mutsukawa, Minami-ku, Yokohama 232-0066, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Vroemen M, Aigner L, Winkler J, Weidner N. Adult neural progenitor cell grafts survive after acute spinal cord injury and integrate along axonal pathways. Eur J Neurosci 2003; 18:743-51. [PMID: 12925000 DOI: 10.1046/j.1460-9568.2003.02804.x] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The main rationale for cell-based therapies following spinal cord injury are: (i) replacement of degenerated spinal cord parenchyma by an axon growth supporting scaffold; (ii) remyelination of regenerating axons; and (iii), local delivery of growth promoting molecules. A potential source to meet these requirements is adult neural progenitor cells, which were examined in the present study. Fibroblast growth factor 2-responsive adult spinal cord-derived syngenic neural progenitor cells were either genetically modified in vitro to express green fluorescent protein (GFP) using retroviral vectors or prelabelled with bromodeoxyuridine (BrdU). Neural progenitor cells revealed antigenic properties of neurons and glial cells in vitro confirming their multipotency. This differentiation pattern was unaffected by retroviral transduction. GFP-expressing or BrdU-prelabelled neural progenitor cells were grafted as neurospheres directly into the acutely injured rat cervical spinal cord. Animals with lesions only served as controls. Three weeks postoperatively, grafted neural progenitor cells integrated along axonal profiles surrounding the lesion site. In contrast to observations in culture, grafted neural progenitor cells differentiated only into astro- and oligodendroglial lineages, supporting the notion that the adult spinal cord provides molecular cues for glial, but not for neuronal, differentiation. This study demonstrates that adult neural progenitor cells will survive after transplantation into the acutely injured spinal cord. The observed oligodendroglial and astroglial differentiation and integration along axonal pathways represent important prerequisites for potential remyelination and support of axonal regrowth.
Collapse
Affiliation(s)
- Maurice Vroemen
- Department of Neurology, University of Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany
| | | | | | | |
Collapse
|
227
|
Abstract
The discovery of neural stem cells (NSCs) has changed our long-held view that the adult mammalian central nervous system (CNS) is postmitotic and lacks the capability for self-repair. The role of NSCs in physiological and pathological processes in the brain is slowly emerging. We are now able to isolate, expand, genetically engineer and transplant NSCs. An important characteristic of NSCs, not fully understood so far, is their migratory ability and their tropism to brain pathology. The migratory ability of NSCs and their capacity to differentiate into all neural phenotypes gives us a potentially powerful tool for the treatment of both diffuse and localised neurologic disorders. The delivery of gene products by NSCs to specific sites in the CNS can maximise the efficiency of delivery and minimise the unwanted exposure of surrounding intact tissue. Here, the recent preclinical advances in the use of NSCs for the delivery of therapeutic products are reviewed, in particular the employment of their migratory potential and the homing ability to pathology in the nervous system.
Collapse
Affiliation(s)
- Peter Kabos
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Ste. 800E, 8631 W 3rd St, Los Angeles, CA 90048, USA
| | | | | | | |
Collapse
|
228
|
Curtis MA, Penney EB, Pearson AG, van Roon-Mom WMC, Butterworth NJ, Dragunow M, Connor B, Faull RLM. Increased cell proliferation and neurogenesis in the adult human Huntington's disease brain. Proc Natl Acad Sci U S A 2003; 100:9023-7. [PMID: 12853570 PMCID: PMC166431 DOI: 10.1073/pnas.1532244100] [Citation(s) in RCA: 404] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neurogenesis has recently been observed in the adult human brain, suggesting the possibility of endogenous neural repair. However, the augmentation of neurogenesis in the adult human brain in response to neuronal cell loss has not been demonstrated. This study was undertaken to investigate whether neurogenesis occurs in the subependymal layer (SEL) adjacent to the caudate nucleus in the human brain in response to neurodegeneration of the caudate nucleus in Huntington's disease (HD). Postmortem control and HD human brain tissue were examined by using the cell cycle marker proliferating cell nuclear antigen (PCNA), the neuronal marker beta III-tubulin, and the glial cell marker glial fibrillary acidic protein (GFAP). We observed a significant increase in cell proliferation in the SEL in HD compared with control brains. Within the HD group, the degree of cell proliferation increased with pathological severity and increasing CAG repeats in the HD gene. Most importantly, PCNA+ cells were shown to coexpress beta III-tubulin or GFAP, demonstrating the generation of neurons and glial cells in the SEL of the diseased human brain. Our results provide evidence of increased progenitor cell proliferation and neurogenesis in the diseased adult human brain and further indicate the regenerative potential of the human brain.
Collapse
Affiliation(s)
- Maurice A Curtis
- Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Wright LS, Li J, Caldwell MA, Wallace K, Johnson JA, Svendsen CN. Gene expression in human neural stem cells: effects of leukemia inhibitory factor. J Neurochem 2003; 86:179-95. [PMID: 12807438 DOI: 10.1046/j.1471-4159.2003.01826.x] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Human neural precursor cells grown in culture provide a source of tissue for drug screening, developmental studies and cell therapy. However, mechanisms underlying their growth and differentiation are poorly understood. We show that epidermal growth factor (EGF) responsive precursors derived from the developing human cortex undergo senescence after 30-40 population doublings. Leukemia inhibitory factor (LIF) increased overall expansion rates, prevented senescence and allowed the growth of a long-term self renewing neural stem cell (ltNSCctx) for up to 110 population doublings. We established basal gene expression in ltNSCctx using Affymetrix oligonucleotide microarrays that delineated specific members of important growth factor and signaling families consistently expressed across three separate lines. Following LIF withdrawal, 200 genes showed significant decreases. Protein analysis confirmed LIF-regulated expression of glial fibrillary acidic protein, CD44, and major histocompatibility complex I. This study provides the first molecular profile of human ltNSCctx cultures capable of long-term self renewal, and reveals specific sets of genes that are directly or indirectly regulated by LIF.
Collapse
Affiliation(s)
- Lynda S Wright
- The Waisman Center Stem Cell Research Program and Department of Anatomy, University of Wisconsin, 53705, USA
| | | | | | | | | | | |
Collapse
|
230
|
Just L, Timmer M, Tinius J, Stahl F, Deiwick A, Nikkhah G, Bader A. Identification of human cells in brain xenografts and in neural co-cultures of rat by in situ hybridisation with Alu probe. J Neurosci Methods 2003; 126:69-77. [PMID: 12788503 DOI: 10.1016/s0165-0270(03)00065-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transplantation of human cells into animal models of neurodegenerative disorders is an important scientific application to analyse the survival and developmental capacity of grafted human cells under in vivo conditions. It is critical, therefore, to have a reliable method to distinguish between human and animal cells. In the present study, we describe a combined in situ hybridisation and immunocytochemistry method for the identification of human cells in cultured rat brain cells and xenografts. The specific Alu probe we utilised, which corresponds to the consensus sequence of human Alu repeats was evaluated by southern blot hybridisation of zoo blot and by in situ hybridisation of primary and neoplastic cells from man, rat, mouse, and hamster. This method allows a definite identification of human cells in neural xenografts and, in combination with additional in situ techniques, a further detection of grafted cells.
Collapse
Affiliation(s)
- Lothar Just
- Organ and Tissue Culturing, German Research Centre for Biotechnology, Mascheroder Weg 1, D-38124 Braunschweig, Germany.
| | | | | | | | | | | | | |
Collapse
|
231
|
Murakami T, Fujimoto Y, Yasunaga Y, Ishida O, Tanaka N, Ikuta Y, Ochi M. Transplanted neuronal progenitor cells in a peripheral nerve gap promote nerve repair. Brain Res 2003; 974:17-24. [PMID: 12742620 DOI: 10.1016/s0006-8993(03)02539-3] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A basic experiment of peripheral nerve regeneration using neuronal progenitor cells embedded in collagen gel was performed in a rat sciatic nerve defect. First, when neuronal progenitor cells derived from the fetal rat hippocampus were cultured in atelocollagen-containing medium, neurospheres positive for anti-nestin antibody were confirmed after 8 days. These cells differentiated into astrocytes positive for anti-glial fibrillary acidic protein (GFAP) antibody, oligodendrocytes positive for anti-galactocerebroside (GalC) antibody and neurons positive for anti-neurofilament 200 (NF200) antibody, and they were capable of extending axons. They also differentiated into Schwann-like supportive cells positive for anti-s100 and anti-p75 antibody. Next, a 15-mm defect was prepared in the sciatic nerve of mature rats, and the nerve was bridged with a silicone tube filled with neuronal progenitor cells (1 x 10(5)) embedded in collagen gel. The transplanted neuronal progenitor cells were labeled in advance with 5-bromo-2-deoxyuridine (BrdU). When the regenerated tissue was examined 6 weeks and 10 weeks after grafting, the number and diameter of myelinated fibers were significantly increased compared with a control tube without neuronal progenitor cells. Action potentials were detected in the regenerated nerve. Also, cells positive for both anti-BrdU antibody and anti-S100 or anti-p75 antibody were observed in the regenerated tissue, and part of the grafted neural stem cells were considered to have differentiated into Schwann cell-like supportive cells. From these results neuronal progenitor cells derived from the fetal rat hippocampus are considered to retain their proliferative and differentiating abilities in collagen gel, and when transplanted to a site of peripheral nerve defect, part of them differentiate into supportive cells and they contributed to promotion of axonal regeneration.
Collapse
Affiliation(s)
- Takeshi Murakami
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima-city 734-8551, Japan.
| | | | | | | | | | | | | |
Collapse
|
232
|
Perrier AL, Studer L. Making and repairing the mammalian brain--in vitro production of dopaminergic neurons. Semin Cell Dev Biol 2003; 14:181-9. [PMID: 12948353 DOI: 10.1016/s1084-9521(03)00010-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Midbrain dopamine (DA) neurons play an essential role in modulating motor control, and their degeneration is the hallmark feature of Parkinson's disease (PD). In vitro production of DA neurons provides insight into the mechanisms that control cell fate choice, and offers an alternative to the use of fetal tissue for experimental cell replacement in PD. Here we will review the advantages and disadvantages of the various renewable cell sources and protocols tested, and discuss their relevance for basic studies and for cell therapy.
Collapse
Affiliation(s)
- Anselme L Perrier
- Laboratory of Stem Cell & Tumor Biology, Neurosurgery and Developmental Biology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 256, New York, NY 10021, USA
| | | |
Collapse
|
233
|
Triarhou LC. Directions for future research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 517:127-42. [PMID: 12580310 DOI: 10.1007/978-1-4615-0699-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Affiliation(s)
- Lazaros C Triarhou
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, Medical Science Building A142, Indiana University Medical Center, 635 Barnhill Drive, Indianapolis, Indiana 46202-5120, USA
| |
Collapse
|
234
|
Abstract
1. Neural stem cells can be cultured from the CNS of different mammalian species at many stages of development. They have an extensive capacity for self-renewal and will proliferate ex vivo in response to mitogenic growth factors or following genetic modification with immortalising oncogenes. Neural stem cells are multipotent since their differentiating progeny will give rise to the principal cellular phenotypes comprising the mature CNS: neurons, astrocytes and oligodendrocytes. 2. Neural stem cells can also be derived from more primitive embryonic stem (ES) cells cultured from the blastocyst. ES cells are considered to be pluripotent since they can give rise to the full cellular spectrum and will, therefore, contribute to all three of the embryonic germ layers: endoderm, mesoderm and ectoderm. However, pluripotent cells have also been derived from germ cells and teratocarcinomas (embryonal carcinomas) and their progeny may also give rise to the multiple cellular phenotypes contributing to the CNS. In a recent development, ES cells have also been isolated and grown from human blastocysts, thus raising the possibility of growing autologous stem cells when combined with nuclear transfer technology. 3. There is now an emerging recognition that the adult mammalian brain, including that of primates and humans, harbours stem cell populations suggesting the existence of a previously unrecognised neural plasticity to the mature CNS, and thereby raising the possibility of promoting endogenous neural reconstruction. 4. Such reports have fuelled expectations for the clinical exploitation of neural stem cells in cell replacement or recruitment strategies for the treatment of a variety of human neurological conditions including Parkinson's disease (PD), Huntington's disease, multiple sclerosis and ischaemic brain injury. Owing to their migratory capacity within the CNS, neural stem cells may also find potential clinical application as cellular vectors for widespread gene delivery and the expression of therapeutic proteins. In this regard, they may be eminently suitable for the correction of genetically-determined CNS disorders and in the management of certain tumors responsive to cytokines. Since large numbers of stem cells can be generated efficiently in culture, they may obviate some of the technical and ethical limitations associated with the use of fresh (primary) embryonic neural tissue in current transplantation strategies. 5. While considerable recent progress has been made in terms of developing new techniques allowing for the long-term culture of human stem cells, the successful clinical application of these cells is presently limited by our understanding of both (i) the intrinsic and extrinsic regulators of stem cell proliferation and (ii) those factors controlling cell lineage determination and differentiation. Although such cells may also provide accessible model systems for studying neural development, progress in the field has been further limited by the lack of suitable markers needed for the identification and selection of cells within proliferating heterogeneous populations of precursor cells. There is a further need to distinguish between the committed fate (defined during normal development) and the potential specification (implying flexibility of fate through manipulation of its environment) of stem cells undergoing differentiation. 6. With these challenges lying ahead, it is the opinion of the authors that stem-cell therapy is likely to remain within the experimental arena for the foreseeable future. In this regard, few (if any) of the in vivo studies employing neural stem cell grafts have shown convincingly that behavioural recovery can be achieved in the various model paradigms. Moreover, issues relating to the quality control of cultured cells and their safety following transplantation have only begun to be addressed. 7. While on the one hand cell biotechnologists have been quick to realise the potential commercial value, human stem cell research and its clinical applications has been the subject of intense ethical and legislative considerations. The present chapter aims to review some recent aspects of stem cell research applicable to developmental neurobiology and the potential applications in clinical neuroscience.
Collapse
Affiliation(s)
- T Ostenfeld
- MRC Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
235
|
Stachowiak EK, Fang X, Myers J, Dunham S, Stachowiak MK. cAMP-induced differentiation of human neuronal progenitor cells is mediated by nuclear fibroblast growth factor receptor-1 (FGFR1). J Neurochem 2003; 84:1296-312. [PMID: 12614330 DOI: 10.1046/j.1471-4159.2003.01624.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Activation of cAMP signaling pathway and its transcriptional factor cyclic AMP response element binding protein (CREB) and coactivator are key determinants of neuronal differentiation and plasticity. We show that nuclear fibroblast growth factor receptor-1 (FGFR1) mediates cAMP-induced neuronal differentiation and regulates CREB and CREB binding protein (CBP) function in alpha-internexin-expressing human neuronal progenitor cells (HNPC). In proliferating HNPC, FGFR1 was associated with the cytoplasm and plasma membrane. Treatment with dB-cAMP induced nuclear accumulation of FGFR1 and caused neuronal differentiation, accompanied by outgrowth of neurites expressing MAP2 and neuron-specific neurofilament-L protein and enolase. HNPC transfected with nuclear/cytoplasmic FGFR1 or non-membrane FGFR1(SP-/NLS), engineered to accumulate exclusively in the cell nucleus, underwent neuronal differentiation in the absence of cAMP stimulation. In contrast, FGFR1/R4, with highly hydrophobic transmembrane domain of FGFR4, was membrane associated, did not enter the nucleus and failed to induce neuronal differentiation. Transfection of tyrosine kinase-deleted dominant negative receptor mutants, cytoplasmic/nuclear FGFR1(TK-) or nuclear FGFR1(SP-/NLS)(TK-), prevented cAMP-induced neurite outgrowth. Nuclear FGFR1 localized in speckle-like domains rich in phosphorylated histone 3 and splicing factors, regions known for active RNA transcription and processing, and activated the neurofilament-L gene promoter. FGFR1(SP-/NLS) transactivated CRE, up-regulated phosphorylation and transcriptional activity of CREB and stimulated the activity of CBP several-fold. Thus, cAMP-induced nuclear accumulation of FGFR1 provides a signal that triggers molecular events leading to neuronal differentiation.
Collapse
Affiliation(s)
- E K Stachowiak
- Molecular and Structural Neurobiology and Gene Therapy Program, Department Pathology and Anatomical Sciences, State University of New York, Buffalo 14214, USA
| | | | | | | | | |
Collapse
|
236
|
Kitamura Y, Nomura Y. Stress proteins and glial functions: possible therapeutic targets for neurodegenerative disorders. Pharmacol Ther 2003; 97:35-53. [PMID: 12493534 DOI: 10.1016/s0163-7258(02)00301-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent findings suggest that unfolded or misfolded proteins participate in the pathology of several neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease. Usually, several stress proteins and glial cells act as intracellular molecular chaperones and show chaperoning neuronal function, respectively. In the brains of patients with neurodegenerative disorders, however, stress proteins are expressed and frequently associated with protein aggregates, and glial cells are activated around degenerative regions. In addition, several stress proteins and glial cells may also regulate neuronal cell death and loss. Therefore, some types of stress proteins and glial cells are considered to be neuroprotective targets. We summarize the current findings regarding the neuroprotective effects of stress proteins and glial cells, and discuss the possibility of using this knowledge to develop new therapeutic strategies to treat neurodegeneration.
Collapse
Affiliation(s)
- Yoshihisa Kitamura
- Department of Neurobiology, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | | |
Collapse
|
237
|
Abstract
BACKGROUND By affecting young people during the most productive period of their lives, spinal cord injury is a devastating problem for modern society. A decade ago, treating SCI seemed frustrating and hopeless because of the tremendous morbidity and mortality, life-shattering impact, and limited therapeutic options associated with the condition. Today, however, an understanding of the underlying pathophysiological mechanisms, the development of neuroprotective interventions, and progress toward regenerative interventions are increasing hope for functional restoration. REVIEW SUMMARY This study addresses the present understanding of SCI, including the etiology, pathophysiology, treatment, and scientific advances. The discussion of treatment options includes a critical review of high-dose methylprednisolone and GM-1 ganglioside therapy. The concept that limited rebuilding can provide a disproportionate improvement in quality of life is emphasized throughout. CONCLUSIONS New surgical procedures, pharmacologic treatments, and functional neuromuscular stimulation methods have evolved over the last decades that can improve functional outcomes after spinal cord injury, but limiting secondary injury remains the primary goal. Tissue replacement strategies, including the use of embryonic stem cells, become an important tool and can restore function in animal models. Controlled clinical trials are now required to confirm these observations. The ultimate goal is to harness the body's own potential to replace lost central nervous system cells by activation of endogenous progenitor cell repair mechanisms.
Collapse
Affiliation(s)
- Daniel Becker
- Department of Neurology, Spinal Cord Injury Neuro-Rehabilitation Section, Restorative Treatment and Research Program, Washington University School of Medicine, St Louis, Missouri 63108, USA
| | | | | |
Collapse
|
238
|
Segovia J. Gene therapy for Parkinson's disease: current status and future potential. AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2002; 2:135-46. [PMID: 12083948 DOI: 10.2165/00129785-200202020-00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Parkinson's disease appears to be a good candidate for gene therapy. The primary biochemical defect associated with the disease has been clearly determined as an absence of dopamine in the caudate-putamen, and the anatomical region where the neuropathologic hallmark of the disease, death of the nigral dopamine-producing neurons, occurs, remains circumscribed. Based on the biochemical and anatomical information gathered on Parkinson's disease, different gene therapy strategies have been devised to treat it. The first, and most explored strategy so far, consists in engineering cells to produce levodopa or dopamine so they will replace dopaminergic neurotransmission. Several types of cells have been employed in these experiments, and behavioral recovery has been reported in animal models of the disease. However, this approach cannot prevent neuronal death, nor reconstruct brain circuits. Another strategy is to protect cells by transferring genes that encode neurotrophic factors. Effort is now being concentrated into this research area, and promising results have recently been reported. Finally, an additional strategy aims at generating cells with a dopaminergic phenotype so they will be capable of replacing the missing dopaminergic neurons in biochemical, anatomical and functional terms. This has the potential to become an important constituent for an effective cure. Gene therapy holds significant promise for the treatment of neurodegenerative disorders, and Parkinson's disease treatment will benefit greatly from the knowledge and information arising from gene therapy research.
Collapse
Affiliation(s)
- José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Distrito Federal, Mexico.
| |
Collapse
|
239
|
Wu P, Tarasenko YI, Gu Y, Huang LYM, Coggeshall RE, Yu Y. Region-specific generation of cholinergic neurons from fetal human neural stem cells grafted in adult rat. Nat Neurosci 2002; 5:1271-8. [PMID: 12426573 DOI: 10.1038/nn974] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2002] [Accepted: 10/22/2002] [Indexed: 02/07/2023]
Abstract
Pluripotent or multipotent stem cells isolated from human embryos or adult central nervous system (CNS) may provide new neurons to ameliorate neural disorders. A major obstacle, however, is that the majority of such cells do not differentiate into neurons when grafted into non-neurogenic areas of the adult CNS. Here we report a new in vitro priming procedure that generates a nearly pure population of neurons from fetal human neural stem cells (hNSCs) transplanted into adult rat CNS. Furthermore, the grafted cells differentiated by acquiring a cholinergic phenotype in a region-specific manner. This technology may advance stem cell-based therapy to replace lost neurons in neural injury or neurodegenerative disorders.
Collapse
Affiliation(s)
- Ping Wu
- Department of Anatomy, University of Texas Medical Branch, Galveston, Texas 77555, USA.
| | | | | | | | | | | |
Collapse
|
240
|
Akita J, Takahashi M, Hojo M, Nishida A, Haruta M, Honda Y. Neuronal differentiation of adult rat hippocampus-derived neural stem cells transplanted into embryonic rat explanted retinas with retinoic acid pretreatment. Brain Res 2002; 954:286-93. [PMID: 12414111 DOI: 10.1016/s0006-8993(02)03356-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The purpose of this study was to evaluate the effects of the retinal environment and retinoic acid (RA) pretreatment on the differentiation of transplanted adult rat hippocampus-derived neural stem cells (AHSCs). AHSCs were transplanted into embryonic (E18) or neonatal (P6) rat retinal explants and the mixture was cultured for 2 weeks. Other AHSCs were stimulated by 0.5 microM all-trans RA for 6 days before transplantation. Immunofluorescent double staining showed that a larger number of AHSCs became beta-tubulin III-positive neurons in the E18 than in P6 retinas. In addition, many AHSCs became MAP2ab-positive and MAP5-positive neurons following RA pretreatment and transplantation. Only a few AHSCs became HPC-1-, calbindin-, PKC- or rhodopsin-positive cells under these conditions. We conclude that the microenvironment supplied by embryonic retinas is conductive to neuronal differentiation in general. RA stimulation before transplantation was also effective in stimulating differentiation.
Collapse
Affiliation(s)
- Joe Akita
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | |
Collapse
|
241
|
Dietrich J, Noble M, Mayer-Proschel M. Characterization of A2B5+ glial precursor cells from cryopreserved human fetal brain progenitor cells. Glia 2002; 40:65-77. [PMID: 12237844 DOI: 10.1002/glia.10116] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The identification and characterization of human neural precursor cells are critical in extending our understanding of central nervous system development from model animal systems to our own species. Moreover, availability of well-characterized populations of human cells is of potential value in endeavors ranging from cell transplantation to drug screening. We have isolated a population of continuously dividing glial-restricted precursor cells from commercially available cryopreserved 18-20 weeks old fetal brain neural progenitor cells. These human glial-restricted precursor cells are A2B5(+) and do not express polysialylated E-NCAM (PSA-NCAM). They can be grown as purified populations in serum-free medium supplemented with basic fibroblast growth factor (bFGF) and can be induced to generate cells with the antigenic characteristics of oligodendrocytes and distinct astrocytic populations.
Collapse
Affiliation(s)
- Joerg Dietrich
- Department of Biomedical Genetics, University of Rochester, Rochester, New York 14642, USA
| | | | | |
Collapse
|
242
|
Okano H, Yoshizaki T, Shimazaki T, Sawamoto K. Isolation and transplantation of dopaminergic neurons and neural stem cells. Parkinsonism Relat Disord 2002; 9:23-8. [PMID: 12217619 DOI: 10.1016/s1353-8020(02)00041-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although transplantation of mesencephalic tissue is considered a promising therapy for Parkinson's disease (PD), its clinical use is still restricted to a very few cases. A major limiting factor of this therapy is the difficulty of obtaining sufficient quantities of viable embryonic mesencephalic tissue. To overcome this limitation, techniques to produce dopaminergic (DA) neurons in vitro have been developed. However, these cultures are likely to contain a variety of unidentified cells, which must be removed before implantation. Specific cell-surface markers to sort DA neurons or their precursors are not available. We have developed an alternative strategy, by which these cells can be labeled with green fluorescent protein and isolated with fluorescent activated cell sorter. Transplantation of the sorted cells resulted in recovery of a rat model of the PD. This strategy should be useful for developing new therapies for PD.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | | | |
Collapse
|
243
|
Galvin KA, Jones DG. Adult human neural stem cells for cell-replacement therapies in the central nervous system. Med J Aust 2002; 177:316-8. [PMID: 12225280 DOI: 10.5694/j.1326-5377.2002.tb04791.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2002] [Accepted: 06/06/2002] [Indexed: 11/17/2022]
Abstract
Human neural stem cells (HNSCs) can be isolated from both the developing and adult central nervous system (CNS). HNSCs can be successfully grown in culture, are self-renewable, and can generate mature neuronal and glial progeny. Embryonic HNSCs can be induced to differentiate into specific neuronal phenotypes. HNSCs successfully integrate into the host environment after transplantation into the developing or adult CNS. HNSCs transplanted into animal models of Parkinson's disease and spinal cord injury have induced functional recovery. The risks associated with stem cell transplantation trials are difficult to assess, but have not become overtly apparent throughout preclinical investigations. Major hurdles remain to be overcome before human clinical trials can be embarked upon.
Collapse
Affiliation(s)
- Kerry A Galvin
- Department of Anatomy and Structural Biology, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
244
|
Abstract
Neural stem cells (NSCs) are multipotential progenitor cells that have self-renewal activities. A single NSC is capable of generating various kinds of cells within the central nervous system (CNS), including neurons, astrocytes, and oligodendrocytes. Because of these characteristics, there is increasing interest in NSCs and neural progenitor cells from the aspects of both basic developmental biology and therapeutic applications to the damaged brain. This special issue, dedicated to understanding the nature of the NSCs present in the CNS, presents an introduction to several avenues of research that may lead to feasible strategies for manipulating cells in situ to treat the damaged brain. The topics covered by these studies include the extracellular factors and signal transduction cascades involved in the differentiation and maintenance of NSCs, the population dynamics and locations of NSCs in embryonic and adult brains, prospective identification and isolation of NSCs, the induction of NSCs to adopt particular neuronal phenotypes, and their transplantation into the damaged CNS.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
245
|
Tamaki S, Eckert K, He D, Sutton R, Doshe M, Jain G, Tushinski R, Reitsma M, Harris B, Tsukamoto A, Gage F, Weissman I, Uchida N. Engraftment of sorted/expanded human central nervous system stem cells from fetal brain. J Neurosci Res 2002; 69:976-86. [PMID: 12205691 DOI: 10.1002/jnr.10412] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Direct isolation of human central nervous system stem cells (CNS-SC) based on cell surface markers yields a highly purified stem cell population that can extensively expand in vitro and exhibit multilineage differentiation potential both in vitro and in vivo. The CNS-SC were isolated from fetal brain tissue using the cell surface markers CD133(+), CD34(-), CD45(-), and CD24(-/lo) (CD133(+) cells). Fluorescence-activated cell sorted (FACS) CD133(+) cells continue to expand exponentially as neurospheres while retaining multipotential differentiation capacity for >10 passages. CD133(-), CD34(-), and CD45(-) sorted cells (approximately 95% of total fetal brain tissue) fail to initiate neurospheres. Neurosphere cells transplanted into neonatal immunodeficient NOD-SCID mice proliferated, migrated, and differentiated in a site-specific manner. However, it has been difficult to evaluate human cell engraftment, because many of the available monoclonal antibodies against neural cells (beta-tubulin III and glial fibrillary acidic protein) are not species specific. To trace the progeny of human cells after transplantation, CD133(+)-derived neurosphere cells were transduced with lentiviral vectors containing enhanced green fluorescent protein (eGFP) expressed downstream of the phosphoglycerate kinase promoter. After transduction, GFP(+) cells were enriched by FACS, expanded, and transplanted into the lateral ventricular space of neonatal immunodeficient NOD-SCID brain. The progeny of transplanted cells were detected by either GFP fluorescence or antibody against GFP. GFP(+) cells were present in the subventricular zone-rostral migrating stream, olfactory bulb, and hippocampus as well as nonneurogenic sites, such as cerebellum, cerebral cortex, and striatum. Antibody against GFP revealed that some of the cells displayed differentiating dendrites and processes with neurons or glia cells. Thus, marking human CNS-SC with reporter genes introduced by lentiviral vectors is a useful tool with which to characterize migration and differentiation of human cells in this mouse transplantation model.
Collapse
|
246
|
Yang M, Stull ND, Berk MA, Snyder EY, Iacovitti L. Neural stem cells spontaneously express dopaminergic traits after transplantation into the intact or 6-hydroxydopamine-lesioned rat. Exp Neurol 2002; 177:50-60. [PMID: 12429210 DOI: 10.1006/exnr.2002.7989] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability to differentiate neural stem cells (NSCs) into dopamine neurons is fundamental to their role in cell replacement therapies for neurodegenerative disorders such as Parkinson's disease. We show here that when a clonal line (C17.2) of undifferentiated NSCs is transplanted into the intact or 6-hydroxydopamine-lesioned striatum, cells withdraw from the cell cycle (BrdU(-)), migrate extensively in the host striatum, and express markers associated with neuronal (beta-tubulin III(+), NSE(+), NeuN(+)) but not glial (GFAP(-), MBP(-), A2B5(-)) differentiation. Importantly, by 2-5 weeks postgrafting, in the majority of these transplants, nearly all engrafted cells express the dopamine-synthesizing enzymes tyrosine hydroxylase and aromatic L-amino decarboxylase, sometimes resulting in changes in motor behavior. In contrast, no NSCs stain for dopamine-beta-hydroxylase, choline acetyltransferase, glutamic acid decarboxylase, or serotonin. We conclude that, following transplantation into the intact or 6-hydroxydopamine-lesioned rat, the adult brain contains intrinsic cues sufficient to direct the specific expression of dopaminergic traits in immature multipotential neural stem cells.
Collapse
Affiliation(s)
- Ming Yang
- Department of Neurology, Thomas Jefferson University Medical College, 1025 Walnut Street, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|
247
|
Suzuki A, Obi K, Urabe T, Hayakawa H, Yamada M, Kaneko S, Onodera M, Mizuno Y, Mochizuki H. Feasibility of ex vivo gene therapy for neurological disorders using the new retroviral vector GCDNsap packaged in the vesicular stomatitis virus G protein. J Neurochem 2002; 82:953-60. [PMID: 12358801 DOI: 10.1046/j.1471-4159.2002.01048.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal progenitor cells (NPC) are particularly suited as the target population for genetic and cellular therapy of neurological disorders such as Parkinson's disease or stroke. However, genetic modification of these cells using retroviral vectors remains a great challenge because of the low transduction rate and the need for fetal calf serum (FCS) during the transduction process that induces the cell differentiation to mature neurons. To overcome these problems, we developed a new retrovirus production system in which the simplified retroviral vector GCDNsap engineered to be resistant to denovo methylation was packaged in the vesicular stomatitis virus G protein (VSV-G), concentrated by centrifugation, and resuspended in serum-free medium (StemPro-34 SFM). In transduction experiments using enhanced green fluorescent protein (EGFP) as a marker, the concentrated FCS-free virus supernatant infected NPC at a high rate, while maintaining the ability of these cells to self-renew and differentiate in vitro. When such cells were grafted into mouse brains, EGFP-expressing NPC were detected in the region around the injection site at 8 weeks post transplantation. These findings suggest that the gene transfer system described here may provide a useful tool to genetically modify NPC for treatments of neurological disorders.
Collapse
Affiliation(s)
- A Suzuki
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Chen J, Li Y, Wang L, Lu M, Chopp M. Caspase inhibition by Z-VAD increases the survival of grafted bone marrow cells and improves functional outcome after MCAo in rats. J Neurol Sci 2002; 199:17-24. [PMID: 12084437 DOI: 10.1016/s0022-510x(02)00075-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Marrow stromal cells (MSCs) transplantation into brain has been employed to treat experimental ischemia. However, MSCs undergo apoptosis and few survive in the ischemic brain. We test the hypotheses that coadministration of bone marrow cells (BMCs) with a cell-permeable inhibitor of caspases, Z-Val-Ala-DL-Asp-fluoromethylketone (Z-VAD), into the ischemic boundary zone (IBZ) of brain promotes BMCs survival and improve outcome. Experimental groups consist of: 24 h after MCAo, either phosphate-buffered saline (PBS, n=4), dead BMC (n=4), fresh BMC (n=10), Z-VAD only (n=4), or BMC with Z-VAD (n=6) were intracerebrally injected. BMCs were harvested from donor adult rats labeled with bromodeoxyuridine (BrdU). Rats were subjected to an adhesive-removal somatosensory and motor-rotarod functional tests before MCAo and at 1 and 7 days after MCAo. Rats treated with a combination of Z-VAD and BMCs exhibited significant improvement in the adhesive-removal test at 7 days compared with the control group (combined MCAo+PBS and MCAo+dead BMC) (p<0.01), and the numbers of BrdU-BMC increased (p<0.05) and apoptotic cells decreased (p<0.05) compared with BMC alone transplantation. Our data suggest that intracerebral coadministration of BMC with Z-VAD enhances the survival of grafted BMC and improves neurological functional recovery after MCAo.
Collapse
Affiliation(s)
- Jieli Chen
- Department of Neurology, Henry Ford Health Sciences Center, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | | | | | | | | |
Collapse
|
249
|
Safford KM, Hicok KC, Safford SD, Halvorsen YDC, Wilkison WO, Gimble JM, Rice HE. Neurogenic differentiation of murine and human adipose-derived stromal cells. Biochem Biophys Res Commun 2002; 294:371-9. [PMID: 12051722 DOI: 10.1016/s0006-291x(02)00469-2] [Citation(s) in RCA: 554] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The identification of cells capable of neuronal differentiation has great potential for cellular therapies. We examined whether murine and human adipose-derived adult stem (ADAS) cells can be induced to undergo neuronal differentiation. We isolated ADAS cells from the adipose tissue of adult BalbC mice or from human liposuction tissue and induced neuronal differentiation with valproic acid, butylated hydroxyanisole, insulin, and hydrocortisone. As early as 1-3 h after neuronal induction, the phenotype of ADAS cells changed towards neuronal morphology. Following neuronal induction, muADAS cells displayed immunocytochemical staining for GFAP, nestin and NeuN and huADAS cells displayed staining for intermediate filament M, nestin, and NeuN. Following neuronal induction of murine and human ADAS cells, Western blot analysis confirmed GFAP, nestin, and NeuN protein expression. Pretreatment with EGF and basic FGF augmented the neuronal differentiation of huADAS cells. The neuronal differentiation of stromal cells from adipose tissue has broad biological and clinical implications.
Collapse
Affiliation(s)
- Kristine M Safford
- Department of Surgery, Division of Pediatric Surgery, Box 3815, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
250
|
Abstract
Neural stem cells (NSCs) have great potential as a therapeutic tool for the repair of a number of CNS disorders. NSCs can either be isolated from embryonic and adult brain tissue or be induced from both mouse and human ES cells. These cells proliferate in vitro through many passages without losing their multipotentiality. Following engraftment into the adult CNS, NSCs differentiate mainly into glia, except in neurogenic areas. After engraftment into the injured and diseased CNS, their differentiation is further retarded. In vitro manipulation of NSC fate prior to transplantation and/or modification of the host environment may be necessary to control the terminal lineage of the transplanted cells to obtain functionally significant numbers of neurons. NSCs and a few types of glial precursors have shown the capability to differentiate into oligodendrocytes and to remyeliate the demyelinated axons in the CNS, but the functional extent of remyelination achieved by these transplants is limited. Manipulation of endogenous neural precursors may be an alternative therapy or a complimentary therapy to stem cell transplantation for neurodegenerative disease and CNS injury. However, this at present is challenging and so far has been unsuccessful. Understanding mechanisms of NSC differentiation in the context of the injured CNS will be critical to achieving these therapeutic strategies.
Collapse
Affiliation(s)
- Qilin Cao
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | | | |
Collapse
|