151
|
Zhang X, Jin G, Wang L, Hu W, Tian M, Qin J, Huang H. Brn-4 is upregulated in the deafferented hippocampus and promotes neuronal differentiation of neural progenitors in vitro. Hippocampus 2009; 19:176-86. [PMID: 18831054 DOI: 10.1002/hipo.20498] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Fimbria-fornix (FF), the septo-hippocampal pathway, was transected to model Alzheimer's disease (AD), which is characterized by loss of cholinergic afferent fibers in hippocampus. Various alternations may happen in the deafferented hippocampus. In this study, we determined the expression of Brn-4 in hippocampus after FF lesion. RT-PCR and Western blot showed that mRNA transcription and protein of Brn-4 increased significantly and reached to the peak at day 14 after FF lesion. Hybridization and immunohistochemistry indicated that Brn-4 signals in hippocampus and dentate gyrus (DG) of the deafferented side were significantly stronger than the normal side. More Brn-4 positive cells were identified in the DG of deafferented hippocampus. In the pyramidal and granular cells, Brn-4 positive cells were all NeuN positive neurons, whereas in the neurogenic area, subgranular zone (SGZ), only a part of Brn-4 positive cells were NeuN positive, and these Brn-4/NeuN double positive neurons in SGZ and hilus of DG increased significantly after the trauma induced by FF lesion. In vitro Brn-4 antibody attenuated the role of extract from deafferented hippocampus in promoting differentiation of hippocampal progenitors into MAP-2 positive neurons. This study demonstrated that after FF lesion, Brn-4 in the deafferented hippocampus was upregulated and might play an important role in inducing local progenitors to differentiate into neurons, which may compensate for the loss of cholinergic afferent fibers or other dysfunctions.
Collapse
Affiliation(s)
- Xinhua Zhang
- Department of Anatomy and Neurobiology, The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, PR China
| | | | | | | | | | | | | |
Collapse
|
152
|
Carlessi L, De Filippis L, Lecis D, Vescovi A, Delia D. DNA-damage response, survival and differentiation in vitro of a human neural stem cell line in relation to ATM expression. Cell Death Differ 2009; 16:795-806. [PMID: 19229246 DOI: 10.1038/cdd.2009.10] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ataxia-telangiectasia (A-T) is a neurodegenerative disorder caused by defects in the ATM kinase, a component of the DNA-damage response (DDR). Here, we employed an immortalized human neural stem-cell line (ihNSC) capable of differentiating in vitro into neurons, oligodendrocytes and astrocytes to assess the ATM-dependent response and outcome of ATM ablation. The time-dependent differentiation of ihNSC was accompanied by an upregulation of ATM and DNA-PK, sharp downregulation of ATR and Chk1, transient induction of p53 and by the onset of apoptosis in a fraction of cells. The response to ionizing radiation (IR)-induced DNA lesions was normal, as attested by the phosphorylation of ATM and some of its substrates (e.g., Nbs1, Smc1, Chk2 and p53), and by the kinetics of gamma-H2AX nuclear foci formation. Depletion in these cells of ATM by shRNA interference (shATM) attenuated the differentiation-associated apoptosis and response to IR, but left unaffected the growth, self-renewal and genomic stability. shATM cells generated a normal number of MAP2/beta-tubulin III+ neurons, but a reduced number of GalC+ oligodendrocytes, which were nevertheless more susceptible to oxidative stress. Altogether, these findings highlight the potential of ihNSCs as an in vitro model system to thoroughly assess, besides ATM, the role of DDR genes in neurogenesis and/or neurodegeneration.
Collapse
Affiliation(s)
- L Carlessi
- Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | |
Collapse
|
153
|
Hadjipanayis CG, Van Meir EG. Tumor initiating cells in malignant gliomas: biology and implications for therapy. J Mol Med (Berl) 2009; 87:363-74. [PMID: 19189072 DOI: 10.1007/s00109-009-0440-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 12/17/2008] [Accepted: 01/09/2009] [Indexed: 12/24/2022]
Abstract
A rare subpopulation of cells within malignant gliomas, which shares canonical properties with neural stem cells (NSCs), may be integral to glial tumor development and perpetuation. These cells, also known as tumor initiating cells (TICs), have the ability to self-renew, develop into any cell in the overall tumor population (multipotency), and proliferate. A defining property of TICs is their ability to initiate new tumors in immunocompromised mice with high efficiency. Mounting evidence suggests that TICs originate from the transformation of NSCs and their progenitors. New findings show that TICs may be more resistant to chemotherapy and radiation than the bulk of tumor cells, thereby permitting recurrent tumor formation and accounting for the failure of conventional therapies. The development of new therapeutic strategies selectively targeting TICs while sparing NSCs may provide for more effective treatment of malignant gliomas.
Collapse
Affiliation(s)
- Costas G Hadjipanayis
- Departments of Neurosurgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 33022, USA.
| | | |
Collapse
|
154
|
Lederer CW, Santama N. Neural stem cells: mechanisms of fate specification and nuclear reprogramming in regenerative medicine. Biotechnol J 2009; 3:1521-38. [PMID: 19072908 DOI: 10.1002/biot.200800193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, intense interest in the potential use of neural stem cells (NSC) in the clinical therapy of brain disease and injury has resulted in rapid progress in research on the properties of NSC, their innate and directed differentiation potential and the induced reprogramming of differentiated somatic cells to revert to a pluripotent NSC-like state. The aim of this review is to give an overview of our current operational definitions of the NSC lineage, the growing understanding of extrinsic and intrinsic mechanisms, including heritable but reversible epigenetic chromatin modifications that regulate the maintenance and differentiation of NSC in vivo, and to emphasize ground-breaking efforts of cellular reprogramming with the view to generating patient-specific stem cells for cell replacement therapy. This is set against a summary of current practical procedures for the isolation, research and application of NSC, and of the state of the art in NSC-based regenerative medicine of the nervous system. Both provide the backdrop for the translation of recent findings into innovative clinical applications, with the hope of increasing the safety, efficiency and ethical acceptability of NSC-based therapies in the near future.
Collapse
|
155
|
Ogawa D, Okada Y, Nakamura M, Kanemura Y, Okano HJ, Matsuzaki Y, Shimazaki T, Ito M, Ikeda E, Tamiya T, Nagao S, Okano H. Evaluation of human fetal neural stem/progenitor cells as a source for cell replacement therapy for neurological disorders: Properties and tumorigenicity after long-term in vitro maintenance. J Neurosci Res 2009; 87:307-17. [DOI: 10.1002/jnr.21843] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
156
|
Pizzi R, Rossetti D, Cino G, Marino D, A.L.Vescovi, Baer W. A cultured human neural network operates a robotic actuator. Biosystems 2009; 95:137-44. [DOI: 10.1016/j.biosystems.2008.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 09/21/2008] [Accepted: 09/24/2008] [Indexed: 11/25/2022]
|
157
|
|
158
|
Abstract
Recent advances in stem cell biology have raised expectations that both diseases of, and injuries to, the central nervous system may be ameliorated by cell transplantation. In particular, cell therapy has been studied for inducing efficient remyelination in disorders of myelin, including both the largely pediatric disorders of myelin formation and maintenance and the acquired demyelinations of both children and adults. Potential cell-based treatments of two major groups of disorders include both delivery of myelinogenic replacements and mobilization of residual oligodendrocyte progenitor cells as a means of stimulating endogenous repair; the choice of modality is then predicated upon the disease target. In this review we consider the potential application of cell-based therapeutic strategies to disorders of myelin, highlighting the promises as well as the problems and potential perils of this treatment approach.
Collapse
Affiliation(s)
- Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
159
|
Transplantation and magnetic resonance imaging of canine neural progenitor cell grafts in the postnatal dog brain. J Neuropathol Exp Neurol 2008; 67:954-62. [PMID: 18800012 DOI: 10.1097/nen.0b013e3181875b2f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cellular transplantation in the form of bone marrow has been one of the primary treatments of many lysosomal storage diseases (LSDs). Although bone marrow transplantation can help central nervous system manifestations in some cases, it has little impact in many LSD patients. Canine models of neurogenetic LSDs provide the opportunity for modeling central nervous system transplantation strategies in brains that more closely approximate the size and architectural complexity of the brains of children. Canine olfactory bulb-derived neural progenitor cells (NPCs) isolated from dog brains were expanded ex vivo and implanted into the caudate nucleus/thalamus or cortex of allogeneic dogs. Canine olfactory bulb-derived NPCs labeled with micron-sized superparamagnetic iron oxide particles were detected by magnetic resonance imaging both in vivo and postmortem. Grafts expressed markers of NPCs (i.e. nestin and glial fibrillary acidic protein), but not the neuronal markers Map2ab or beta-tubulin III. The NPCs were from dogs with the LSD mucopolysaccharidosis VII, which is caused by a deficiency of beta-glucuronidase. When mucopolysaccharidosis VII canine olfactory bulb-NPCs that were genetically corrected with a lentivirus vector ex vivo were transplanted into mucopolysaccharidosis VII recipient brains, they were detected histologically by beta-glucuronidase expression in areas identified by antemortem magnetic resonance imaging tracking. These results demonstrate the potential for ex vivo stem cell-based gene therapy and noninvasive tracking of therapeutic grafts in vivo.
Collapse
|
160
|
Immortalization of human neural stem cells with the c-myc mutant T58A. PLoS One 2008; 3:e3310. [PMID: 18836552 PMCID: PMC2561001 DOI: 10.1371/journal.pone.0003310] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Accepted: 08/27/2008] [Indexed: 11/19/2022] Open
Abstract
Human neural stem cells (hNSC) represent an essential source of renewable brain cells for both experimental studies and cell replacement therapies. Their relatively slow rate of proliferation and physiological senescence in culture make their use cumbersome under some experimental and pre-clinical settings. The immortalization of hNSC with the v-myc gene (v-IhNSC) has been shown to generate stem cells endowed with enhanced proliferative capacity, which greatly facilitates the study of hNSCs, both in vitro and in vivo. Despite the excellent safety properties displayed by v-IhNSCs – which do not transform in vitro and are not tumorigenic in vivo – the v-myc gene contains several mutations and recombination elements, whose role(s) and effects remains to be elucidated, yielding unresolved safety concerns. To address this issue, we used a c-myc T58A retroviral vector to establish an immortal cell line (T-IhNSC) from the same hNSCs used to generate the original v-IhNSCs and compared their characteristics with the latter, with hNSC and with hNSC immortalized using c-myc wt (c-IhNSC). T-IhNSCs displayed an enhanced self-renewal ability, with their proliferative capacity and clonogenic potential being remarkably comparable to those of v-IhNSC and higher than wild type hNSCs and c-IhNSCs. Upon growth factors removal, T-IhNSC promptly gave rise to well-differentiated neurons, astrocytes and most importantly, to a heretofore undocumented high percentage of human oligodendrocytes (up to 23%). Persistent growth-factor dependence, steady functional properties, lack of ability to generate colonies in soft-agar colony-forming assay and to establish tumors upon orthotopic transplantation, point to the fact that immortalization by c-myc T58A does not bring about tumorigenicity in hNSCs. Hence, this work describes a novel and continuous cell line of immortalized human multipotent neural stem cells, in which the immortalizing agent is represented by a single gene which, in turn, carries a single and well characterized mutation. From a different perspective, these data report on a safe approach to increase human neural stem cells propagation in culture, without altering their basic properties. These T-IhNSC line provides a versatile model for the elucidation of the mechanisms involved in human neural stem cells expansion and for development of high throughput assays for both basic and translational research on human neural cell development. The improved proclivity of T-IhNSC to generate human oligodendrocytes propose T-IhNSC as a feasible candidate for the design of experimental and, perhaps, therapeutic approaches in demyelinating diseases.
Collapse
|
161
|
Setting the conditions for efficient, robust and reproducible generation of functionally active neurons from adult subventricular zone-derived neural stem cells. Cell Death Differ 2008; 15:1847-56. [PMID: 19011641 DOI: 10.1038/cdd.2008.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although new culture conditions enable homogeneous and long-term propagation of radial glia-like neural stem (NS) cells in monolayer and serum-free conditions, the efficiency of the conversion of NS cells into terminally differentiated, functionally mature neurons is relatively limited and poorly characterized. We demonstrate that NS cells derived from adult mouse subventricular zone robustly develop properties of mature neurons when exposed to an optimized neuronal differentiation protocol. A high degree of cell viability was preserved. At 22 days in vitro, most cells (65%) were microtubule-associated protein 2(+) and coexpressed gamma-aminobutyric acid (GABA), GAD67, calbindin and parvalbumin. Nearly all neurons exhibited sodium, potassium and calcium currents, and 70% of them fired action potentials. These neurons expressed functional GABA(A) receptors, whereas activable kainate, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and N-methyl-D-aspartic acid receptors were present in approximately 80, 30 and 2% of cells, respectively. Antigenic and functional properties were efficiently and reliably reproduced across experiments and cell passages (up to 68). This is the first report showing a consistent and reproducible generation of large amounts of neurons from long-term passaged adult neural stem cells. Remarkably, the neuronal progeny carries a defined set of antigenic, biochemical and functional characteristics that make this system suitable for studies of NS cell biology as well as for genetic and chemical screenings.
Collapse
|
162
|
Zhang H, Wang Y, Zhao Y, Yin Y, Xu Q, Xu Q. Immortalized human neural progenitor cells from the ventral telencephalon with the potential to differentiate into GABAergic neurons. J Neurosci Res 2008; 86:1217-26. [PMID: 18189314 DOI: 10.1002/jnr.21581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Human neural progenitor cells (hNPCs) are believed to have important potential in clinical applications and basic neuroscience research. In the present study, we created a new immortalized human neural cell line, hSN12W-TERT, derived from human fetal ventral telencephalon, using IRES-based retroviral overexpression of human telomerase reverse transcriptase. We showed that after more than 40 passages, hSN12W-TERT cells possess high telomerase activity, maintain a normal diploid karyotype, and retain the characteristics of hNPCs. Under proliferative conditions, these cells remained undifferentiated, expressing the neural progenitor cell markers nestin, vimentin, and Sox2. The cells were able to differentiate into neurons, astrocytes, and oligodendrocytes after a significant decrease in the level of telomerase following withdrawal of growth factors. The neurons were postmitotic and achieved electrophysiologic competence. Furthermore, we showed that most neurons were GABAergic, especially on differentiation induced by bone morphogenetic protein-2 (BMP2). RT-PCR analysis also confirmed that hSN12W-TERT cells expressed mammalian achaete-scute homolog 1 (Mash1) and Dlx2, genes associated with the development of GABAergic cortical interneurons. BMP2 exposure may activate a positive-feedback loop of BMP signaling in hSN12W-TERT cells. Our data indicated that this hSN12W-TERT cell line could be a valuable experimental tool with which to study the regulatory roles of intrinsic and extrinsic factors in human neural stem cell biology and that it would be useful in basic research and in research seeking to discover novel drug targets for clinical candidates.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Cell Biology, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
163
|
Pharmacologic blockade of chloride channel synergistically enhances apoptosis of chemotherapeutic drug-resistant cancer stem cells. Biochem Biophys Res Commun 2008; 373:539-44. [PMID: 18590702 DOI: 10.1016/j.bbrc.2008.06.070] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Accepted: 06/16/2008] [Indexed: 01/06/2023]
Abstract
1,3-Bis(2-chloroethyl)-1-nitrosourea (BCNU) is the most commonly used chemotherapeutic agent in the treatment of human glioblastoma multiforme (GBM). However, BCNU chemotherapy fails due to subpopulations of intrinsic resistant-cells within the tumor mass. In our previous study, we dissociated BCNU-resistant cancer stem cells (CSCs) and observed the over-expression of multiple ion channel genes related to drug efflux. In the present study, we identified chloride intracellular channel 1 (CLIC1) in dissociated-BCNU-resistant CSCs using 2-DE and MALDI-TOF/MS analysis. To develop the specific target therapy of BCNU-resistant CSCs, we evaluated the drug-sensitivity of these CSCs using the chloride channel blocker, 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS). When combined with BCNU, DIDS synergistically increased the apoptotic events of BCNU-resistant CSCs in vitro and augmented BCNU sensitivity ex vivo. These findings suggest that CLIC1 is involved in the resistance of BCNU-resistant CSCs and BCNU/DIDS combined-therapy can provide valuable insight for promoting apoptosis or sensitizing glioblastomas to BCNU chemotherapy.
Collapse
|
164
|
Pax6 promotes neurogenesis in human neural stem cells. Mol Cell Neurosci 2008; 38:616-28. [PMID: 18595732 DOI: 10.1016/j.mcn.2008.05.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/15/2008] [Accepted: 05/09/2008] [Indexed: 12/14/2022] Open
Abstract
During brain embryogenesis, transcription factors drive stem cells towards neuronal fate. Here we show that the transcription factor Pax6 increased in vitro generation of neurons from striatal but not cortical neural stem cells (NSCs), derived from 6 to 9 weeks old human fetuses, without affecting survival and proliferation. Overexpression of mouse Pax6 produced increased numbers of GABA+ and DARPP-32+ (characteristic of striatum) but not glutamate+ neurons (characteristic of cortex). Pax6-overexpressing cells survived and migrated to the same extent as control cells at 1 month after intrastriatal transplantation into newborn rats and generated more neuroblasts. Overexpression of mouse Pax6 in human NSCs also leads to altered levels of lineage-appropriate genes as revealed by Q-PCR. Our data suggest that Pax6 function is conserved between species since its overexpression activates similar genes in mouse and human NSCs. Also, that Pax6 overexpression in striatal NSCs increases the number of neurons but their region-specificity is maintained.
Collapse
|
165
|
Induction of neural stem cell-like cells (NSCLCs) from mouse astrocytes by Bmi1. Biochem Biophys Res Commun 2008; 371:267-72. [PMID: 18439910 DOI: 10.1016/j.bbrc.2008.04.068] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 11/22/2022]
Abstract
Recently, Bmi1 was shown to control the proliferation and self-renewal of neural stem cells (NSCs). In this study, we demonstrated the induction of NSC-like cells (NSCLCs) from mouse astrocytes by Bmi1 under NSC culture conditions. These NSCLCs exhibited the morphology and growth properties of NSCs, and expressed NSC marker genes, including nestin, CD133, and Sox2. In vitro differentiation of NSCLCs resulted in differentiated cell populations containing astrocytes, neurons, and oligodendrocytes. Following treatment with histone deacetylase inhibitors (trichostatin A and valproic acid), the potential of NSCLCs for proliferation, dedifferentiation, and self-renewal was significantly inhibited. Our data indicate that multipotent NSCLCs can be generated directly from astrocytes by the addition of Bmi1.
Collapse
|
166
|
Bhattacharyya A, McMillan E, Wallace K, Tubon TC, Capowski EE, Svendsen CN. Normal Neurogenesis but Abnormal Gene Expression in Human Fragile X Cortical Progenitor Cells. Stem Cells Dev 2008; 17:107-17. [PMID: 18225979 DOI: 10.1089/scd.2007.0073] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human stem and progenitor cells offer an innovative way to study early events in development. An exciting new opportunity for these cells is their application to study the underlying developmental consequences of genetic diseases. Because many diseases, ranging from leukemias to developmental disorders, are caused by single-gene defects, stem and progenitor cells that carry disease-causing genetic mutations are invaluable in understanding and treating disease. We have characterized human neural progenitor (hNPCs) cells that carry a single-gene defect that leads to the neurodevelopmental disorder Fragile X syndrome (FX). A loss-of-function mutation in the FMR1 gene leads to subtle changes in neural development and subsequent mental impairment characteristic of FX. hNPCs were isolated from fetal cortex carrying the FMR1 mutation to determine whether aberrations occur in their proliferation and differentiation. As expected, FX hNPCs have reduced expression of the FMR1 gene product Fragile X mental retardation protein (FMRP), and this decrease is maintained in culture and following differentiation. In contrast to a previously published report, the proliferation of FX hNPCs and their differentiation into neurons is not different from unaffected controls. Although the early development of FX hNPCs is essentially normal, microarray analysis reveals novel changes in the expression of signal transduction genes in FX hNPCs. Therefore, hNPCs have intrinsic characteristics that can be investigated to further our understanding and potential treatment of developmental disorders such as FX.
Collapse
|
167
|
Production and characterization of immortal human neural stem cell line with multipotent differentiation property. Methods Mol Biol 2008; 438:103-21. [PMID: 18369753 DOI: 10.1007/978-1-59745-133-8_10] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We document the protocols and methods for the production of immortalized cell lines of human neural stem cells from the human fetal central nervous system (CNS) cells by using a retroviral vector encoding v-myc oncogene. One of the human neural stem cell lines (HB1.F3) was found to express nestin and other specific markers for human neural stem cells, giving rise to three fundamental cell types of the CNS: neurons, astrocytes, and oligodendrocytes. After transplantation into the brain of mouse model of stroke, implanted human neural stem cells were observed to migrate extensively from the site of implantation into other anatomical sites and to differentiate into neurons and glial cells.
Collapse
|
168
|
Novel and immortalization-based protocols for the generation of neural CNS stem cell lines for gene therapy approaches. Methods Mol Biol 2008. [PMID: 18369767 DOI: 10.1007/978-1-59745-133-8_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Transplantation of neural cells engineered to produce growth factors or molecules with antitumor effects have the potential of grafted cells to be used as vectors for protein delivery in animal models of diseases. In this context, neural stem cells (NSCs), since their identification, have been considered an attractive subject for therapeutic applications to the damaged brain. NSCs have been shown to include attributes important for potential successful ex vivo gene therapy approaches: they show extensive in vitro expansion and, in some cases, a particular tropism toward pathological brain areas. Clearly, the challenges for future clinical development of this approach are in the definition of the most appropriate stem cells for a given application, what genes or chemicals can be delivered, and what diseases are suitable targets. Ideally, NSC lines should be homogeneous and well characterized in terms of their in vitro stability and grafting capacity. We discuss two possible approaches to produce homogeneous and stable progenitor and NSC lines that exploit an oncogene-based immortalization, or, in the second case, a novel protocol for growth factor expansion of stem cells with radial glia-like features. Furthermore, we describe the use of retroviral particles for genetic engineering.
Collapse
|
169
|
Lu H, Li M, Song T, Qian Y, Xiao X, Chen X, Zhang P, Feng X, Parker T, Liu Y. Retrovirus delivered neurotrophin-3 promotes survival, proliferation and neuronal differentiation of human fetal neural stem cells in vitro. Brain Res Bull 2008; 77:158-64. [PMID: 19875351 DOI: 10.1016/j.brainresbull.2008.02.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/10/2008] [Accepted: 02/18/2008] [Indexed: 12/21/2022]
Abstract
Poor survival and insufficient neuronal differentiation are the main obstacles to neural stem cell (NSC) transplantation therapy. Genetic modification of NSCs with neurotrophins is considered a promising approach to overcome these difficulties. In this study, the effects on survival, proliferation and neuronal differentiation of human fetal NSCs (hfNSCs) were observed after infection by a neurotrophin-3 (NT-3) recombinant retrovirus. The hfNSCs, from 12-week human fetal brains formed neurospheres, expressed the stem cell marker nestin and differentiated into the three main cell types of the nervous system. NT-3 recombinant retrovirus (Retro-NT-3) infected hfNSCs efficiently expressed NT-3 gene for at least 8 weeks, presented an accelerated proliferation, and therefore produced an increased number of neurospheres and after differentiation in vitro, contained a higher percentage of neuronal cells. Eight weeks after infection, 37.9+/-4.2% of hfNSCs in the Retro-NT-3 infection group expressed the neuronal marker, this was significantly higher than the control and mock infection groups. NT-3 transduced hfNSCs also displayed longer protruding neurites compared with other groups. Combined these results demonstrate that NT-3 modification promote the survival/proliferation, neuronal differentiation and growth of neurites of hfNSCs in vitro. This study proposes recombinant retrovirus mediated NT-3 modification may provide a promising means to resolve the poor survival and insufficient neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Haixia Lu
- Institute of Neurobiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Therapeutic strategies for Parkinson's disease: the ancient meets the future--traditional Chinese herbal medicine, electroacupuncture, gene therapy and stem cells. Neurochem Res 2008; 33:1956-63. [PMID: 18404373 DOI: 10.1007/s11064-008-9691-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 03/28/2008] [Indexed: 10/22/2022]
Abstract
In China, it has been estimated that there are more than 2.0 million people suffering from Parkinson's disease, which is currently becoming one of the most common chronic neurodegenerative disorders during recent years. For many years, scientists have struggled to find new therapeutic approaches for this disease. Since 1994, our research group led by Drs. Ji-Sheng Han and Xiao-Min Wang of Neuroscience Research Institute, Peking University has developed several prospective treatment strategies for the disease. These studies cover the traditional Chinese medicine-herbal formula or acupuncture, and modern technologies such as gene therapy or stem cell replacement therapy, and have achieved some original results. It hopes that these data may be beneficial for the research development and for the future clinical utility for treatment of Parkinson's disease.
Collapse
|
171
|
Abstract
New fundamental results on stem cell biology have been obtained in the past 15 years. These results allow us to reinterpret the functioning of the cerebral tissue in health and disease. Proliferating stem cells have been found in the adult brain, which can be involved in postinjury repair and can replace dead cells under specific conditions. Numerous genomic mechanisms controlling stem cell proliferation and differentiation have been identified. The involvement of stem cells in the genesis of malignant tumors has been demonstrated. Neural stem cell tropism toward tumors has been shown. These findings suggest new lines of research on brain functioning and development. Stem cells can be used to develop radically new treatments of neurodegenerative and cancer diseases of the brain.
Collapse
|
172
|
Ayuso-Sacido A, Roy NS, Schwartz TH, Greenfield JP, Boockvar JA. Long-term expansion of adult human brain subventricular zone precursors. Neurosurgery 2008; 62:223-9; discussion 229-31. [PMID: 18300911 DOI: 10.1227/01.neu.0000311081.50648.4c] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Many common neurosurgical procedures, including anterior temporal lobectomy and endoscopic ventricular puncture, allow neurosurgeons to retrieve portions of the germinal subventricular zone (SVZ). Isolation and maintenance of precursor cells from this zone can be used for hypothesis-driven experiments with a goal of improving our understanding of the basic mechanisms of central nervous system injury or disease and the potential of cell-based therapies to treat them. This article details our ability to reliably harvest, isolate, characterize, and maintain normal adult human brain SVZ precursor cells. METHODS Normal SVZ specimens were retrieved as part of anterior temporal lobe resections during planned epilepsy surgery. Dissociated SVZ specimens were plated and incubated in epidermal growth factor and basic fibroblast growth factor for more than 1 year to select for and expand normal neural precursor cells. RESULTS Self-renewal and immunocytochemical experiments proved the feasibility of long-term expansion of a slowly dividing nestin+, vimentin+, and glial fibrillary acidic protein-positive astrocyte capable of generating new neurons and glia. These mitotically active bipotent human precursors generated a large number of progeny and possessed significant self-renewal capacity, demonstrated by their ability to generate neurospheres. Cryopreservation was reliable with no loss of the precursor phenotype. CONCLUSION We have adapted techniques to allow for the isolation and long-term propagation of human adult neural precursors that are capable of generating both neurons and astrocytes in vitro. We have exploited the cell's self-renewal capacity to significantly and consistently expand human neural precursor cells for as long as 20 months. These findings suggest that cells derived from the SVZ during routine surgery may provide a renewable source of human neural precursor cells to study the biological mechanism of central nervous system disease or for application in cell-based human transplantation paradigms.
Collapse
Affiliation(s)
- Angel Ayuso-Sacido
- Laboratory for Translational Stem Cell Research, Department of Neurological Surgery, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
173
|
Mokrý J, Karbanová J, Filip S, Čížková D, Pazour J, English D. Phenotypic and Morphological Characterization of In Vitro Oligodendrogliogenesis. Stem Cells Dev 2008; 17:333-41. [DOI: 10.1089/scd.2007.0091] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- J. Mokrý
- Department of Histology and Embryology, Charles University in Prague, Faculty of Medicine in Hradec Králové, 50038 Hradec Králové, Czech Republic
| | - J. Karbanová
- Department of Histology and Embryology, Charles University in Prague, Faculty of Medicine in Hradec Králové, 50038 Hradec Králové, Czech Republic
| | - S. Filip
- Department of Histology and Embryology, Charles University in Prague, Faculty of Medicine in Hradec Králové, 50038 Hradec Králové, Czech Republic
- Department of Oncology and Radiotherapy, Faculty Hospital, 50005 Hradec Králové, Czech Republic
| | - D. Čížková
- Department of Histology and Embryology, Charles University in Prague, Faculty of Medicine in Hradec Králové, 50038 Hradec Králové, Czech Republic
| | - J. Pazour
- Department of Histology and Embryology, Charles University in Prague, Faculty of Medicine in Hradec Králové, 50038 Hradec Králové, Czech Republic
| | - D. English
- Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL 33612
| |
Collapse
|
174
|
Walton RM, Wolfe JH. In vitro growth and differentiation of canine olfactory bulb-derived neural progenitor cells under variable culture conditions. J Neurosci Methods 2008; 169:158-67. [PMID: 18261803 PMCID: PMC2377027 DOI: 10.1016/j.jneumeth.2007.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Revised: 12/05/2007] [Accepted: 12/05/2007] [Indexed: 10/22/2022]
Abstract
The dog serves as a large animal model for multiple neurologic diseases that may potentially benefit from neural progenitor cell (NPC) transplantation. In the adult brain, multipotent NPCs reside in the subventricular zone and its rostral and caudal extensions into the olfactory bulb and hippocampus. The olfactory bulb represents a surgically accessible site for obtaining cells for autologous NPC transplantation. To model conditions that would occur for ex vivo gene therapy in the postnatal brain, NPCs were isolated from the canine olfactory bulb, expanded ex vivo under different culture conditions, and compared quantitatively for growth and immunophenotype. Under standard growth conditions, canine olfactory bulb-derived NPCs (OB-cNPCs) could be expanded nearly 500-fold in the time evaluated. Canine OB-cNPCs grown on poly-d-lysine (PDL) or on PDL-fibronectin had similar growth rates, whereas supplementation with leukemia inhibitory factor (LIF) resulted in significantly slower growth. However, when OB-cNPC cultures were grown on PDL-fibronectin or PDL supplemented with LIF, a greater proportion of cells with neuronal markers were generated upon differentiation.
Collapse
Affiliation(s)
- Raquel M. Walton
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania; and Stokes Research Institute, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - John H. Wolfe
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania; and Stokes Research Institute, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| |
Collapse
|
175
|
Dirks PB. Brain tumour stem cells: the undercurrents of human brain cancer and their relationship to neural stem cells. Philos Trans R Soc Lond B Biol Sci 2008; 363:139-52. [PMID: 17309866 PMCID: PMC2605491 DOI: 10.1098/rstb.2006.2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Conceptual and technical advances in neural stem cell biology are being applied to the study of human brain tumours. These studies suggest that human brain tumours are organized as a hierarchy and are maintained by a small number of tumour cells that have stem cell properties. Most of the bulk population of human brain tumours comprise cells that have lost the ability to initiate and maintain tumour growth. Although the cell of origin for human brain tumours is uncertain, recent evidence points towards the brain's known proliferative zones. The identification of brain tumour stem cells has important implications for understanding brain tumour biology and these cells may be critical cellular targets for curative therapy.
Collapse
Affiliation(s)
- Peter B Dirks
- Division of Neurosurgery and Program in Developmental Biology, University of Toronto, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada.
| |
Collapse
|
176
|
Denham M, Conley B, Olsson F, Cole TJ, Mollard R. Stem cells: an overview. ACTA ACUST UNITED AC 2008; Chapter 23:Unit 23.1. [PMID: 18228471 DOI: 10.1002/0471143030.cb2301s28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stem cells are specialized cells that possess a capacity to undergo self-renewal while at the same time having the ability to give rise to at least one or more differentiated or mature cell type. They therefore represent a fundamental cornerstone during the life of all vertebrates, playing central roles in the production of new and replacement cells for tissues during development and homeostasis, including repair following disease or injury. This unit is a review of stem cells, their roles in development, and their potentials as therapeutic agents.
Collapse
|
177
|
McAdoo DJ, Wu P. Microdialysis in central nervous system disorders and their treatment. Pharmacol Biochem Behav 2008; 90:282-96. [PMID: 18436292 DOI: 10.1016/j.pbb.2008.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 02/27/2008] [Accepted: 03/05/2008] [Indexed: 01/17/2023]
Abstract
Central nervous system (CNS) insults elevate endogenous toxins and alter levels of indicators of metabolic disorder. These contribute to neurotrauma, neurodegenerative diseases and chronic pain and are possible targets for pharmaceutical treatment. Microdialysis samples substances in the extracellular space for chemical analysis. It has demonstrated that toxic levels of glutamate are released and that toxic levels of the reactive species O(2)(-), H(2)O(2), HO. NO and HOONO are generated upon CNS injury. Agent administration by microdialysis can also help elucidate mechanisms of damage and protection, and to identify targets for clinical application. Microdialysis sampling indicates that circuits descending from the brain to the spinal cord transmit and modulate pain signals by releasing neurotransmitter amines and amino acids. Efforts are under way to develop microdialysis into a technique for intensive care monitoring and predicting outcomes of brain insults. Finally, microdialysis sampling has demonstrated in vivo elevation of glial cell line-derived neurotrophic factor following grafting of primed fetal human neural stem cells into brain-injured rats, the first in vivo demonstration of the release of a neurotrophic factor by grafted stem cells. This increased release correlated with significantly improved spatial learning and memory.
Collapse
Affiliation(s)
- David J McAdoo
- Department of Neurosciences and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, United States.
| | | |
Collapse
|
178
|
Isolation, characterization and gene modification of fetal neural stem/progenitor cells from cynomolgus monkey. Neuroreport 2008; 19:419-24. [DOI: 10.1097/wnr.0b013e3282f76136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
179
|
Abstract
OBJECTIVES The aim of this review is to provide an overview of the fundamental features of the neurosphere assay (NSA), which was initially described in 1992, and has since been used not only to detect the presence of stem cells in embryonic and adult mammalian neural tissues, but also to study their characteristics in vitro. Implicit in this review is a detailed examination of the limitations of the NSA, and how this assay is most accurately and appropriately used. Finally we will point out criteria that should be challenged to design alternative ways to overcome the limits of this assay. METHODS NSA is used to isolate putative neural stem cells (NSCs) from the central nervous system (CNS) and to demonstrate the critical stem cell attributes of proliferation, extensive self-renewal and the ability to give rise to a large number of differentiated and functional progeny. Nevertheless, the capability of neural progenitor cells to form neurospheres precludes its utilisation to accurately quantify bona fide stem cell frequency based simply on neurosphere numbers. New culture conditions are needed to be able to distinguish the activity of progenitor cells from stem cells. CONCLUSION A commonly used, and arguably misused, methodology, the NSA has provided a wealth of information on precursor activity of cells derived from the embryonic through to the aged CNS. Importantly, the NSA has contributed to the demise of the 'no new neurogenesis' dogma, and the beginning of a new era of CNS regenerative medicine. Nevertheless, the interpretations arising from the utilisation of the NSA need to take into consideration its limits, so as not to be used beyond its specificity and sensitivity.
Collapse
Affiliation(s)
- Loic P Deleyrolle
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Rodney L Rietze
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Brent A Reynolds
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
180
|
Kang MK, Hur BI, Ko MH, Kim CH, Cha SH, Kang SK. Potential identity of multi-potential cancer stem-like subpopulation after radiation of cultured brain glioma. BMC Neurosci 2008; 9:15. [PMID: 18230189 PMCID: PMC2266936 DOI: 10.1186/1471-2202-9-15] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 01/30/2008] [Indexed: 01/08/2023] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most frequently encountered brain cancer. Although the existence of cancer stem cells in GBM has been previously established, there is little evidence to explain the difference between cancer stem cells and radio-resistant cells in GBM. In an effort to increase our understanding of whether cellular radio-resistance is a characteristic associated with cancer stem cells, we developed a dissociated cell system of subpopulations derived from GBM, and demonstrated radiotherapy resistance therein. Results The radio-resistant cancer cell subpopulations of GBM abundantly express CD133, CD117, CD71, and CD45 surface markers, and these radio-resistant cancer cell subpopulations have the capacity for extensive proliferation, self-renewal, and pluripotency. These radio-resistant cancer subpopulations have been shown to initiate tumorigenesis when transplanted into SCID mouse brains. Moreover, these tumors evidenced highly peculiar nest-like shapes harboring both vascular and cancerous tissue structures, which expressed the blood vessel specific marker, the von Willebrand factor. Accordingly, subpopulations of radio-resistant cells in GBM have been shown to be very similar to hematopoietic stem cells (HSCs) in the circulating blood. This similarity may contribute to increased tumor growth and GBM recurrence. Conclusion The results of the present study provide further evidence for radio resistant subpopulations of cancer stem cells in GBM. Also, our results will assist in the identification and characterization of cancer stem cell populations in glioma, and will help to improve the therapeutic outcomes of GBM.
Collapse
Affiliation(s)
- Mi K Kang
- Department of Physiology, College of Medicine, Pusan National University, Busan, Korea.
| | | | | | | | | | | |
Collapse
|
181
|
|
182
|
Kang MK, Kang SK. Tumorigenesis of chemotherapeutic drug-resistant cancer stem-like cells in brain glioma. Stem Cells Dev 2008; 16:837-47. [PMID: 17999604 DOI: 10.1089/scd.2007.0006] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequently occurring brain cancer. Although the existence of cancer stem cells (CSCs) in GBM has been established, there is little evidence to explain the link between CSCs and chemoresistance. In this study, we developed a dissociated cell system of human GBM cells, A172 and established GBM2 cells, that have shown resistance to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). After exposure to a lethal dose of BCNU, the small population of GBM cancer cells survived and proliferated, as opposed to direct inhibition of the apoptosis and activation of the proliferation signal. Also, these cells contained subpopulations of stem-like cells, expressing CD133, CD117, CD90, CD71, and CD45 cell-surface markers, and had the capacity for multipotency. Moreover, we observed that BCNU-resistant subpopulations derived from GBM cancer cells can be grown to tumors when transplanted into severe combined immunodeficient (SCID) mouse brain. These results demonstrated that BCNU-resistant subpopulations derived from GBM have cancer stem-like cell properties. These findings provide further evidence that CSCs in GBM display chemotherapeutic drug resistance. Hopefully, it will be possible to improve the therapeutic outcome of GBM, leading to better anticancer strategies.
Collapse
Affiliation(s)
- Mi-Kyung Kang
- Department of Physiology, College of Medicine, Pusan National University, Busan 602-739, South Korea
| | | |
Collapse
|
183
|
Willing AE, Garbuzova-Davis S, Sanberg PR, Saporta S. Routes of stem cell administration in the adult rodent. Methods Mol Biol 2008; 438:383-401. [PMID: 18369773 DOI: 10.1007/978-1-59745-133-8_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Stem cell transplantation to replace damaged tissue or correct metabolic disease holds the promise of helping a myriad of human afflictions. Although a great deal of attention has focused on pluripotent stem cells derived from embryos, adult stem cells have been described in a variety of tissues, and they likely will prove to be as beneficial as embryonic stem cells in cell replacement therapy and control of inbred errors of metabolism. We describe methods by which stem cells can be introduced into the nervous system, although the techniques are applicable to any portion of the body to be targeted or any cell that may be used for cell therapy. The first and most straight-forward method is introduction of stem cells directly into the brain parenchyma. The second, which in our hands has proven to be superior in some instances, is introduction of the stem cells into the circulatory system.
Collapse
Affiliation(s)
- Alison E Willing
- Center for Aging and Brain Repair Cell Biology, University of South Florida College of Medicine, Tampa, FL, USA
| | | | | | | |
Collapse
|
184
|
Gritti A, Galli R, Vescovi AL. Clonal analyses and cryopreservation of neural stem cell cultures. Methods Mol Biol 2008; 438:173-84. [PMID: 18369757 DOI: 10.1007/978-1-59745-133-8_14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The discovery of stem cell populations in the adult central nervous system (CNS) that continually produce neurons and glial cells, and the hypothesis that they could contribute to neural plasticity/repair, has opened new and exciting areas of research in basic cell biology and regenerative medicine. The success of these studies relies on understanding the functional features and the normal fate of neural stem cells (NSCs) in vivo as well on the development of in vitro culture conditions enabling isolation, extensive propagation, and rigorous characterization of the "putative" NSCs. The neurosphere assay (NSA) has emerged as a valuable tool for isolating embryonic and adult CNS stem cells and for studying their biology. However, because this assay may select and expand a heterogeneous stem/progenitor cell population, rigorous clonal and serial subcloning analyses are required to detect and document stem cell activity and to unequivocally identify bona fide stem cells. We illustrate and discuss methods for the isolation, propagation, cryopreservation, and functional characterization of NSCs, focusing on the essential issue of their clonogenic capacity.
Collapse
Affiliation(s)
- Angela Gritti
- Telethon Institute for Gene Therapy, DIBIT-San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
185
|
Maurer MH, Thomas C, Bürgers HF, Kuschinsky W. Transplantation of adult neural progenitor cells transfected with vascular endothelial growth factor rescues grafted cells in the rat brain. Int J Biol Sci 2007; 4:1-7. [PMID: 18167530 PMCID: PMC2140152 DOI: 10.7150/ijbs.4.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 11/30/2007] [Indexed: 01/12/2023] Open
Abstract
Growth factors are currently evaluated as therapeutics in stroke and neurodegeneration. Besides direct neurotrophic effects, they promote proliferation, survival, and differentiation of both transplanted and endogenous neural precursor cells (NPCs). In the current study, we investigated whether NPCs expressing Vascular Endothelial Growth Factor VEGF-A165 are a useful vehicle for growth factor delivery after transplantation into the caudate putamen of the rat brain. We found an increased survival of adenovirally transfected NPCs after 11 days, but not after 24 hours or 4 days. Additional brain immunohistochemistry revealed increased expression of the endothelial cell marker PECAM-1 (CD31) after 24 hours, 4 day, and 11 days after transplantation. In conclusion, we show that the graft itself is a useful vehicle for growth factor delivery, promoting the survival of NPCs. Moreover, transplantation of VEGF-expressing NPCs supports angiogenesis in the brain, which may contribute to potential brain repair.
Collapse
Affiliation(s)
- Martin H Maurer
- Department of Physiology, Pathophysiology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
186
|
Nelson AD, Suzuki M, Svendsen CN. A high concentration of epidermal growth factor increases the growth and survival of neurogenic radial glial cells within human neurosphere cultures. Stem Cells 2007; 26:348-55. [PMID: 18032704 DOI: 10.1634/stemcells.2007-0299] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Human neural progenitor cells (hNPC) isolated from the fetal cortex can be expanded as aggregates of cells termed neurospheres. Traditional methods have used 20 ng/ml epidermal growth factor (EGF) to drive the proliferation of these cells. Here, we show that 100 ng/ml EGF can significantly increase growth rates of hNPC at later passages. This was through increased survival of dividing cells rather than increased proliferation and associated with prolonged activation of ErbB2 and phosphorylated Akt. High EGF also resulted in a larger proportion of elongated "radial glial"-like cells within the growing neurospheres and increased expression of the radial glial markers. The number of new neurons generated from cultures maintained in 100 ng/ml EGF was significantly higher than from 20 ng/ml EGF. Thus, high concentrations of EGF increase the survival of a highly neurogenic human radial glial cell.
Collapse
Affiliation(s)
- Aaron D Nelson
- Department of Anatomy and Neurology, Waisman Center, University of Wisconsin-Madison, 1500 Highland Avenue, Madison, Wisconsin 53705-2280, USA
| | | | | |
Collapse
|
187
|
Neri M, Maderna C, Cavazzin C, Deidda-Vigoriti V, Politi LS, Scotti G, Marzola P, Sbarbati A, Vescovi AL, Gritti A. Efficient in vitro labeling of human neural precursor cells with superparamagnetic iron oxide particles: relevance for in vivo cell tracking. Stem Cells 2007; 26:505-16. [PMID: 17975226 DOI: 10.1634/stemcells.2007-0251] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Recent studies have raised appealing possibilities of replacing damaged or lost neural cells by transplanting in vitro-expanded neural precursor cells (NPCs) and/or their progeny. Magnetic resonance (MR) tracking of superparamagnetic iron oxide (SPIO)-labeled cells is a noninvasive technique to track transplanted cells in longitudinal studies on living animals. Murine NPCs and human mesenchymal or hematopoietic stem cells can be efficiently labeled by SPIOs. However, the validation of SPIO-based protocols to label human neural precursor cells (hNPCs) has not been extensively addressed. Here, we report the development and validation of optimized protocols using two SPIOs (Sinerem and Endorem) to label human hNPCs that display bona fide stem cell features in vitro. A careful titration of both SPIOs was required to set the conditions resulting in efficient cell labeling without impairment of cell survival, proliferation, self-renewal, and multipotency. In vivo magnetic resonance imaging (MRI) combined with histology and confocal microscopy indicated that low numbers (5 x 10(3) to 1 x 10(4)) of viable SPIO-labeled hNPCs could be efficiently detected in the short term after transplantation in the adult murine brain and could be tracked for at least 1 month in longitudinal studies. By using this approach, we also clarified the impact of donor cell death to the MR signal. This study describes a simple protocol to label NPCs of human origin using SPIOs at optimized low dosages and demonstrates the feasibility of noninvasive imaging of labeled cells after transplantation in the brain; it also evidentiates potential limitations of the technique that have to be considered, particularly in the perspective of neural cell-based clinical applications.
Collapse
Affiliation(s)
- Margherita Neri
- aStem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Hong S, Kang UJ, Isacson O, Kim KS. Neural precursors derived from human embryonic stem cells maintain long-term proliferation without losing the potential to differentiate into all three neural lineages, including dopaminergic neurons. J Neurochem 2007; 104:316-24. [PMID: 17944878 DOI: 10.1111/j.1471-4159.2007.04952.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human embryonic stem (hES) cells have the ability to renew themselves and differentiate into multiple cell types upon exposure to appropriate signals. In particular, the ability of hES cells to differentiate into defined neural lineages, such as neurons, astrocytes, and oligodendrocytes, is fundamental to developing cell-based therapies for neurodegenerative disorders and studying developmental mechanisms. However, the utilization of hES cells for basic and applied research is hampered by the lack of well-defined methods to maintain their self-renewal and direct their differentiation. Recently we reported that neural precursor (NP) cells derived from mouse ES cells maintained their potential to differentiate into dopaminergic (DA) neurons after significant expansion in vitro. We hypothesized that NP cells derived from hES cells (hES-NP) could also undergo the same in vitro expansion and differentiation. To test this hypothesis, we passaged hES-NP cells and analyzed their proliferative and developmental properties. We found that hES-NP cells can proliferate approximately 380 000-fold after in vitro expansion for 12 weeks and maintain their potential to generate Tuj1+ neurons, GFAP+ astrocytes, and O4+ oligodendrocytes as well as tyrosine hydroxylase-positive (TH+) DA neurons. Furthermore, TH+ neurons originating from hES-NP cells expressed other midbrain DA markers, including Nurr1, Pitx3, Engrail-1, and aromatic l-amino acid decarboxylase, and released significant amounts of DA. In addition, hES-NP cells maintained their developmental potential through long-term storage (over 2 years) in liquid nitrogen and multiple freeze-thaw cycles. These results demonstrate that hES-NP cells have the ability to provide an expandable and unlimited human cell source that can develop into specific neuronal and glial subtypes.
Collapse
Affiliation(s)
- Sunghoi Hong
- Udall Parkinson's Disease Research Center of Excellence, McLean Hospital, Harvard Medical School, Belmont, Massachusetts 02478, USA
| | | | | | | |
Collapse
|
189
|
Paynter SJ. Principles and practical issues for cryopreservation of nerve cells. Brain Res Bull 2007; 75:1-14. [PMID: 18158089 DOI: 10.1016/j.brainresbull.2007.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 08/23/2007] [Indexed: 11/26/2022]
Abstract
Nerve cells isolated from the brain have a number of research and clinical applications, not the least of which is their transplantation to patients with Parkinson's disease. Neural primary and precursor cells of several areas of the brain are potential candidates for transplantation and research. However, supply of suitable tissue is one of the major problems associated with the widespread application of such techniques. The ability to store such tissue for prolonged periods would greatly alleviate this problem. Cryopreservation allows indefinite storage, provided the storage temperature is sufficiently low. Whilst many of the potentially usable cell types have been shown to be capable of surviving cryopreservation to some degree, survival post-thaw needs to be considerably improved. Cryopreservation techniques applied to date are mostly crude and often adopted from those used for unrelated cell types. Studies involving cryopreservation of primary neural cells and stem cells are reviewed, the basic principles of cryopreservation explained and suggestions made for improvements to the low temperature storage of these cells.
Collapse
Affiliation(s)
- Sharon J Paynter
- Department of Obstetrics & Gynaecology, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
190
|
Wang L, Martin DR, Baker HJ, Zinn KR, Kappes JC, Ding H, Gentry AS, Harper S, Snyder EY, Cox NR. Neural progenitor cell transplantation and imaging in a large animal model. Neurosci Res 2007; 59:327-40. [PMID: 17897743 DOI: 10.1016/j.neures.2007.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 07/10/2007] [Accepted: 08/06/2007] [Indexed: 01/19/2023]
Abstract
To evaluate neural stem/progenitor cell (NPC) transplantation therapy in cat models of neurodegenerative diseases, we have isolated, expanded and characterized feline NPCs (fNPCs) from normal fetal cat brain. Feline NPCs responsive to both human epidermal growth factor (hEGF) and human fibroblast growth factor 2 (hFGF2) proliferated as neurospheres, which were able to differentiate to neurons and glial cells. The analysis of growth factors indicated that both hEGF and hFGF2 were required for proliferation of fNPCs. In contrast to the effect on human NPCs, human leukemia inhibitory factor (hLIF) enhanced differentiation of fNPCs. Expanded fNPCs were injected into the brains of normal adult cats. Immunohistochemical analysis showed that the majority of transplanted cells were located adjacent to the injection site and some fNPCs differentiated into neurons. The survival of transplanted fNPCs over time was monitored using non-invasive bioluminescent imaging technology. This study provided the first evidence of allotransplantation of fNPCs into feline CNS. Cats have heterogeneous genetic backgrounds and possess neurological diseases that closely resemble analogous human diseases. The characterization of fNPCs and exploration of non-invasive bioluminescent imaging to track transplanted cells in this study will allow evaluation of NPC transplantation therapy using feline models of human neurological diseases.
Collapse
Affiliation(s)
- Lei Wang
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849-5525, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
De Filippis L, Foglieni C, Silva S, Vescovi AL, Lusso P, Malnati MS. Differentiated human neural stem cells: a new ex vivo model to study HHV-6 infection of the central nervous system. J Clin Virol 2007; 37 Suppl 1:S27-32. [PMID: 17276364 DOI: 10.1016/s1386-6532(06)70008-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND HHV-6 is the etiologic agent of exanthem subitum, a pediatric illness that may be associated with clinical and laboratory signs of central nervous system involvement. The absence of suitable experimental models has so far hampered the elucidation of the mechanisms of HHV-6-mediated neural cell damage. Recently, the growing knowledge in neurobiology has permitted the establishment of long-term cultures of human neural stem cells (hNSC) that, by virtue of their self-renewal capacity and multipotentiality, provide a valuable tool for the study of neurodegenerative disorders. OBJECTIVES AND STUDY DESIGN We studied the effects of HHV-6 infection in differentiated cultures of hNSC derived from the telencephalic and diencephalic regions of a 13.5 week post conception (pcw) fetal brain. The prototypic HHV-6 strain GS (subgroup A) was used. RESULTS hNSC were differentiated ex vivo to obtain mixed cultures encompassing astrocytes, neurons and oligodendrocytes. These differentiated hNSC cultures were found to be susceptible to productive HHV-6A infection, resulting in the formation of syncytia associated with phenotypic alterations. CONCLUSION These results demonstrate that hNSC may provide a physiologically relevant model to investigate the pathogenic role of HHV-6 in central nervous system disorders.
Collapse
Affiliation(s)
- Lidia De Filippis
- Stem Cell Research Institute, DIBIT, San Raffaele Scientific Institute 20132 Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
192
|
Invernici G, Cristini S, Madeddu P, Brock S, Spillmann F, Bernasconi P, Cappelletti C, Calatozzolo C, Fascio U, Bisleri G, Muneretto C, Alessandri G, Parati EA. Human adult skeletal muscle stem cells differentiate into cardiomyocyte phenotype in vitro. Exp Cell Res 2007; 314:366-76. [PMID: 17888423 DOI: 10.1016/j.yexcr.2007.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 07/13/2007] [Accepted: 08/08/2007] [Indexed: 10/22/2022]
Abstract
Cell transplantation to repair or regenerate injured myocardium is a new frontier in the treatment of cardiovascular disease. Most studies on stem cell transplantation therapy in both experimental heart infarct and in phase-I human clinical trials have focused on the use of undifferentiated stem cells. Based on our previous observations demonstrating the presence of multipotent progenitor cells in human adult skeletal muscle, in this study we investigated the capacity of these progenitors to differentiate into cardiomyocytes. Here we show an efficient protocol for the cardiomyogenic differentiation of human adult skeletal muscle stem cells in vitro. We found that treatment with Retinoic Acid directed cardiomyogenic differentiation of skeletal muscle stem cells in vitro. After Retinoic Acid treatment, cells expressed cardiomyocyte markers and acquired spontaneous contraction. Functional assays exhibited cardiac-like response to increased extracellular calcium. When cocultured with mouse cardiomyocytes, Retinoic Acid-treated skeletal muscle stem cells expressed connexin43 and when transplanted into ischemic heart were detectable even 5 weeks after injection. Based on these results, we can conclude that human adult skeletal muscle stem cells, if opportunely treated, can transdifferentiate into cells of cardiac lineage and once injected into infarcted heart can integrate, survive in cardiac tissue and improve the cardiac function.
Collapse
Affiliation(s)
- Gloria Invernici
- Neurobiology and Neuroregenerative Therapies Unit, Fondazione IRCCS Neurological Institute C. Besta, Milan #20133, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Anderson L, Caldwell MA. Human neural progenitor cell transplants into the subthalamic nucleus lead to functional recovery in a rat model of Parkinson’s disease. Neurobiol Dis 2007; 27:133-40. [PMID: 17587588 DOI: 10.1016/j.nbd.2007.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 03/21/2007] [Accepted: 03/27/2007] [Indexed: 01/03/2023] Open
Abstract
Despite the success of foetal nigral transplantation for the treatment of Parkinson's disease, supply limitations of tissue means that alternative sources must be found. Transplantation of human neural progenitor cells (HNPCs) may offer a solution, however few studies have shown functional recovery in animal models of PD without cell modification. Here we show that unmodified HNPC grafted into the subthalamic nucleus (STN) show excellent survival of up to 5 months and induce significant functional recovery following amphetamine-induced rotations within 4 weeks. For the first time we also show that HNPCs, which remain in an immature nestin-positive state, produce VEGF in vivo allowing further modification of the host brain. This suggests that even in the absence of cell replacement strategies utilising immature progenitor cells could be of real therapeutic value.
Collapse
Affiliation(s)
- Lucy Anderson
- Centre for Brain Repair and Department of Clinical Neurosciences, University Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK
| | | |
Collapse
|
194
|
Ubiali F, Nava S, Nessi V, Frigerio S, Parati E, Bernasconi P, Mantegazza R, Baggi F. Allorecognition of human neural stem cells by peripheral blood lymphocytes despite low expression of MHC molecules: role of TGF-beta in modulating proliferation. Int Immunol 2007; 19:1063-74. [PMID: 17660500 DOI: 10.1093/intimm/dxm079] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells (NSCs) transplantation has been proposed as a means of restoring damaged brain tissue, a possibility rendered more likely by reports of low NSCs immunogenicity in various experimental models because of low expression of MHC class I and II as well as co-stimulatory molecules. We investigated the immunogenicity of a human NSC line grown in normal culture conditions and in the presence of pro-inflammatory cytokines IFN-gamma and tumor necrosis factor alpha by one-way mixed lymphocyte reaction (MLR) experiments with peripheral blood lymphocytes from eight HLA-incompatible donors. NSCs stimulated lymphocyte proliferation in almost all donors tested, with stimulation indices in the range of the low-end distribution curve of MLR between donors. The healthy subject that gave negative MLR results was the best compatible donor with respect to NSC haplotype. Since we observed low MLR responses overall, we studied if NSCs might exert any immunomodulatory activity. We detected transcription and release of the immunomodulatory molecule transforming growth factor beta (TGF-beta)-1; moreover, the addition of TGF-beta1 in MLR experiments down-regulated proliferative responses. To further confirm the immunological potential of human NSCs, we studied xenogeneic recognition of NSCs by immunocompetent cells derived from C57BL/6 mice, showing that NSCs can elicit an allo(xeno) response ex vivo. Our data indicate that NSCs have low but not negligible immunogenic potential that is sufficient to activate peripheral lymphocytes. Secretion of TGF-beta1 might balance the immunogenicity of NSCs. Nevertheless, the possibility that allo-NSCs grafting might induce in the long term an immune activation, thus vanishing their therapeutical effect, should not be overlooked and deserves further investigation.
Collapse
Affiliation(s)
- Federica Ubiali
- Neurology IV, Neurological Institute Foundation Carlo Besta, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
195
|
Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, Dimeco F, Olivi A, Eberhart CG. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 2007; 25:2524-33. [PMID: 17628016 PMCID: PMC2610257 DOI: 10.1634/stemcells.2007-0166] [Citation(s) in RCA: 446] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brain tumors can arise following deregulation of signaling pathways normally activated during brain development and may derive from neural stem cells. Given the requirement for Hedgehog in non-neoplastic stem cells, we investigated whether Hedgehog blockade could target the stem-like population in glioblastoma multiforme (GBM). We found that Gli1, a key Hedgehog pathway target, was highly expressed in 5 of 19 primary GBM and in 4 of 7 GBM cell lines. Shh ligand was expressed in some primary tumors, and in GBM-derived neurospheres, suggesting a potential mechanism for pathway activation. Hedgehog pathway blockade by cyclopamine caused a 40%-60% reduction in growth of adherent glioma lines highly expressing Gli1 but not in those lacking evidence of pathway activity. When GBM-derived neurospheres were treated with cyclopamine and then dissociated and seeded in media lacking the inhibitor, no new neurospheres formed, suggesting that the clonogenic cancer stem cells had been depleted. Consistent with this hypothesis, the stem-like fraction in gliomas marked by both aldehyde dehydrogenase activity and Hoechst dye excretion (side population) was significantly reduced or eliminated by cyclopamine. In contrast, we found that radiation treatment of our GBM neurospheres increased the percentage of these stem-like cells, suggesting that this standard therapy preferentially targets better-differentiated neoplastic cells. Most importantly, viable GBM cells injected intracranially following Hedgehog blockade were no longer able to form tumors in athymic mice, indicating that a cancer stem cell population critical for ongoing growth had been removed. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Eli E. Bar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aneeka Chaudhry
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alex Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xing Fan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karisa Schreck
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sara Piccirillo
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | - Angelo L. Vescovi
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | | | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G. Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
196
|
Hovakimyan M, Haas SJP, Schmitt O, Gerber B, Wree A, Andressen C. Mesencephalic human neural progenitor cells transplanted into the neonatal hemiparkinsonian rat striatum differentiate into neurons and improve motor behaviour. J Anat 2007; 209:721-32. [PMID: 17118060 PMCID: PMC2049002 DOI: 10.1111/j.1469-7580.2006.00654.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Neural stem cell transplantation is a promising strategy for the treatment of neurodegenerative diseases. To evaluate the differentiation potential of human neural progenitor cells (hNPCs) as a prerequisite for clinical trials, we intracerebrally transplanted in vitro expanded fetal mesencephalic hNPCs into hemiparkinsonian rats. On postnatal day one (P1), 17 animals underwent a unilateral intraventricular 6-hydroxydopamine injection into the right lateral ventricle. At P3, animals (n = 10) received about 100,000 hNPCs (1 microL) in the right striatum. Five weeks after birth, animals underwent behaviour tests prior to fixation, followed by immunohistochemistry on brain slices for human nuclei, glial fibrillary acidic protein, S100beta, neuronal nuclei antigen, neuron-specific enolase and tyrosine hydroxylase. Compared with the apomorphine-induced rotations in the lesioned-only group (7.4 +/- 0.5 min(-1)), lesioned and successfully transplanted animals (0.3 +/- 0.1 min(-1)) showed a significant therapeutic improvement. Additionally, in the cylinder test, the lesioned-only animals preferred to use the ipsilateral forepaw. Conversely, the lesioned and transplanted animals showed no significant side bias similar to untreated control animals. Transplanted human nuclei-immunoreactive cells were found to survive and migrate up to 2000 microm into the host parenchyma, many containing the pan-neuronal markers neuronal nuclei antigen and neuron-specific enolase. In the striatum, tyrosine hydroxylase-immunoreactive somata were also found, indicating a dopaminergic differentiation capacity of transplanted hNPCs in vivo. However, the relative number of tyrosine hydroxylase-immunoreactive neurons in vivo seemed to be lower than in corresponding in vitro differentiation. To minimize donor tissue necessary for transplantation, further investigations will aim to enhance dopaminergic differentiation of transplanted cells in vivo.
Collapse
Affiliation(s)
- Marine Hovakimyan
- Institute of Anatomy, Medical Faculty, University of Rostock, Germany
| | | | | | | | | | | |
Collapse
|
197
|
McLeod M, Hong M, Sen A, Sadi D, Ulalia R, Behie LA, Mendez I. Transplantation of bioreactor-produced neural stem cells into the rodent brain. Cell Transplant 2007; 15:689-97. [PMID: 17269440 DOI: 10.3727/000000006783464426] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The development of new cell replacement strategies using neural stem cells (NSC) may provide an alternative and unlimited cell source for clinical neural transplantation in neurodegenerative diseases such as Parkinson's and Huntington's disease. The clinical application of neural transplantation using NSC will therefore depend upon the availability of clinical grade NSC that are generated in unlimited quantities in a standardized manner. In order to investigate the utility of NSC in clinical neural transplantation, undifferentiated murine NSC were first expanded for an extended period of time in suspension bioreactors containing a serum-free medium. Following expansion in suspension bioreactors, NSC were still able to differentiate in vitro into both astrocytes and neurons after exposure to brain-derived neurotrophic factor (BDNF), suggesting that bioreactor expansion does not alter cell lineage potentiality. Undifferentiated bioreactor-expanded NSC were then transplanted into the rodent striatum. Immunohistochemical examination revealed undifferentiated bioreactor-expanded NSC survived transplantation for up to 8 weeks and expressed the astrocytic immunohistochemical marker glial fibrillary acidic protein (GFAP), suggesting that the host striatal environment influences NSC cell fate upon transplantation. Moreover, no tumor formation was observed within the graft site, indicating that NSC expanded in suspension bioreactors for an extended period of time are a safe source of tissue for transplantation. Future studies should focus on predifferentiating NSC towards specific neuronal phenotypes prior to transplantation in order to restore behavioral function in rodent models of neurodegenerative disease.
Collapse
Affiliation(s)
- M McLeod
- Cell Restoration Laboratory, Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | | | | | | | |
Collapse
|
198
|
De Filippis L, Lamorte G, Snyder EY, Malgaroli A, Vescovi AL. A novel, immortal, and multipotent human neural stem cell line generating functional neurons and oligodendrocytes. Stem Cells 2007; 25:2312-21. [PMID: 17556596 DOI: 10.1634/stemcells.2007-0040] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The discovery and study of neural stem cells have revolutionized our understanding of the neurogenetic process, and their inherent ability to adopt expansive growth behavior in vitro is of paramount importance for the development of novel therapeutics based on neural cell replacement. Recent advances in high-throughput assays for drug development and gene discovery dictate the need for rapid, reproducible, long-term expansion of human neural stem cells (hNSCs). In this view, the complement of wild-type cell lines currently available is insufficient. Here we report the establishment of a stable human neural stem cell line (immortalized human NSCs [IhNSCs]) by v-myc-mediated immortalization of previously derived wild-type hNSCs. These cells demonstrate three- to fourfold faster proliferation than wild-type cells in response to growth factors but retain rather similar properties, including multipotentiality. By molecular biology, biochemistry, immunocytochemistry, fluorescence microscopy, and electrophysiology, we show that upon growth factor removal, IhNSCs completely downregulate v-myc expression, cease proliferation, and differentiate terminally into three major neural lineages: astrocytes, oligodendrocytes, and neurons. The latter are functional, mature cells displaying clear-cut morphological and physiological features of terminally differentiated neurons, encompassing mostly the GABAergic, glutamatergic, and cholinergic phenotypes. Finally, IhNSCs produce bona fide oligodendrocytes in fractions up to 20% of total cell number. This is in contrast to the negligible propensity of hNSCs to generate oligodendroglia reported so far. Thus, we describe an immortalized hNSC line endowed with the properties of normal hNSCs and suitable for developing the novel, reliable assays and reproducible high-throughput gene and drug screening that are essential in both diagnostics and cell therapy studies.
Collapse
Affiliation(s)
- Lidia De Filippis
- Department of Biotechnologies, Fondazione Centro San Raffaele del Monte Tabor, Milan, Italy
| | | | | | | | | |
Collapse
|
199
|
Pisati F, Bossolasco P, Meregalli M, Cova L, Belicchi M, Gavina M, Marchesi C, Calzarossa C, Soligo D, Lambertenghi-Deliliers G, Bresolin N, Silani V, Torrente Y, Polli E. Induction of neurotrophin expression via human adult mesenchymal stem cells: implication for cell therapy in neurodegenerative diseases. Cell Transplant 2007; 16:41-55. [PMID: 17436854 DOI: 10.3727/000000007783464443] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In animal models of neurological disorders for cerebral ischemia, Parkinson's disease, and spinal cord lesions, transplantation of mesenchymal stem cells (MSCs) has been reported to improve functional outcome. Three mechanisms have been suggested for the effects of the MSCs: transdifferentiation of the grafted cells with replacement of degenerating neural cells, cell fusion, and neuroprotection of the dying cells. Here we demonstrate that a restricted number of cells with differentiated astroglial features can be obtained from human adult MSCs (hMSCs) both in vitro using different induction protocols and in vivo after transplantation into the developing mouse brain. We then examined the in vitro differentiation capacity of the hMSCs in coculture with slices of neonatal brain cortex. In this condition the hMSCs did not show any neuronal transdifferentiation but expressed neurotrophin low-affinity (NGFR(p75)) and high-affinity (trkC) receptors and released nerve growth factor (NGF) and neurotrophin-3 (NT-3). The same neurotrophin's expression was demonstrated 45 days after the intracerebral transplantation of hMSCs into nude mice with surviving astroglial cells. These data further confirm the limited capability of adult hMSC to differentiate into neurons whereas they differentiated in astroglial cells. Moreover, the secretion of neurotrophic factors combined with activation of the specific receptors of transplanted hMSCs demonstrated an alternative mechanism for neuroprotection of degenerating neurons. hMSCs are further defined in their transplantation potential for treating neurological disorders.
Collapse
Affiliation(s)
- Federica Pisati
- Fondazione IRCCS Ospedale Maggiore, Department of Neurological Sciences, Stem Cell Laboratory, Dino Ferrari Center, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Invernici G, Emanueli C, Madeddu P, Cristini S, Gadau S, Benetti A, Ciusani E, Stassi G, Siragusa M, Nicosia R, Peschle C, Fascio U, Colombo A, Rizzuti T, Parati E, Alessandri G. Human fetal aorta contains vascular progenitor cells capable of inducing vasculogenesis, angiogenesis, and myogenesis in vitro and in a murine model of peripheral ischemia. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1879-92. [PMID: 17525256 PMCID: PMC1899439 DOI: 10.2353/ajpath.2007.060646] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2007] [Indexed: 12/19/2022]
Abstract
Vasculogenesis, the formation of blood vessels in embryonic or fetal tissue mediated by immature vascular cells (ie, angioblasts), is poorly understood. We report the identification of a population of vascular progenitor cells (hVPCs) in the human fetal aorta composed of undifferentiated mesenchymal cells that coexpress endothelial and myogenic markers. Under culture conditions that promoted cell differentiation, hVPCs gave rise to a mixed population of mature endothelial and mural cells when progenitor cells were stimulated with vascular endothelial growth factor-A or platelet-derived growth factor-betabeta. hVPCs grew as nonadherent cells and, when embedded in a three-dimensional collagen gel, reorganized into cohesive cellular cords that resembled mature vascular structures. hVPC-conditioned medium contained angiogenic substances (vascular endothelial growth factor-A and angiopoietin-2) and strongly stimulated the proliferation of endothelial cells. We also demonstrate the therapeutic efficacy of a small number of hVPCs transplanted into ischemic limb muscle of immunodeficient mice. hVPCs markedly improved neovascularization and inhibited the loss of endogenous endothelial cells and myocytes, thus ameliorating the clinical outcome from ischemia. We conclude that fetal aorta represents an important source for the investigation of the phenotypic and functional features of human vascular progenitor cells.
Collapse
Affiliation(s)
- Gloria Invernici
- Neurobiology and Neuroregenerative Therapies Unit, Carlo Besta Neurological Institute, Milan 20133, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|