901
|
Rohrschneider LR, Custodio JM, Anderson TA, Miller CP, Gu H. The intron 5/6 promoter region of the ship1 gene regulates expression in stem/progenitor cells of the mouse embryo. Dev Biol 2005; 283:503-21. [PMID: 15978570 DOI: 10.1016/j.ydbio.2005.04.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 04/07/2005] [Accepted: 04/26/2005] [Indexed: 10/25/2022]
Abstract
The s-SHIP protein is a shorter isoform of the longer SHIP1 protein and lacks the N-terminal SH2 domain region contained in SHIP1. s-SHIP is expressed in ES cells and in enriched bone marrow stem cells, and may be controlled by a promoter within intron 5 of the ship1 gene. We therefore examined the potential specificity of promoter activity in ES cells of an intron 5/intron 6 ship1 genomic segment and its tissue specificity within transgenic mice expressing GFP from this promoter region. The results indicate that s-SHIP promoter activity is specific for ES cells in vitro and for known and presumptive stem/progenitor cells throughout embryo development of the transgenic mice. Specific GFP expression was observed in the blastocyst, primordial germ cells, thymus, arterioles, osteoblasts, and skin epidermis. The epidermis/epithelium is the progenitor for hair follicles, mammary tissue, and prostate. Interestingly, each of these latter tissues acquired a few GFP-positive cells in the course of their development from the epithelial layers, and these cells express marker proteins for stem/progenitor cells. These results identify potential stem cell populations, mark these cells for analyses in normal and cancer development, and implicate s-SHIP as an important protein in stem/progenitor cell function.
Collapse
Affiliation(s)
- Larry R Rohrschneider
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | | | | | |
Collapse
|
902
|
Curtis MA, Waldvogel HJ, Synek B, Faull RLM. A histochemical and immunohistochemical analysis of the subependymal layer in the normal and Huntington's disease brain. J Chem Neuroanat 2005; 30:55-66. [PMID: 16108100 DOI: 10.1016/j.jchemneu.2005.05.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Previous studies in the rodent brain have characterised the cell types present in the subependymal layer, however the general organisation and cellular morphology of the adult human subependymal layer has not been demonstrated previously. In this study, we have demonstrated that the normal human brain subependymal layer contains three morphologically distinct types of cells, A, B and C type cells. The type A cells resembling migrating neuroblasts were located in the superficial part of the subependymal layer, type B cells resembling glial cells were evenly distributed throughout the subependymal layer and caudate nucleus, and type C cells that resembled progenitor cells were located in the deeper regions of the subependymal layer close to the caudate nucleus. We also examined the subependymal layer in the Huntington's disease brain to determine whether neurodegenerative pathology of the caudate nucleus (adjacent to the subependymal layer) altered the cellular composition of the subependymal layer. In the Huntington's disease subependymal layer there was a significant increase in the thickness of the subependymal layer compared with the normal subependymal layer (p < 0.01) and there was a 2.8-fold increase in the number of cells present in the Huntington's disease subependymal layer compared with the normal subependymal layer but the density of cells remained unchanged. As the grade of Huntington's disease increased, so did the overall number of cells in the subependymal layer. An increase in the number of type B cells was responsible for most of the increase demonstrated, however there was also an increase in the numbers of type A and C cells. To further characterise the human normal and Huntington's disease subependymal layer we used immunohistochemistry and antibodies against a range of projection neuron markers, interneuron markers, glial cell markers and GABAA receptor subunits. The results demonstrated the presence of increased numbers of neuropeptide Y positive cells in the Huntington's disease subependymal layer compared with the normal subependymal layer, suggesting that neuropeptide Y neurons may play a role in progenitor cell proliferation. Also there was an increased level of the developmentally active GABAA receptor subunit gamma 2 that indicates that the adult subependymal layer still retains the ability to proliferate. Taken together our results give a detailed description of the adult human subependymal layer and also demonstrate the plasticity of the human subependymal layer in response to Huntington's disease.
Collapse
Affiliation(s)
- Maurice A Curtis
- Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | |
Collapse
|
903
|
Domínguez-Pinos MD, Páez P, Jiménez AJ, Weil B, Arráez MA, Pérez-Fígares JM, Rodríguez EM. Ependymal Denudation and Alterations of the Subventricular Zone Occur in Human Fetuses With a Moderate Communicating Hydrocephalus. J Neuropathol Exp Neurol 2005; 64:595-604. [PMID: 16042311 DOI: 10.1097/01.jnen.0000171648.86718.bb] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In mutant rodents, ependymal denudation occurs early in fetal life, preceding the onset of a communicating hydrocephalus, and is a key event in the etiology of this disease. The present investigation was designed to obtain evidence whether or not ependymal denudation occurs in 16- to 40-week-old human fetuses developing a communicating hydrocephalus (n = 8) as compared to fetuses of similar ages with no neuropathologic alterations (n = 15). Sections through the walls of the cerebral aqueduct and lateral ventricles were processed for lectin binding and immunocytochemistry using antibodies against ependyma, astroglia, neuroblasts, and macrophages markers. Anticaveolin was used as a functional marker of the fetal ependyma. The structural and functional molecular markers are differentially expressed throughout the differentiation of the human fetal ependyma. Denudation of the ependyma of the aqueduct and lateral ventricles occurred in all fetuses developing a communicating hydrocephalus, including the youngest ones studied. The denuded surface area increased in parallel with the fetus age. The possibility is advanced that in many or most cases of human fetal hydrocephalus there is a common defect at the ependymal cell lineage leading to ependymal detachment. Evidence was obtained that in hydrocephalic human fetuses a process to repair the denuded areas takes place during the fetal life. In hydrocephalic fetuses, detachment of the ependyma of the lateral ventricles resulted in the (i) loss of the germinal ependymal zone, (ii) disorganization of the subventricular zone and, (iii) abnormal migration of neuroblasts into the ventricular cavity. Thus, detachment of the ependymal layer in hydrocephalic fetuses would not only be associated with the pathogenesis of hydrocephalus but also to abnormal neurogenesis.
Collapse
|
904
|
Yoshino J, Tochinai S. Successful reconstitution of the non-regenerating adult telencephalon by cell transplantation in Xenopus laevis. Dev Growth Differ 2005; 46:523-34. [PMID: 15610142 DOI: 10.1111/j.1440-169x.2004.00767.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The South African clawed frog (Xenopus laevis) can regenerate the anterior half of the telencephalon only during larval life, but such regeneration is no longer possible after metamorphosis. In order to gain a better understanding of differences between larvae and adults that are potentially related to regeneration, several experiments were conducted on larvae and froglets after the partial removal of the telencephalon. As a result, it was found that the cells in the brain proliferated actively, even in non-regenerating froglets, just as was observed in regenerating larvae after the partial removal of the telencephalon. Moreover, it was shown that although the structure was usually imperfect, even isolated single cells derived from the frog brain were able to reconstitute the lost portion when the cells were transplanted to the partially truncated telencephalon. It is therefore likely to be critical for massive organ regeneration that ependymal layer cells promptly cover the cerebral lateral ventricles at an initial stage of wound healing, as is the case observed in larvae. However, in froglets, these cells strongly adhere to one another, and they are therefore unable to move to seal off the exposed ventricle, which in turn is likely to render the froglet brain non-regenerative.
Collapse
Affiliation(s)
- Jun Yoshino
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, N10W8, Kita-ku, Sapporo 060-0810, Japan.
| | | |
Collapse
|
905
|
Timiras PS, Yaghmaie F, Saeed O, Thung E, Chinn G. The ageing phenome: caloric restriction and hormones promote neural cell survival, growth, and de-differentiation. Mech Ageing Dev 2005; 126:3-9. [PMID: 15610756 DOI: 10.1016/j.mad.2004.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The phenome represents the observable properties of an organism that have developed under the continued influences of both genome and environmental factors. Phenotypic properties are expressed through the functions of cells, organs and body systems that operate optimally, close to equilibrium. In complex organisms, maintenance of the equilibrium is achieved by the interplay of several regulatory mechanisms. In the elderly, dynamic instability may lead to progressive loss of normal function, failure of adaptation and increased pathology. Extensive research (reported elsewhere in this journal) has demonstrated that genetic manipulations of endocrine signaling in flies, worms and mice increase longevity. Another effective strategy for prolonging the lifespan is caloric restriction: in data presented here, the persistence of estrogen-sensitive cells in the hypothalamus of caloric restricted 22-month-old female mice, may explain the persistence of reproductive function at an age, when reproductive function has long ceased in ad libitum fed controls. Still another strategy utilizes the effects of epidermal growth factor (EGF) to promote in vitro proliferation of neuroglia, astrocytes and oligodendrocytes. Their subsequent de-differentiation generates immature precursor cells potentially capable of differentiating into neuroblasts and neurons. These and other examples suggest that, in terms of functional outcomes, "the genome proposes but the phenome disposes".
Collapse
Affiliation(s)
- Paola S Timiras
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3202, USA.
| | | | | | | | | |
Collapse
|
906
|
Kubis N, Catala M. [Where have the neuronal stem cells of the subependymal zone gone in human beings?]. Morphologie 2005; 89:49-54. [PMID: 15943081 DOI: 10.1016/s1286-0115(05)83238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Stem cells are characterized by their ability for self-renewal (allowing them to be present throughout the entire life of the organism) and their ability to give rise to differentiated cells belong to one or more lineages. The strict definition of these cells is however still a matter of debate. There is new experimental evidence (including in human beings) that stem cells are present within the brain and may give rise to neurons. Ependymal cells have been proposed to play such a role. In fact, subependymal cells expressing GFAP would be more likely candidates. Such cells are observed in the brain of human beings. They are able to differentiate into neurons in vitro but such potential appears to be repressed in vivo.
Collapse
Affiliation(s)
- N Kubis
- Laboratoire d'Explorations Fonctionnelles Multidisciplinaires, Hôpital Lariboisière, Paris
| | | |
Collapse
|
907
|
Yamashita YM, Fuller MT, Jones DL. Signaling in stem cell niches: lessons from the Drosophila germline. J Cell Sci 2005; 118:665-72. [PMID: 15701923 DOI: 10.1242/jcs.01680] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stem cells are cells that, upon division, can produce new stem cells as well as daughter cells that initiate differentiation along a specific lineage. Studies using the Drosophila germline as a model system have demonstrated that signaling from the stem cell niche plays a crucial role in controlling stem cell behavior. Surrounding support cells secrete growth factors that activate signaling within adjacent stem cells to specify stem cell self-renewal and block differentiation. In addition, cell-cell adhesion between stem cells and surrounding support cells is important for holding stem cells close to self-renewal signals. Furthermore, a combination of localized signaling and autonomously acting proteins might polarize stem cells in such a way as to ensure asymmetric stem cell divisions. Recent results describing stem cell niches in other adult stem cells, including hematopoietic and neural stem cells, have demonstrated that the features characteristic of stem cell niches in Drosophila gonads might be conserved.
Collapse
Affiliation(s)
- Yukiko M Yamashita
- Department of Development Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
908
|
Karl C, Couillard-Despres S, Prang P, Munding M, Kilb W, Brigadski T, Plötz S, Mages W, Luhmann H, Winkler J, Bogdahn U, Aigner L. Neuronal precursor-specific activity of a human doublecortin regulatory sequence. J Neurochem 2005; 92:264-82. [PMID: 15663475 DOI: 10.1111/j.1471-4159.2004.02879.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The doublecortin (DCX) gene encodes a 40-kDa microtubule-associated protein specifically expressed in neuronal precursors of the developing and adult CNS. Due to its specific expression pattern, attention was drawn to DCX as a marker for neuronal precursors and neurogenesis, thereby underscoring the importance of its promoter identification and promoter analysis. Here, we analysed the human DCX regulatory sequence and confined it to a 3.5-kb fragment upstream of the ATG start codon. We demonstrate by transient transfection experiments that this fragment is sufficient and specific to drive expression of reporter genes in embryonic and adult neuronal precursors. The activity of this regulatory fragment overlapped with the expression of endogenous DCX and with the young neuronal markers class III beta-tubulin isotype and microtubule-associated protein Map2ab but not with glial or oligodendroglial markers. Electrophysiological data further confirmed the immature neuronal nature of these cells. Deletions within the 3.5-kb region demonstrated the relevance of specific regions containing transcription factor-binding sites. Moreover, application of neurogenesis-related growth factors in the neuronal precursor cultures suggested the lack of direct signalling of these factors on the DCX promoter construct.
Collapse
Affiliation(s)
- Claudia Karl
- Volkswagen-Foundation-Research Group, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
909
|
Abstract
Transplantation of functional islets of Langerhans may emerge as a useful therapy for some patients with type 1 diabetes mellitus (DM), but donor islet shortages motivate the search for new sources of transplantable islets. Pluripotent embryonic stem (ES) cells are expandable in culture and have the potential to give rise to all cell types in the body. The recent isolation of pluripotent ES cells from humans has generated excitement over the possibility of engineering glucose-responsive islet replacement tissue from these cells in large quantities. In this study, we review the recent advances in generating insulin-producing cells (IPC) from mouse and human ES (hES) cells.
Collapse
Affiliation(s)
- Jeremy J Heit
- Department of Developmental Biology, Stanford University, 279 Campus Drive, Stanford, CA 94305, USA
| | | |
Collapse
|
910
|
Abstract
The genetic analysis of common neurological disorders will be a difficult and protracted endeavour. Genetics is only one of many disciplines that will be required but it has already thrown considerable light on the aetiology of several major neurological disorders through the analysis of rare inherited subgroups. The identification of individual susceptibility genes with variants of smaller effect will be more difficult but there is no sharp demarcation between large and small genetic effects, so that many new and important insights will emerge using existing and new technologies. The availability of improved neuroimaging, better animal models of disease and new genetic tools, such as high-throughput gene chips, expression microarrays and proteomics, are extending the range of traditional genetic mapping tools. Finally, an understanding of the genetic and epigenetic mechanisms that restrain the differentiation and integration of human neural stem cells into mature neuronal networks could have a major impact on clinical practice. These approaches will be illustrated in the context of Alzheimer disease, Parkinson disease and synucleinopathies, tauopathies, amyotrophic lateral sclerosis and stroke.
Collapse
Affiliation(s)
- A F Wright
- MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
911
|
Lledo PM, Saghatelyan A. Integrating new neurons into the adult olfactory bulb: joining the network, life–death decisions, and the effects of sensory experience. Trends Neurosci 2005; 28:248-54. [PMID: 15866199 DOI: 10.1016/j.tins.2005.03.005] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In contrast to the situation in the developing brain, neurons born during adulthood must integrate into established neuronal networks characterized by ongoing activity. For sensory systems, this neuronal activity is driven mainly by external stimuli that can lead to experience-dependent morpho-functional changes in adult circuits. Here, we describe new insights into the mechanisms by which sensory experience might govern the targeting of adult-generated neurons to appropriate regions, their differentiation into distinct neuronal subtypes, and finally their survival in the adult olfactory bulb. We propose not only that neurogenesis depends on the degree of sensory experience, but also that new neurons bring unique features to the operational network, allowing a continuous adjustment of information processing in response to an ever-changing external word.
Collapse
Affiliation(s)
- Pierre-Marie Lledo
- Laboratory of Perception and Memory, Centre National de la Recherche Scientifique, Unité de Recherche Associée 2182, Pasteur Institute, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France.
| | | |
Collapse
|
912
|
Mitchell BD, Emsley JG, Magavi SSP, Arlotta P, Macklis JD. Constitutive and induced neurogenesis in the adult mammalian brain: manipulation of endogenous precursors toward CNS repair. Dev Neurosci 2005; 26:101-17. [PMID: 15711054 DOI: 10.1159/000082131] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2004] [Accepted: 03/07/2004] [Indexed: 12/31/2022] Open
Abstract
Over most of the past century of modern neuroscience, it was thought that the adult brain was completely incapable of generating new neurons. During the past 3 decades, research exploring potential neuronal replacement therapies has focused on replacing lost neurons by transplanting cells or grafting tissue into diseased regions of the brain. However, in the last decade, the development of new techniques has resulted in an explosion of new research showing that neurogenesis, the birth of new neurons, normally occurs in two limited and specific regions of the adult mammalian brain and that there are significant numbers of multipotent neural precursors in many parts of the adult mammalian brain. Recent advances in our understanding of related events of neural development and plasticity, including the role of radial glia in developmental neurogenesis and the ability of endogenous precursors present in the adult brain to be induced to produce neurons and partially repopulate brain regions affected by neurodegenerative processes, have led to fundamental changes in the views about how the brain develops as well as to approaches by which endogenous precursors might be recruited to repair the adult brain. Recruitment of new neurons can be induced in a region-specific, layer-specific and neuronal-type-specific manner, and, in some cases, newly recruited neurons can form long-distance connections to appropriate targets. Elucidation of the relevant molecular controls may both allow control over transplanted precursor cells and potentially allow the development of neuronal replacement therapies for neurodegenerative disease and other CNS injuries that do not require transplantation of exogenous cells.
Collapse
Affiliation(s)
- Bartley D Mitchell
- MGH-HMS Center for Nervous System Repair, Department of Neurosurgery, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
913
|
Williams R, Ryves WJ, Dalton EC, Eickholt B, Shaltiel G, Agam G, Harwood AJ. A molecular cell biology of lithium. Biochem Soc Trans 2005; 32:799-802. [PMID: 15494019 DOI: 10.1042/bst0320799] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lithium (Li(+)), a mood stabilizer, has profound effects on cultured neurons, offering an opportunity to investigate its cellular biological effects. Here we consider the effect of Li(+) and other psychotropic drugs on growth cone morphology and chemotaxis. Li(+) inhibits GSK-3 (glycogen synthase kinase-3) at a therapeutically relevant concentration. Treated cells show a number of features that arise due to GSK-3 inhibition, such as altered microtubule dynamics, axonal branching and loss of semaphorin 3A-mediated growth cone collapse. Li(+) also causes growth cones to spread; however, a similar effect is seen with two other mood stabilizers, valproic acid and carbamazepine, but without changes in microtubules or axon branching. This common effect of mood stabilizers is mediated by changes in inositol phosphate signalling, not GSK-3 activity. Given the presence of neurogenesis in the adult brain, we speculate that changes in growth cone behaviour could also occur during treatment of mental disorders.
Collapse
Affiliation(s)
- R Williams
- MRC Laboratory for Molecular Cell Biology and Department of Biology, University College London, Gower St, London WC1E 6BT, UK
| | | | | | | | | | | | | |
Collapse
|
914
|
Santner-Nanan B, Peek MJ, McCullagh P, Nanan R. Therapeutic potential of stem cells in perinatal medicine. Aust N Z J Obstet Gynaecol 2005; 45:102-7. [PMID: 15760308 DOI: 10.1111/j.1479-828x.2005.00362.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Increasing evidence suggests that stem cells have tremendous potential to facilitate repair of damaged tissue and to exert protective influences that limit the extent of damage. Their inherent capacity to respond to signals generated by damaged tissue, migrate to these regions and either replace dead tissue or deliver protection by secretion of specific growth hormones and protective factors, suggests that they might have unrivalled therapeutic potential in perinatal medicine. A further potential of stem cells is their use in gene repair strategies for genetic disorders; an application which is exceedingly interesting from a perinatal perspective. Because of the relatively small size of infants and their capacity for future growth, stem cell therapy could be more successful in newborns than in older children or adults. In practical terms, the placenta, with its large reservoir of fetal blood, offers the ideal source of autologous stem cells. This affords the opportunity for stem cells to be collected and used, either directly ex vivo or after in vitro modulation, both for disorders in the neonatal period and for those arising later in life. The organs most affected from tissue damage in the neonatal period are the brain and the lung. So far, the most promising application of stem cells might be in the treatment of neurological injury. In this review we discuss recent research findings with adult stem cell therapy and their potential use in perinatal medicine. Furthermore, specific animal models suitable to explore the patho-physiological mechanisms of stem cell transplantation after neurological injury will be discussed. This review gives an overview of basic science findings and their possible role for clinical application with regards to the therapeutic potential of stem cells in perinatal medicine. Medline was searched for journal selection in peer-reviewed journals with high impact scores, which were relevant to this topic. All articles were in English and the search was not limited by publication year. However, the oldest publication was dated 1988 (reference 1).
Collapse
Affiliation(s)
- Brigitte Santner-Nanan
- Department of Women's and Children's Health, Western Clinical School, The University of Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
915
|
Pluchino S, Zanotti L, Deleidi M, Martino G. Neural stem cells and their use as therapeutic tool in neurological disorders. ACTA ACUST UNITED AC 2005; 48:211-9. [PMID: 15850660 DOI: 10.1016/j.brainresrev.2004.12.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 10/25/2022]
Abstract
Spontaneous neural tissue repair occurs in patients affected by inflammatory and degenerative disorders of the central nervous system (CNS). However, this process is not robust enough to promote a functional and stable recovery of the CNS architecture. The development of cell-based therapies aimed at promoting brain repair, through damaged cell-replacement, is therefore foreseen. Several experimental cell-based strategies aimed at replacing damaged neural cells have been developed in the last 30 years. Although successful in promoting site-specific repair in focal CNS disorders, most of these therapeutic approaches have failed to foster repair in multifocal CNS diseases where the anatomical and functional damage is widespread. Stem cell-based therapies have been recently proposed and might represent in the near future a plausible alternative strategy in these disorders. However, before envisaging any human applications of stem cell-based therapies in neurological diseases, we need to consider some preliminary and still unsolved issues: (i) the ideal stem cell source for transplantation, (ii) the most appropriate route of stem cell administration, and, last but not least, (iii) the best approach to achieve an appropriate, functional, and long-lasting integration of transplanted stem cells into the host tissue.
Collapse
Affiliation(s)
- Stefano Pluchino
- Department of Neuroscience, San Raffaele Scientific Institute, Neuroimmunology Unit-DIBIT, Milano, Italy
| | | | | | | |
Collapse
|
916
|
Couillard-Despres S, Winner B, Schaubeck S, Aigner R, Vroemen M, Weidner N, Bogdahn U, Winkler J, Kuhn HG, Aigner L. Doublecortin expression levels in adult brain reflect neurogenesis. Eur J Neurosci 2005; 21:1-14. [PMID: 15654838 DOI: 10.1111/j.1460-9568.2004.03813.x] [Citation(s) in RCA: 799] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Progress in the field of neurogenesis is currently limited by the lack of tools enabling fast and quantitative analysis of neurogenesis in the adult brain. Doublecortin (DCX) has recently been used as a marker for neurogenesis. However, it was not clear whether DCX could be used to assess modulations occurring in the rate of neurogenesis in the adult mammalian central nervous system following lesioning or stimulatory factors. Using two paradigms increasing neurogenesis levels (physical activity and epileptic seizures), we demonstrate that quantification of DCX-expressing cells allows for an accurate measurement of modulations in the rate of adult neurogenesis. Importantly, we excluded induction of DCX expression during physiological or reactive gliogenesis and excluded also DCX re-expression during regenerative axonal growth. Our data validate DCX as a reliable and specific marker that reflects levels of adult neurogenesis and its modulation. We demonstrate that DCX is a valuable alternative to techniques currently used to measure the levels of neurogenesis. Importantly, in contrast to conventional techniques, analysis of neurogenesis through the detection of DCX does not require in vivo labelling of proliferating cells, thereby opening new avenues for the study of human neurogenesis under normal and pathological conditions.
Collapse
Affiliation(s)
- Sebastien Couillard-Despres
- Volkswagen-Foundation Junior Group, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
917
|
Abstract
PURPOSE OF REVIEW Conventional surgery, radio- and chemotherapy have failed to significantly improve the prognosis of patients with malignant astrocytomas--hence the need for understanding their molecular biology. Harvesting this understanding to yield novel biological targeted therapies has approached the clinical doorstep. Therapeutic efficacy will likely require combinatorial therapy involving biologicals and conventional therapies, with small incremental efficacy in selected sub-groups. This review highlights some of the findings over the past year (June 2003-2004) that have contributed to this slow but essential journey towards our understanding of the biology of astrocytomas. RECENT FINDINGS The accumulation of loss and/or gain of function molecular alterations underlying astrocytoma formation, progression and key growth parameters including proliferation, angiogenesis, apoptosis, invasion and resistance are emerging. These alterations involve those regulating the growth factor/receptor and downstream signaling networks, cell cycle, immune modulators and other key biological processes. The advances are facilitated by interactions amongst clinician and basic scientists, in both academia and industry. They have incorporated high-throughput bioinformatics analysis of genomic and expression array data, the emerging field of proteomics and development of various genetically engineered models of astrocytomas. SUMMARY Astrocytomas, like other cancers, are a result of several molecular alterations, some of which strongly correlate to their pathological grade. However, molecular heterogeneity exists between astrocytomas of similar grades and likely between varying micro-environmental regions of a single tumor. Characterization of the molecular signature of an astrocytoma and linking with the appropriate 'tailored' therapie(s) is the hope of the future.
Collapse
Affiliation(s)
- Abhijit Guha
- Western Hospital, University of Toronto, Toronto, Ontario M5T 2S8, Canada.
| | | |
Collapse
|
918
|
Stein CS, Martins I, Davidson BL. The Lymphocytic Choriomeningitis Virus Envelope Glycoprotein Targets Lentiviral Gene Transfer Vector to Neural Progenitors in the Murine Brain. Mol Ther 2005; 11:382-9. [PMID: 15727934 DOI: 10.1016/j.ymthe.2004.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Revised: 10/29/2004] [Accepted: 11/08/2004] [Indexed: 11/29/2022] Open
Abstract
Feline immunodeficiency virus (FIV)-based lentiviral vectors can be targeted to restricted cell types by pseudotyping with envelopes from other viruses. An FIV vector expressing bacterial beta-galactosidase (beta-gal) and pseudotyped with lymphocytic choriomeningitis virus (LCMV) envelope glycoprotein was injected into postnatal mouse brain striatum to determine neural cell-type transduction. After 3 or 7.5 weeks, the beta-gal-expressing cells included astrocytes in the striatum and in the subventricular zone (SVZ), neuroblasts along the rostral migratory stream, and neurons in the olfactory bulb. This pattern was suggestive of transduction of neural stem cells/progenitors that reside in the SVZ and continually generate olfactory bulb neurons. To test for transduction of SVZ type B astrocyte/stem cells, LCMV-pseudotyped FIV encoding Cre recombinase driven by an astrocyte-specific promoter was injected into the striatum of ROSA26 Cre reporter mice. beta-Gal expression in these mice depends on Cre recombinase-mediated DNA recombination. beta-Gal-expressing neuroblasts and neurons were detected in the rostral migratory stream and olfactory bulb, respectively, indicating that these cells derived from an astrocytic-type stem cell. Thus, LCMV (WE54)-pseudotyped FIV provides a novel vector for transducing neural stem cells/progenitors in vivo and may prove valuable as a gene transfer vector for therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Colleen S Stein
- Program in Gene Therapy, Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
919
|
Lemkine GF, Raj A, Alfama G, Turque N, Hassani Z, Alegria-Prévot O, Samarut J, Levi G, Demeneix BA. Adult neural stem cell cycling in vivo requires thyroid hormone and its alpha receptor. FASEB J 2005; 19:863-5. [PMID: 15728663 DOI: 10.1096/fj.04-2916fje] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Thyroid hormones (TH) are essential for brain development. However, information on if and how this key endocrine factor affects adult neurogenesis is fragmentary. We thus investigated the effects of TH on proliferation and apoptosis of stem cells in the subventricular zone (SVZ), as well as on migration of transgene-tagged neuroblasts out of the stem cell niche. Hypothyroidism significantly reduced all three of these processes, inhibiting generation of new cells. To determine the mechanisms relaying TH action in the SVZ, we analyzed which receptor was implicated and whether the effects were played out directly at the level of the stem cell population. The alpha TH receptor (TRalpha), but not TRbeta, was found to be expressed in nestin positive progenitor cells of the SVZ. Further, use of TRalpha mutant mice showed TRalpha to be required to maintain full proliferative activity. Finally, a direct TH transcriptional effect, not mediated through other cell populations, was revealed by targeted gene transfer to stem cells in vivo. Indeed, TH directly modulated transcription from the c-myc promoter reporter construct containing a functional TH response element containing TRE but not from a mutated TRE sequence. We conclude that liganded-TRalpha is critical for neurogenesis in the adult mammalian brain.
Collapse
Affiliation(s)
- G F Lemkine
- UMR CNRS 5166, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
920
|
Környei Z, Szlávik V, Szabó B, Gócza E, Czirók A, Madarász E. Humoral and contact interactions in astroglia/stem cell co-cultures in the course of glia-induced neurogenesis. Glia 2005; 49:430-44. [PMID: 15546152 DOI: 10.1002/glia.20123] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Astroglial cells support or restrict the migration and differentiation of neural stem cells depending on the developmental stage of the progenitors and the physiological state of the astrocytes. In the present study, we show that astroglial cells instruct noncommitted, immortalized neuroectodermal stem cells to adopt a neuronal fate, while they fail to induce neuronal differentiation of embryonic stem cells under similar culture conditions. Astrocytes induce neuron formation by neuroectodermal progenitors both through direct cell-to-cell contacts and via short-range acting humoral factors. Neuron formation takes place inside compact stem cell assemblies formed 30- 60 h after the onset of glial induction. Statistical analyses of time-lapse microscopic recordings show that direct contacts with astrocytes hinder the migration of neuroectodermal progenitors, while astroglia-derived humoral factors increase their motility. In non-contact co-cultures with astrocytes, altered adhesiveness prevents the separation of frequently colliding neural stem cells. By contrast, in contact co-cultures with astrocytes, the restricted migration on glial surfaces keeps the cell progenies together, resulting in the formation of clonally proliferating stem cell aggregates. The data indicate that in vitro maintained parenchymal astrocytes (1) secrete factors, which initiate neuronal differentiation of neuroectodermal stem cells; and (2) provide a cellular microenvironment where stem cell/stem cell interactions can develop and the sorting out of the future neurons can proceed. In contrast to noncommitted progenitors, postmitotic neuronal precursors leave the stem cell clusters, indicating that astroglial cells selectively support the migration of maturing neurons as well as the elongation of neurites.
Collapse
Affiliation(s)
- Zsuzsanna Környei
- Institute of Experimental Medicine, Hungarian Academy of Sciences, P.O. Box 67, H-1450 Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|
921
|
Colodner KJ, Montana RA, Anthony DC, Folkerth RD, De Girolami U, Feany MB. Proliferative Potential of Human Astrocytes. J Neuropathol Exp Neurol 2005; 64:163-9. [PMID: 15751231 DOI: 10.1093/jnen/64.2.163] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although a number of studies have demonstrated proliferation of nonneoplastic astrocytes in experimental animal models, the proliferative potential of human astrocytes has not been well defined. Using double-label immunohistochemistry, we identified proliferating cells with the proliferation marker MIB-1 and astrocytes with glial fibrillary acidic protein staining in human biopsy and autopsy tissue. MIB-1 labeling of astrocytes was monitored in a variety of conditions containing significant numbers of reactive astrocytes, including infections, arteriovenous malformations, demyelinating lesions, metastatic tumors, and long-standing gliosis. Twenty-nine of a total of 54 cases showed no evidence of astrocyte-specific MIB-1 labeling despite prominent reactive changes. An average proliferation rate of 0.9% was present in the remaining 25 cases. Labeling indices were highest in infectious conditions and acute demyelinating lesions. We also examined astrocyte proliferation in 5 cases of progressive multifocal leukoencephalopathy. Astrocytic labeling indices were notably elevated in these cases, with an average labeling index of 5.8%. We conclude that low, but appreciable, astrocytic proliferation may occur in nonneoplastic human astrocytes. These findings have implications for astrocyte function in the normal and disease states and for the diagnostic distinction between reactive lesions and low-grade astrocytic neoplasms.
Collapse
Affiliation(s)
- Kenneth J Colodner
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
922
|
Abstract
Recently, modern neuroscience has made considerable progress in understanding how the brain perceives, discriminates, and recognizes odorant molecules. This growing knowledge took over when the sense of smell was no longer considered only as a matter for poetry or the perfume industry. Over the last decades, chemical senses captured the attention of scientists who started to investigate the different stages of olfactory pathways. Distinct fields such as genetic, biochemistry, cellular biology, neurophysiology, and behavior have contributed to provide a picture of how odor information is processed in the olfactory system as it moves from the periphery to higher areas of the brain. So far, the combination of these approaches has been most effective at the cellular level, but there are already signs, and even greater hope, that the same is gradually happening at the systems level. This review summarizes the current ideas concerning the cellular mechanisms and organizational strategies used by the olfactory system to process olfactory information. We present findings that exemplified the high degree of olfactory plasticity, with special emphasis on the first central relay of the olfactory system. Recent observations supporting the necessity of such plasticity for adult brain functions are also discussed. Due to space constraints, this review focuses mainly on the olfactory systems of vertebrates, and primarily those of mammals.
Collapse
|
923
|
Cooper O, Isacson O. Intrastriatal transforming growth factor alpha delivery to a model of Parkinson's disease induces proliferation and migration of endogenous adult neural progenitor cells without differentiation into dopaminergic neurons. J Neurosci 2005; 24:8924-31. [PMID: 15483111 PMCID: PMC2613225 DOI: 10.1523/jneurosci.2344-04.2004] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We examined the cell proliferative, neurogenic, and behavioral effects of transforming growth factor alpha (TGFalpha) in a 6-OHDA Parkinson's disease model when compared with naive rats. Intrastriatal TGFalpha infusion induced significant proliferation, hyperplastic nodules, and substantial migratory waves of nestin-positive progenitor cells from the adult subventricular zone (SVZ) of dopamine-denervated rats. Interestingly, SVZ cells in naive rats displayed proliferation but minimal migration in response to the TGFalpha infusion. The cells in the expanded SVZ accumulated cytoplasmic beta-catenin, indicating activation of classical Wnt signaling. However, no evidence of any neuronal differentiation was found of these recruited progenitor cells anywhere examined in the brain. Consequently, no evidence of dopaminergic (DA) neurogenesis was found in the striatum or substantia nigra in any experimental group, and amphetamine-induced behavioral rotations did not improve. In summary, the cells in the TGFalpha-induced migratory cellular wave remain undifferentiated and do not differentiate into midbrain-like DA neurons.
Collapse
Affiliation(s)
- Oliver Cooper
- Harvard University and McLean Hospital, National Institute of Neurological Disorders and Stroke Udall Parkinson's Disease Research Center of Excellence, Belmont, Massachusetts 02478, USA
| | | |
Collapse
|
924
|
Wada A, Yokoo H, Yanagita T, Kobayashi H. Lithium: Potential Therapeutics Against Acute Brain Injuries and Chronic Neurodegenerative Diseases. J Pharmacol Sci 2005; 99:307-21. [PMID: 16340157 DOI: 10.1254/jphs.crj05009x] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
In addition to the well-documented mood-stabilizing effects of lithium in manic-depressive illness patients, recent in vitro and in vivo studies in rodents and humans have increasingly implicated that lithium can be used in the treatment of acute brain injuries (e.g., ischemia) and chronic neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, tauopathies, and Huntington's disease). Consistent with this novel view, substantial evidences suggest that depressive illness is not a mere neurochemical disease, but is linked to gray matter atrophy due to the reduced number/size of neurons and glia in brain. Importantly, neurogenesis, that is, birth/maturation of functional new neurons, continues to occur throughout the lifetime in human adult brains (e.g., hippocampus); the neurogenesis is impaired by multiple not-fully defined factors (e.g., aging, chronic stress-induced increase of glucocorticoids, and excitotoxicity), accounting for brain atrophy in patients with depressive illness and neurodegenerative diseases. Chronic treatment of lithium, in agreement with the delayed-onset of mood-stabilizing effects of lithium, up-regulates cell survival molecules (e.g., Bcl-2, cyclic AMP-responsive element binding protein, brain-derived neurotrophic factor, Grp78, Hsp70, and beta-catenin), while down-regulating pro-apoptotic activities (e.g., excitotoxicity, p53, Bax, caspase, cytochrome c release, beta-amyloid peptide production, and tau hyperphosphorylation), thus preventing or even reversing neuronal cell death and neurogenesis retardation.
Collapse
Affiliation(s)
- Akihiko Wada
- Department of Pharmacology, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
| | | | | | | |
Collapse
|
925
|
Alexanian AR, Kurpad SN. Quiescent neural cells regain multipotent stem cell characteristics influenced by adult neural stem cells in co-culture. Exp Neurol 2005; 191:193-7. [PMID: 15589526 DOI: 10.1016/j.expneurol.2004.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Revised: 10/01/2004] [Accepted: 10/13/2004] [Indexed: 12/23/2022]
Abstract
The source of cells participating in central nervous system (CNS) tissue repair and regeneration is poorly defined. One possible source is quiescent neural cells that can persist in CNS in the form of dormant progenitors or highly specialized cell types. Under appropriate conditions, these quiescent cells may be capable of re-entering the mitotic cell cycle and contributing to the stem cell pool. The aim of this study was to determine whether in vitro differentiated neural stem cells (NSC) can regain their multipotent-like stem cell characteristics in co-culture with NSC. To this end, we induced neural differentiation by plating NSC, derived from the periventricular subependymal zone (SEZ) of ROSA26 transgenic mice in Neurobasal A/B27 medium in the absence of bFGF. Under these conditions, NSC differentiated into neurons, glia, and oligodendrocytes. While the level of Nestin expression was downregulated, persistence of dormant progenitors could not be ruled out. However, further addition of bFGF or bFGF/EGF with conditioned medium derived from adult NSC did not induce any noticeable cell proliferation. In another experiment, differentiated neural cells were cultured with adult NSC, isolated from the hippocampus of Balb/c mice, in the presence bFGF. This resulted in proliferating colonies of ROSA26 derived cells that mimicked NSC in their morphology, growth kinetics, and expressed NSC marker proteins. The average nuclear area and DAPI fluorescence intensity of these cells were similar to that of NSC grown alone. We conclude that reactivation of quiescent neural cells can be initiated by NSC-associated short-range cues but not by cell fusion.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Neuroscience Research Laboratory, Deparment of Neurosurgery, Medical College of Wisconsin, VAMC, 5000 W. National Avenue 151, Building 70-D, Milwaukee, WI 53295, USA.
| | | |
Collapse
|
926
|
Mori T, Buffo A, Götz M. The novel roles of glial cells revisited: the contribution of radial glia and astrocytes to neurogenesis. Curr Top Dev Biol 2005; 69:67-99. [PMID: 16243597 DOI: 10.1016/s0070-2153(05)69004-7] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most frequent cell type in the adult mammalian brain, and the number and range of their diverse functions are still increasing. One of their most striking roles is their function as adult neural stem cells and contribution to neurogenesis. This chapter discusses first the role of the ubiquitous glial cell type in the developing nervous system, the radial glial cells. Radial glial cells share several features with neuroepithelial cells, but also with astrocytes in the mature brain, which led to the name "radial glia." At the end of neurogenesis in the mammalian brain, radial glial cells disappear, and a subset of them transforms into astroglial cells. Interestingly, only some astrocytes maintain their neurogenic potential and continue to generate neurons throughout life. We discuss the current knowledge about the differences between the adult astroglial cells that remain neurogenic and act as neural stem cells and the majority of other astroglial cells that have apparently lost the capacity to generate neurons. Additionally, we review the changes in glial cells upon brain lesion, their dedifferentiation and recapitulation of radial glial properties, and the conditions under which reactive glia may reinitiate some neurogenic potential. Given that the astroglial cells are not only the most frequent cell type in an adult mammalian brain, but also the key cell type in the wound reaction of the brain to injury, it is essential to further understand their heterogeneity and molecular specification, with the final aim of using this unique source for neuronal replacement. Therefore, one of the key advances in the field of neurobiology is the discovery that astroglial cells can generate neurons not only during development, but also throughout adult life and potentially even after brain lesion.
Collapse
Affiliation(s)
- Tetsuji Mori
- Institute for Stem Cell Research, GSF-National Research Center for Environment and Health, D-85764 Neuherberg/Munich, Germany
| | | | | |
Collapse
|
927
|
Cacci E, Claasen JH, Kokaia Z. Microglia-derived tumor necrosis factor-α exaggerates death of newborn hippocampal progenitor cells in vitro. J Neurosci Res 2005; 80:789-97. [PMID: 15884015 DOI: 10.1002/jnr.20531] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Production of new hippocampal neurons continues in adult mammals and different brain insults can significantly increase this process. However, many hippocampal progenitor cells (HPC) die shortly after birth. Here we investigated the possibility that increased release of cytokines by activated microglia contributes to the death of HPC. We showed that addition of tumor necrosis factor-alpha (TNFalpha) to the medium of a cultured HPC line (HiB5) shortly after the cells stopped division causes significant apoptotic cell death. Conditioned medium from an activated microglial cell line (BV-2) had a similar effect, though conditioned medium from nonactivated microglia increased the survival of HPC. Reverse transcription-PCR indicated that HPC and microglial cells express both TNF receptors, TNF-R1 and TNF-R2. Coculturing of HPC with activated microglial cells aggravated death of hippocampal progenitors and also caused death of microglial cells themselves. Our data indicate that activated microglia-released TNFalpha might be an important contributor in inflammation-induced exaggeration of death of newly formed HPC in the adult brain after an insult.
Collapse
Affiliation(s)
- Emanuele Cacci
- Laboratory of Neural Stem Cell Biology, Section of Restorative Neurology, University Hospital, and Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Sweden
| | | | | |
Collapse
|
928
|
Abstract
The brain was traditionally viewed as a static organ, without turnover of neurons or significant capacity for self-repair after insults. Research in the last years has established that neurons are renewed in certain areas throughout life. The prospect of stimulation of endogenous neurogenesis as well as cell transplantation has raised hope for new therapies for neurological diseases.
Collapse
Affiliation(s)
- Anna Falk
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
929
|
Mueller FJ, McKercher SR, Imitola J, Loring JF, Yip S, Khoury SJ, Snyder EY. At the interface of the immune system and the nervous system: how neuroinflammation modulates the fate of neural progenitors in vivo. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2005:83-114. [PMID: 16315610 DOI: 10.1007/3-540-27626-2_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Neural stem and progenitor cells express a variety of receptors that enable them to sense and react to signals emanating from physiological and pathophysiological conditions in the brain as well as elsewhere in the body. Many of these receptors and were first described in investigations of the immune system, particularly with respect to hematopoietic stem cells. This emerging view of neurobiology has two major implications. First, many phenomena known from the hematopoietic system may actually be generalizable to stem cells from many organ systems, reflecting the cells' progenitor-mediated regenerative potential. Second, regenerative interfaces may exist between diverse organ systems; populations of cells of neuroectodermal and hematopoietic origin may interact to play a crucial role in normal brain physiology, pathology, and repair. An understanding of the origins of signals and the neural progenitors' responses might lead to the development of effective therapeutic strategies to counterbalance acute and chronic neurodegenerative processes. Such strategies may include modifying and modulating cells with regenerative potential in subtle ways. For example, stem cells might be able to detect pathology-associated signals and be used as "interpreters" to mediate drug and other therapeutic interventions. This review has focused on the role of inflammation in brain repair. We propose that resident astroglia and blood-born cells both contribute to an inflammatory signature that is unique to each kind of neuronal degeneration or injury. These cells play a key role in coordinating the neural progenitor cell response to brain injury by exerting direct and indirect environmentally mediated influence on neural progenitor cells. We suggest that investigations of the neural progenitor-immunologic interface will provide valuable data related to the mechanisms by which endogenous and exogenous neural progenitor cells react to brain pathology, ultimately aiding in the design of more effective therapeutic applications of stem cell biology. Such improvements will include: (1) ascertaining the proper timing for implanting exogenous neural progenitor cells in relation to the administration of anti-inflammatory agents; (2) identifying what types of molecules might be administered during injury to enhance the mobilization and differentiation of endogenous and exogenous neural progenitor cells while also inhibiting the detrimental aspects of the inflammatory reaction; (3) divining clues as to which molecules may be required to change the lesioned environment in order to invite the homing of reparative neural progenitor cells.
Collapse
Affiliation(s)
- F J Mueller
- Program in Developmental Regenerative Cell Biology, The Burnham Institute, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
930
|
Abstract
Inositol, a simple six-carbon sugar, forms the basis of a number of important intracellular signaling molecules. Over the last 35 years, a series of biochemical and cell biological experiments have shown that lithium (Li(+)) reduces the cellular concentration of myo-inositol and as a consequence attenuates signaling within the cell. Based on these observations, inositol-depletion was proposed as a therapeutic mechanism in the treatment of bipolar mood disorder. Recent results have added significant new dimensions to the original hypothesis. However, despite a number of clinical studies, this hypothesis still remains to be either proven or refuted. In this review of our current knowledge, I will consider where the inositol-depletion hypothesis stands today and how it may be further investigated in the future.
Collapse
Affiliation(s)
- A J Harwood
- MRC Laboratory for Molecular Biology, Department of Biology, University College London, London, UK.
| |
Collapse
|
931
|
Komitova M, Eriksson PS. Sox-2 is expressed by neural progenitors and astroglia in the adult rat brain. Neurosci Lett 2004; 369:24-7. [PMID: 15380301 DOI: 10.1016/j.neulet.2004.07.035] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Accepted: 07/12/2004] [Indexed: 11/22/2022]
Abstract
Sox-2 is a transcription factor that is expressed by self-renewing and multipotent stem cells of the embryonic neuroepithelium. Very little is however known about Sox-2 expression in the adult brain and therefore we used immunohistochemistry to examine its distribution and co-localization with specific cell markers. We found that Sox-2 was expressed by actively dividing neural progenitor cells in the neurogenic regions in the adult rat brain, the subventricular zone of the forebrain and the subgranular zone of the dentate gyrus in the hippocampus. Cells expressing immature neuronal markers were essentially Sox-2 immunonegative. Sox-2 was also found to be expressed by glial fibrillary acidic protein immunopositive astroglia, widely distributed in the brain parenchyma. Given the fact that several studies have established the neurogenic capacity of a specialized type of astroglia in the adult brain, the findings of Sox-2 expression in parenchymal astroglia are of potential interest. We conclude that Sox-2 might, in combination with appropriate cell-specific markers, constitute a useful marker to study the in vivo dynamics of the neural progenitor cell compartment also in the adult brain.
Collapse
Affiliation(s)
- Mila Komitova
- The Arvid Carlsson Institute at the Institute of Clinical Neuroscience, Blå Stråket 7, Sahlgrenska University Hospital, SE-413 45 Göteborg, Sweden
| | | |
Collapse
|
932
|
Gurok U, Steinhoff C, Lipkowitz B, Ropers HH, Scharff C, Nuber UA. Gene expression changes in the course of neural progenitor cell differentiation. J Neurosci 2004; 24:5982-6002. [PMID: 15229246 PMCID: PMC6729244 DOI: 10.1523/jneurosci.0809-04.2004] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The molecular changes underlying neural progenitor differentiation are essentially unknown. We applied cDNA microarrays with 13,627 clones to measure dynamic gene expression changes during the in vitro differentiation of neural progenitor cells that were isolated from the subventricular zone of postnatal day 7 mice and grown in vitro as neurospheres. In two experimental series in which we withdrew epidermal growth factor and added the neurotrophins Neurotrophin-4 or BDNF, four time points were investigated: undifferentiated cells grown as neurospheres, and cells 24, 48, and 96 hr after differentiation. Expression changes of selected genes were confirmed by semiquantitative RT-PCR. Ten different groups of gene expression dynamics obtained by cluster analysis are described. To correlate selected gene expression changes to the localization of respective proteins, we performed immunostainings of cultured neurospheres and of brain sections from adult mice. Our results provide new insights into the genetic program of neural progenitor differentiation and give strong hints to as yet unknown cellular communications within the adult subventricular zone stem cell niche.
Collapse
Affiliation(s)
- Ulf Gurok
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
933
|
Krtolica A. Stem cell: balancing aging and cancer. Int J Biochem Cell Biol 2004; 37:935-41. [PMID: 15743668 DOI: 10.1016/j.biocel.2004.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 09/25/2004] [Accepted: 10/04/2004] [Indexed: 10/26/2022]
Abstract
Stem cells are defined by their self-renewing capacity and the ability to differentiate into one or more cell types. Stem cells can be divided, depending on their origin, into embryonic or adult. Embryonic stem cells derive from early stage embryos and can give rise to cells from all three germ layers. Adult stem cells, first identified in hematopoietic tissue, reside in a variety of adult tissues. Under normal physiologic conditions, adult stem cells are capable of differentiating into the limited cell types that comprise the particular tissue or organ. Adult stem cells are responsible for tissue renewal and exhaustion of their replicative capacity may contribute to tissue aging. Loss of unlimited proliferative capacity in some of the adult stem cells and/or their progenitors may have involved the evolutionary trade-off: senescence prevents cancer but may promote aging. Embryonic stem cells exhibit unlimited self-renewal capacity due to the expression of telomerase. Although they possess some cancer cell characteristics, embryonic stem cells exhibit a remarkable resistance to genomic instability and malignant transformation. Understanding the tumor suppressive mechanisms employed by embryonic stem cells may contribute to the development of novel cancer treatments and safe cell-based therapies for age-related diseases.
Collapse
Affiliation(s)
- Ana Krtolica
- Lawrence Berkley National Laboratory, 1 Cyclotron Road, MS 84-171, Berkeley, CA 94720, USA.
| |
Collapse
|
934
|
Schinder AF, Gage FH. A hypothesis about the role of adult neurogenesis in hippocampal function. Physiology (Bethesda) 2004; 19:253-61. [PMID: 15381753 DOI: 10.1152/physiol.00012.2004] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The functional relevance of adult hippocampal neurogenesis has long been a matter of intense experimentation and debate, but the precise role of new neurons has not been sufficiently elaborated. Here we propose a hypothesis in which specific features of newly generated neurons contribute to hippocampal plasticity and function and discuss the most recent and relevant findings in the context of the proposed hypothesis.
Collapse
Affiliation(s)
- Alejandro F Schinder
- Laboratory of Neuronal Plasticity, Leloir Institute Foundation, 1405 Buenos Aires, Argentina.
| | | |
Collapse
|
935
|
Abstract
Neurogenesis occurs in two germinal centres of the adult brain and persists with increasing age, although at a reduced level. This observation, that the mature brain can support neurogenesis, has given rise to the hope that neural stem cells could be used to repair the brain by repopulating regions suffering from neuronal loss as a result of injury or disease. The aging brain is vulnerable to mild cognitive impairment, increasing incidence of stroke, and a variety of neurodegenerative diseases. However, most studies to date have focused on the young adult brain, and relatively little information is available about the regulation of neurogenesis in the aged brain or the potential of using neural stem cells to repair the aged brain. This review summarizes the current state of knowledge on neurogenesis in the young adult brain and discusses the information available on age-related changes in neurogenesis. Possible therapeutic strategies using neural stem cells for repair of the aging brain are considered.
Collapse
Affiliation(s)
- Giovanna M Bernal
- Department of Neuroscience, The Chicago Medical School at, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | | |
Collapse
|
936
|
Abstract
Stem cells in animal tissues are often located and controlled by special tissue microenvironments known as niches. Studies of stem cell niches in model systems such as Drosophila have revealed adhesive interactions, cell cycle modifications and intercellular signals that operate to control stem cell behavior. Candidate niches and regulatory molecules have also been identified in many mammalian tissues, including bone marrow, skin, gut and brain. While niches are an ancient evolutionary device with conserved features across diverse organisms, we suggest that certain niches display important differences in their organization and function.
Collapse
Affiliation(s)
- Benjamin Ohlstein
- Howard Hughes Medical Institute, Carnegie Institution of Washington, Department of Embryology, 115 W. University Pkwy., Baltimore, Maryland 21210, USA
| | | | | | | |
Collapse
|
937
|
|
938
|
Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 2004; 64:7011-21. [PMID: 15466194 DOI: 10.1158/0008-5472.can-04-1364] [Citation(s) in RCA: 1923] [Impact Index Per Article: 91.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transformed stem cells have been isolated from some human cancers. We report that, unlike other brain cancers, the lethal glioblastoma multiforme contains neural precursors endowed with all of the critical features expected from neural stem cells. Similar, yet not identical, to their normal neural stem cell counterpart, these precursors emerge as unipotent (astroglial) in vivo and multipotent (neuronal-astroglial-oligodendroglial) in culture. More importantly, these cells can act as tumor-founding cells down to the clonal level and can establish tumors that closely resemble the main histologic, cytologic, and architectural features of the human disease, even when challenged through serial transplantation. Thus, cells possessing all of the characteristics expected from tumor neural stem cells seem to be involved in the growth and recurrence of adult human glioblastomas multiforme.
Collapse
Affiliation(s)
- Rossella Galli
- Stem Cell Research Institute and Laboratory of Molecular Diagnostics, H. S. Raffaele, Milan, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
939
|
Abstract
Neural stem cells contribute to neurogenesis in both the embryonic and adult brain. However, while adult neural stem cells produce new neurons that populate the olfactory bulb and the granule cell layer of the hippocampus, they do not normally participate in reparative neurogenesis following injury or disease affecting regions distant from the subventricular zone or the dentate gyrus. Here we review differences between neural stem cells found in the embryo and the adult, and describe factors that enhance neuronal output from these cells in vivo. Additionally, we review evidence that neural stem cells can be transplanted into injured regions of the adult brain to enhance compensatory neurogenesis from endogenous precursors. Pre-differentiation of neural stem cells into immature neurons prior to transplantation can also aid in functional recovery following injury or disease.
Collapse
Affiliation(s)
- Christine Y Brazel
- Laboratory of Neurosciences, National Institute on Aging, 333 Cassell Dr., Triad 406A, Baltimore, MD 21224, USA.
| | | |
Collapse
|
940
|
Coles BLK, Angénieux B, Inoue T, Del Rio-Tsonis K, Spence JR, McInnes RR, Arsenijevic Y, van der Kooy D. Facile isolation and the characterization of human retinal stem cells. Proc Natl Acad Sci U S A 2004; 101:15772-7. [PMID: 15505221 PMCID: PMC524825 DOI: 10.1073/pnas.0401596101] [Citation(s) in RCA: 287] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This study identifies and characterizes retinal stem cells (RSCs) in early postnatal to seventh-decade human eyes. Different subregions of human eyes were dissociated and cultured by using a clonal sphere-forming assay. The stem cells were derived only from the pars plicata and pars plana of the retinal ciliary margin, at a frequency of approximately 1:500. To test for long-term self-renewal, both the sphere assay and monolayer passaging were used. By using the single sphere passaging assay, primary spheres were dissociated and replated, and individual spheres demonstrated 100% self-renewal, with single spheres giving rise to one or more new spheres in each subsequent passage. The clonal retinal spheres were plated under differentiation conditions to assay the differentiation potential of their progeny. The spheres were produced all of the different retinal cell types, demonstrating multipotentiality. Therefore, the human eye contains a small population of cells (approximately equal to 10,000 cells per eye) that have retinal stem-cell characteristics (proliferation, self-renewal, and multipotentiality). To test the in vivo potential of the stem cells and their progeny, we transplanted dissociated human retinal sphere cells, containing both stem cells and progenitors, into the eyes of postnatal day 1 NOD/SCID mice and embryonic chick eyes. The progeny of the RSCs were able to survive, migrate, integrate, and differentiate into the neural retina, especially as photoreceptors. Their facile isolation, integration, and differentiation suggest that human RSCs eventually may be valuable in treating human retinal diseases.
Collapse
Affiliation(s)
- Brenda L K Coles
- Departments of Medical Genetics and Microbiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario, Canada M5S 1A8
| | | | | | | | | | | | | | | |
Collapse
|
941
|
Emsley JG, Mitchell BD, Magavi SSP, Arlotta P, Macklis JD. The repair of complex neuronal circuitry by transplanted and endogenous precursors. Neurotherapeutics 2004. [DOI: 10.1007/bf03206630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
942
|
Abstract
Huntington's disease is an autosomal dominant genetic disease, which results in progressive neuronal degeneration in the neostriatum and neocortex, and associated functional impairments in motor, cognitive, and psychiatric domains. Although the genetic mutation is identified, involving an abnormal CAG expansion within the htt gene on chromosome 4, the mechanism by which this leads to neuronal cell death and the question of why striatal neurones are targeted both remain unknown. Thus, in addition to the search for molecular and genetic strategies to inhibit development of the disease, we still need to identify effective strategies for cellular repair in affected individuals. Aspects of the human neuropathology can be well modeled by excitotoxic or metabolic lesions in experimental animals, and in transgenic mice carrying the htt mutation, providing the basis for testing alternative therapeutic strategies. The rationale and efficacy of alternative cell therapies are reviewed, including transplantation repair with embryonic striatal tissues, expansion and differentiation of striatal-like cells from stem cells, and in vivo and ex vivo gene therapy for delivery of neuroprotective growth factor molecules. Pilot and experimental clinical trials of several approaches are now also underway, and the alternative strategies are compared.
Collapse
Affiliation(s)
- Stephen B Dunnett
- School of Biosciences, Cardiff University, Cardiff CF10 3US, Wales, United Kingdom.
| | | |
Collapse
|
943
|
Chu K, Kim M, Park KI, Jeong SW, Park HK, Jung KH, Lee ST, Kang L, Lee K, Park DK, Kim SU, Roh JK. Human neural stem cells improve sensorimotor deficits in the adult rat brain with experimental focal ischemia. Brain Res 2004; 1016:145-53. [PMID: 15246850 DOI: 10.1016/j.brainres.2004.04.038] [Citation(s) in RCA: 180] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2004] [Indexed: 01/10/2023]
Abstract
Ischemic stroke is caused by the interruption of cerebral blood flow that leads to brain damage with long-term sensorimotor deficits. Stem cell transplantation may recover functional deficit by replacing damaged brain. In this study, we attempted to test whether the human neural stem cells (NSCs) can improve the outcome in the rat brain with intravenous injection and also determine the migration, differentiation and the long-term viabilities of human NSCs in the rat brain. Focal cerebral ischemia was induced by intraluminal thread occlusion of middle cerebral artery (MCA). One day after surgery, the rats were randomly divided into two groups: NSCs-ischemia vs. Ischemia-only. Human NSCs infected with retroviral vector encoding beta galactosidase were intravenously injected in NSCs-ischemia group (5 x 10(6) cells) and the same amount of saline was injected in Ischemia-only group for control. The animals were evaluated for 4 weeks using turning in an alley (TIA) test, modified limb placing test (MLPT) and rotarod test. Transplanted cells were detected by X gal cytohistochemistry or beta gal immunohistochemistry with double labeling of other cell markers. The NSCs-ischemia group showed better performance on TIA test at 2 weeks, and MLPT and rotarod test from 3 weeks after ischemia compared with the Ischemia-only group. Human NSCs were detected in the lesion side and labeled with marker for neurons or astrocytes. Postischemic hemispheric atrophy was noted but reduced in NSCs-ischemia group. X gal+ cells were detected in the rat brain as long as 540 days after transplantation. Our data suggest intravenously transplanted human NSCs can migrate and differentiate in the rat brain with focal ischemia and improve functional recovery.
Collapse
Affiliation(s)
- Kon Chu
- Department of Neurology, Stroke and Neural Stem Cell Laboratory in Clinical Research Institute, Seoul National University Hospital, Seoul National University, 28, Yongon-Dong, Chongro-Gu, Seoul 110-744, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
944
|
Lindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neurodegenerative disorders-how to make it work. Nat Med 2004; 10 Suppl:S42-50. [PMID: 15272269 DOI: 10.1038/nm1064] [Citation(s) in RCA: 676] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Accepted: 03/30/2004] [Indexed: 02/08/2023]
Abstract
Recent progress shows that neurons suitable for transplantation can be generated from stem cells in culture, and that the adult brain produces new neurons from its own stem cells in response to injury. These findings raise hope for the development of stem cell therapies in human neurodegenerative disorders. Before clinical trials are initiated, we need to know much more about how to control stem cell proliferation and differentiation into specific phenotypes, induce their integration into existing neural and synaptic circuits, and optimize functional recovery in animal models closely resembling the human disease.
Collapse
Affiliation(s)
- Olle Lindvall
- Laboratory of Neurogenesis and Cell Therapy, Section of Restorative Neurology, Department of Clinical Neuroscience, Wallenberg Neuroscience Center, University Hospital, SE-221 84 Lund, Sweden.
| | | | | |
Collapse
|
945
|
Peterson DA. Umbilical cord blood cells and brain stroke injury: bringing in fresh blood to address an old problem. J Clin Invest 2004; 114:312-4. [PMID: 15286794 PMCID: PMC484988 DOI: 10.1172/jci22540] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Degeneration of brain tissue following stroke leads to functional impairment with limited brain self-repair. New evidence suggests that delivery of circulating CD34(+) human umbilical cord blood cells can produce functional recovery in an animal stroke model with concurrent angiogenesis and neurogenesis leading to some restoration of cortical tissue. While some alternative interpretations of this data are offered herein, the study provides encouraging evidence of functional recovery from stroke in an animal model using stem cell therapy.
Collapse
Affiliation(s)
- Daniel A Peterson
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA.
| |
Collapse
|
946
|
The Neuroscientist Comments. Neuroscientist 2004. [DOI: 10.1177/107385840401000401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
947
|
Kabos P, Matundan H, Zandian M, Bertolotto C, Robinson ML, Davy BE, Yu JS, Krueger RC. Neural precursors express multiple chondroitin sulfate proteoglycans, including the lectican family. Biochem Biophys Res Commun 2004; 318:955-63. [PMID: 15147965 DOI: 10.1016/j.bbrc.2004.04.114] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2004] [Indexed: 02/05/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) abnormally accumulate in cerebrospinal fluid (CSF) of both human neonates with preterm hydrocephalus, and P8 hydrocephalic mice. We hypothesized CSF CSPGs are synthesized by neural precursors, separated from ventricular CSF by ependyma, which is often disrupted in hydrocephalus. Western blotting demonstrates that neural precursors cultured as neurospheres secrete CSPGs (> 30 microg/ml) into their media which appear to be very similar to these CSF CSPGs. Some CSPGs bear the stage-specific embryonic antigen-1 (ssea-1), associated with embryonic/neural stem cells. Neurospheres transcribe many CSPG genes, including the entire aggrecan/lectican family, phosphacan, and tenascin. Phosphacan can be detected in media by Western blotting. Aggrecan can be detected in media after purification using hyaluronic acid affinity chromatography. During differentiation, neurospheres downregulate CSPGs. This is the first report to show that proliferating neural precursors synthesize lecticans, including aggrecan, which are downregulated with differentiation. These observations suggest novel links between CSPGs and CNS precursor biology.
Collapse
Affiliation(s)
- Peter Kabos
- Department of Pediatrics, Division of Molecular and Human Genetics, Children's Research Institute, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | | | |
Collapse
|
948
|
Divinski I, Mittelman L, Gozes I. A Femtomolar Acting Octapeptide Interacts with Tubulin and Protects Astrocytes against Zinc Intoxication. J Biol Chem 2004; 279:28531-8. [PMID: 15123709 DOI: 10.1074/jbc.m403197200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An octapeptide was previously described that protects neurons against a wide variety of insults directly and indirectly as a result of interactions (at femtomolar concentrations) with supporting glial cells. The current study set out to identify the octapeptide binding molecules so as to understand the high affinity mechanisms of cellular protection. Studies utilizing affinity chromatography of brain extracts identified tubulin, the brain major protein, as the octapeptide-binding ligand. Dot blot analysis with pure tubulin and the biotinylated octapeptide verified this finding. When added to cerebral cortical astrocytes, the octapeptide (10(-15)-10(-10) m) induced a rapid microtubule reorganization into distinct microtubular structures that were stained by monoclonal tubulin antibodies and visualized by confocal microscopy. Fluorescein-labeled octapeptide induced a similar change and was detected in the intracellular milieu, even when cells were incubated at 4 degrees C or at low pH. In a cell-free system, the octapeptide stimulated tubulin assembly into microtubules. Furthermore, treatment of astrocytes with zinc chloride resulted in microtubule disassembly and cell death that was protected by the octapeptide. In conclusion, the results suggest that the octapeptide crosses the plasma membrane and interacts directly with tubulin, the microtubule subunit, to induce microtubule reorganization and improved survival. Because microtubules are the key component of the neuronal and glial cytoskeleton that regulates cell division, differentiation, and protection, this finding may explain the breadth and efficiency of the cellular protective capacities of the octapeptide.
Collapse
Affiliation(s)
- Inna Divinski
- Department of Clinical Biochemistry and Interdepartmental Core Facility, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | |
Collapse
|
949
|
Linazasoro G. [Investigation in stem cells]. Med Clin (Barc) 2004; 123:93-4. [PMID: 15225473 DOI: 10.1016/s0025-7753(04)74423-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
950
|
Höglinger GU, Rizk P, Muriel MP, Duyckaerts C, Oertel WH, Caille I, Hirsch EC. Dopamine depletion impairs precursor cell proliferation in Parkinson disease. Nat Neurosci 2004; 7:726-35. [PMID: 15195095 DOI: 10.1038/nn1265] [Citation(s) in RCA: 696] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Accepted: 04/26/2004] [Indexed: 01/19/2023]
Abstract
Cerebral dopamine depletion is the hallmark of Parkinson disease. Because dopamine modulates ontogenetic neurogenesis, depletion of dopamine might affect neural precursors in the subependymal zone and subgranular zone of the adult brain. Here we provide ultrastructural evidence showing that highly proliferative precursors in the adult subependymal zone express dopamine receptors and receive dopaminergic afferents. Experimental depletion of dopamine in rodents decreases precursor cell proliferation in both the subependymal zone and the subgranular zone. Proliferation is restored completely by a selective agonist of D2-like (D2L) receptors. Experiments with neural precursors from the adult subependymal zone grown as neurosphere cultures confirm that activation of D2L receptors directly increases the proliferation of these precursors. Consistently, the numbers of proliferating cells in the subependymal zone and neural precursor cells in the subgranular zone and olfactory bulb are reduced in postmortem brains of individuals with Parkinson disease. These observations suggest that the generation of neural precursor cells is impaired in Parkinson disease as a consequence of dopaminergic denervation.
Collapse
|