Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Electr Bioimpedance ; 12(1): 34-49, 2021 Jan.
Article in English | MEDLINE | ID: mdl-34966467

ABSTRACT

We present here the first impedance-based characterization of the differentiation process of two human mesencephalic fetal neural stem lines. The two dopaminergic neural stem cell lines used in this study, Lund human mesencephalic (LUHMES) and human ventral mesencephalic (hVM1 Bcl-XL), have been developed for the study of Parkinsonian pathogenesis and its treatment using cell replacement therapy. We show that if only relying on impedance magnitude analysis, which is by far the most usual approach in, e.g., cytotoxicity evaluation and drug screening applications, one may not be able to distinguish whether the neural stem cells in a population are proliferating or differentiating. However, the presented results highlight that equivalent circuit analysis can provide detailed information on cellular behavior, e.g. simultaneous changes in cell morphology, cell-cell contacts, and cell adhesion during formation of neural projections, which are the fundamental behavioral differences between proliferating and differentiating neural stem cells. Moreover, our work also demonstrates the sensitivity of impedance-based monitoring with capability to provide information on changes in cellular behavior in relation to proliferation and differentiation. For both of the studied cell lines, in already two days (one day after induction of differentiation) equivalent circuit analysis was able to show distinction between proliferation and differentiation conditions, which is significantly earlier than by microscopic imaging. This study demonstrates the potential of impedance-based monitoring as a technique of choice in the study of stem cell behavior, laying the foundation for screening assays to characterize stem cell lines and testing the efficacy epigenetic control.

2.
Mol Neurobiol ; 56(6): 4023-4036, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30259399

ABSTRACT

Amyloid-ß 42 [Aß1-42 (Aß42)] is one of the main Aß peptide isoforms found in amyloid plaques of brains with Alzheimer's disease (AD). Although Aß42 is associated with neurotoxicity, it might mediate several normal physiological processes during embryonic brain development and in the adult brain. However, due to the controversy that exists in the field, relatively little is known about its physiological function. In the present work, we have analyzed the effects of different concentrations of monomeric Aß42 on cell death, proliferation, and cell fate specification of human neural stem cells (hNSCs), specifically the hNS1 cell line, undergoing differentiation. Our results demonstrate that at higher concentrations (1 µM), Aß42 increases apoptotic cell death and DNA damage, indicating that prolonged exposure of hNS1 cells to higher concentrations of Aß42 is neurotoxic. However, at lower concentrations, Aß42 significantly promotes cell proliferation and glial cell specification of hNS1 cells by increasing the pool of proliferating glial precursors, without affecting neuronal differentiation, in a concentration-dependent manner. At the molecular level, these effects could be mediated, at least in part, by GSK3ß, whose expression is increased by treatment with Aß42 and whose inhibition prevents the glial specification induced by Aß42. Since the cellular and molecular effects are known to appear decades before the first clinical symptoms, these types of studies are important in discovering the underlying pathophysiological processes involved in the development of AD. This knowledge could then be used in diagnosing the disease at early stages and be applied to the development of new treatment options.


Subject(s)
Amyloid beta-Peptides/toxicity , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neuroglia/pathology , Peptide Fragments/toxicity , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Cell Proliferation/drug effects , Humans , Neural Stem Cells/drug effects , Neuroglia/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects
3.
J Neurovirol ; 21(4): 346-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25851778

ABSTRACT

Cytomegalovirus (CMV) is the most common cause of congenital infection in developed countries and a major cause of neurological disability in children. Although CMV can affect multiple organs, the most important sequelae of intrauterine infection are related to lesions of the central nervous system. However, little is known about the pathogenesis and the cellular events responsible for neuronal damage in infants with congenital infection. Some studies have demonstrated that neural precursor cells (NPCs) show the greatest susceptibility to CMV infection in the developing brain. We sought to establish an in vitro model of CMV infection of the developing brain in order to analyze the cellular events associated with invasion by this virus. To this end, we employed two cell lines as a permanent source of NPC, avoiding the continuous use of human fetal tissue, the human SK-N-MC neuroblastoma cell line, and an immortalized cell line of human fetal neural origin, hNS-1. We also investigated the effect of the differentiation stage in relation to the susceptibility of these cell lines by comparing the neuroblastoma cell line with the multipotent cell line hNS-1. We found that the effects of the virus were more severe in the neuroblastoma cell line. Additionally, we induced hNS-1 to differentiate and evaluated the effect of CMV in these differentiated cells. Like SK-N-MC cells, hNS-1-differentiated cells were also susceptible to infection. Viability of differentiated hNS-1 cells decreased after CMV infection in contrast to undifferentiated cells. In addition, differentiated hNS-1 cells showed an extensive cytopathic effect whereas the effect was scarce in undifferentiated cells. We describe some of the effects of CMV in neural stem cells, and our observations suggest that the degree of differentiation is important in the acquisition of susceptibility.


Subject(s)
Cytomegalovirus Infections/virology , Neural Stem Cells/cytology , Neural Stem Cells/virology , Cell Differentiation/physiology , Cell Line , Humans , Immunohistochemistry , Polymerase Chain Reaction
4.
Cell Death Differ ; 14(11): 1880-92, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17673921

ABSTRACT

Understanding basic processes of human neural stem cell (hNSC) biology and differentiation is crucial for the development of cell replacement therapies. Bcl-X(L) has been reported to enhance dopaminergic neuron generation from hNSCs and mouse embryonic stem cells. In this work, we wanted to study, at the cellular level, the effects that Bcl-X(L) may exert on cell death during differentiation of hNSCs, and also on cell fate decisions and differentiation. To this end, we have used both v-myc immortalized (hNS1 cell line) and non-immortalized neurosphere cultures of hNSCs. In culture, using different experimental settings, we have consistently found that Bcl-X(L) enhances neuron generation while precluding glia generation. These effects do not arise from a glia-to-neuron shift (changes in fate decisions taken by precursors) or by only cell death counteraction, but, rather, data point to Bcl-X(L) increasing proliferation of neuronal progenitors, and inhibiting the differentiation of glial precursors. In vivo, after transplantation into the aged rat striatum, Bcl-X(L) overexpressing hNS1 cells generated more neurons and less glia than the control ones, confirming the results obtained in vitro. These results indicate an action of Bcl-X(L) modulating hNSCs differentiation, and may be thus important for the future development of cell therapy strategies for the diseased mammalian brain.


Subject(s)
Neurons/cytology , Stem Cells/cytology , bcl-X Protein/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Corpus Striatum/cytology , Humans , Intermediate Filament Proteins/isolation & purification , Intermediate Filament Proteins/metabolism , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Neuroglia/cytology , Neuroglia/physiology , Neurons/physiology , Rats , Stem Cell Transplantation , Stem Cells/physiology , Tubulin/isolation & purification , Tubulin/metabolism , bcl-X Protein/isolation & purification
5.
Exp Cell Res ; 313(3): 588-601, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17156776

ABSTRACT

Isolation and expansion of neural stem cells (NSCs) of human origin are crucial for successful development of cell therapy approaches in neurodegenerative diseases. Different epigenetic and genetic immortalization strategies have been established for long-term maintenance and expansion of these cells in vitro. Here we report the generation of a new, clonal NSC (hc-NSC) line, derived from human fetal cortical tissue, based on v-myc immortalization. Using immunocytochemistry, we show that these cells retain the characteristics of NSCs after more than 50 passages. Under proliferation conditions, when supplemented with epidermal and basic fibroblast growth factors, the hc-NSCs expressed neural stem/progenitor cell markers like nestin, vimentin and Sox2. When growth factors were withdrawn, proliferation and expression of v-myc and telomerase were dramatically reduced, and the hc-NSCs differentiated into glia and neurons (mostly glutamatergic and GABAergic, as well as tyrosine hydroxylase-positive, presumably dopaminergic neurons). RT-PCR analysis showed that the hc-NSCs retained expression of Pax6, Emx2 and Neurogenin2, which are genes associated with regionalization and cell commitment in cortical precursors during brain development. Our data indicate that this hc-NSC line could be useful for exploring the potential of human NSCs to replace dead or damaged cortical cells in animal models of acute and chronic neurodegenerative diseases. Taking advantage of its clonality and homogeneity, this cell line will also be a valuable experimental tool to study the regulatory role of intrinsic and extrinsic factors in human NSC biology.


Subject(s)
Cell Transformation, Viral , Cerebral Cortex/cytology , Fetal Stem Cells/physiology , Neurons/physiology , Oncogene Protein p55(v-myc)/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Clone Cells/physiology , Down-Regulation , Fetal Stem Cells/enzymology , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Oncogene Protein p55(v-myc)/genetics , Phenotype , Telomerase , Tubulin/metabolism
6.
Hip Int ; 14(1): 34-38, 2004.
Article in English | MEDLINE | ID: mdl-28247376

ABSTRACT

Occult fractures of the hip are rare; they are almost absent from published medical literature and the studies that are reported concern fractures in the elderly population. In this paper the cases of two children, nine and 16 years old are presented who suffered occult fractures of the femoral head during high-energy traumatisms. Conventional X-ray did not reveal bone lesions, although it did show a discrete eccentration of the cephalic nucleus and the diagnosis was delayed until more specific image techniques were applied: bone scanning with Tc99, CT and MRI, revealed fracture from compression in Case 1 and osteochondral fracture in Case 2. The treatment consisted of non-weight-bearing of the joint in the first case and removal of the fragment by arthroscopy in the second. The outcome was good in both patients who had no pain in the hip and complete mobility one year later. (Hip International 2004; 14: 34-8).

7.
J Neurosurg ; 94(5): 765-74, 2001 May.
Article in English | MEDLINE | ID: mdl-11354408

ABSTRACT

OBJECT: Immortalized neural progenitor cells derived from embryonic rat hippocampus (HiB5), were transduced ex vivo with the gene for mouse nerve growth factor (NGF) to secrete NGF (NGF-HiB5) at 2 ng/hr/10(5) cells in culture. METHODS: Fifty-nine male Wistar rats weighing 300 to 370 g each were anesthetized with 60 mg/kg sodium pentobarbital and subjected to lateral fluid-percussion brain injury of moderate severity (2.3-2.4 atm, 34 rats) or sham injury (25 rats). At 24 hours postinjury, 2 microl (150,000 cells/microl) of [3H]thymidine-labeled NGF-HiB5 cells were transplanted stereotactically into three individual sites in the cerebral cortex adjacent to the injury site (14 rats). Separate groups of brain-injured rats received nontransfected (naive [n])-HiB5 cells (12 animals) or cell suspension vehicle (eight animals). One week postinjury, animals underwent neurological evaluation for motor function and cognition (Morris water maze) and were killed for histological, autoradiographic, and immunocytochemical analysis. Viable HiB5 cell grafts were identified in all animals, together with reactive microglia and macrophages located throughout the periinjured parenchyma and grafts (OX-42 immunohistochemistry). Brain-injured animals transplanted with either NGF-HiB5 or n-HiB5 cells displayed significantly improved neuromotor function (p < 0.05) and spatial learning behavior (p < 0.005) compared with brain-injured animals receiving microinjections of vehicle alone. A significant reduction in hippocampal CA3 cell death was observed in brain-injured animals receiving transplants of NGF-HiB5 cells compared with those receiving n-HiB5 cells or vehicle (p < 0.025). CONCLUSIONS: This study demonstrates that immortalized neural stem cells that have been retrovirally transduced to produce NGF can markedly improve cognitive and neuromotor function and rescue hippocampal CA3 neurons when transplanted into the injured brain during the acute posttraumatic period.


Subject(s)
Antigens, CD , Antigens, Neoplasm , Antigens, Surface , Avian Proteins , Blood Proteins , Brain Injuries/therapy , Brain Tissue Transplantation , Genetic Therapy/methods , Nerve Growth Factor/genetics , Neurons/transplantation , Stem Cell Transplantation , Animals , Basigin , Behavior, Animal , Brain Injuries/surgery , Cell Line, Transformed/transplantation , Cerebral Cortex/cytology , Cerebral Cortex/surgery , Cognition , Conditioning, Psychological , Gene Expression , Graft Survival , Hippocampus/cytology , Male , Membrane Glycoproteins/analysis , Memory , Motor Activity , Neurologic Examination , Neurons/chemistry , Neurons/cytology , Rats , Rats, Wistar , Recovery of Function , Stem Cells/chemistry , Stem Cells/cytology
8.
Biomed Pharmacother ; 55(2): 91-5, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11293818

ABSTRACT

Because of their ability to generate all the cell types in the nervous system, neural stem cells are promising candidates for the development of cellular and genetic therapies for nervous system disorders and, in particular, neurodegenerative diseases. In recent years, researchers have discovered ways of expanding and perpetuating these cells in culture, as well as different sources for these tissue-specific stem cells, ranging from embryonic to adult tissue, and also from human pluripotent stem cells. Current efforts are oriented to the understanding of the molecular mechanisms controlling their fate decisions, their genetic engineering, and how to harness their potential to make them useful from a therapeutic point of view.


Subject(s)
Neurons/metabolism , Stem Cells/metabolism , Cells, Cultured , Genetic Therapy , Humans , Neurons/physiology , Stem Cells/physiology , Thymidine/metabolism
9.
Curr Gene Ther ; 1(3): 279-99, 2001 Sep.
Article in English | MEDLINE | ID: mdl-12109143

ABSTRACT

The generation of unlimited quantities of neural stem and/or progenitor cells derived from the human brain holds great interest for basic and applied neuroscience. In this article we critically review the origins and recent developments of procedures developed for the expansion, perpetuation, identification, and isolation of human neural precursors, as well as their attributes. Factors influencing their in vitro properties, both under division and after differentiation conditions, are evaluated, with the aim of identifying properties common to the different culture systems reported. This analysis suggests that different culture procedures result in cells with different properties, or even in different cells being isolated. With respect to in vivo performance, present evidence obtained in rodents indicate that cultured human neural precursors, in general, are endowed with excellent integrative properties. Differentiation of the implanted cells, in particular in the case of adult recipients, seems not to be complete, and functionality still needs to be demonstrated. In relation to gene transfer and therapy, aspects currently underexplored, initial data support the view that human neural stem and progenitor cells may serve a role as a platform cell for the delivery of bioactive substances to the diseased CNS. Although a large deal of basic research remains to be done, available data illustrate the enormous potential that human neural precursors isolated, expanded, and characterized in vitro hold for therapeutic applications. In spite of this potential, maintaining a critical view on many unresolved questions will surely help to drive this research field to a good end, that is, the development of real therapies for diseases of the human nervous system.


Subject(s)
Central Nervous System/physiology , Neuroglia/physiology , Neurons/physiology , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Central Nervous System/cytology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/therapy , Genetic Therapy , Humans , Neuroglia/cytology , Neurons/cytology , Rats , Stem Cells/cytology
10.
Mol Cell Neurosci ; 16(1): 1-13, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10882478

ABSTRACT

Human neural stem cells (HNSCs) may serve as a cellular vehicle for molecular therapies as well as for cell replacement in the human CNS. The survival, integration, and differentiation of HNSC.100, a multipotent cell line of HNSCs (A. Villa et al. (2000), Exp. Neurol. 161, 67-84), conditionally perpetuated by genetic and epigenetic means, was investigated after transplantation to the striatum and substantia nigra of the adult, intact rat brain. These are two key regions in the mammalian brain involved in the control of voluntary movement and motor coordination, among other functions. Soon after transplantation (1 week), the cells had already integrated in a nondisruptive manner into the surrounding tissue and migrated out of the implantation site to different distances depending on graft location (in the range of 0.5-2.5 mm). Cell migration was markedly more extensive in the striatum, where the cells colonized the whole extent of the caudate-putamen, than in the substantia nigra region. The engrafted cells completely downregulated the stem cell marker nestin and, due to their multipotential nature, differentiated and expressed mature neural markers. As expected from cells grafted into nonneurogenic regions of the intact brain, the majority of differentiated cells expressed GFAP (astroglia), but expression of other markers, like GalC (oligodendroglia) and MAP2, beta-tubulin III, NeuN, and NSE (for mature neurons) could also be detected. These results demonstrate that genetically perpetuated HNSCs, once transplanted, find residence in the host brain, where they differentiate, generating mature neural cells in the host, chimeric, adult mammalian brain. HNSCs cell lines may be a highly useful model for the development of humanized systems for cell replacement and/or gene transfer to the CNS, which will likely be strong candidates for future therapeutic application in human neurodegenerative conditions.


Subject(s)
Brain/cytology , Graft Survival , Nerve Tissue Proteins , Neurons/cytology , Neurons/transplantation , Stem Cell Transplantation , Stem Cells/cytology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Autoradiography , Brain/metabolism , Brain/surgery , Cell Differentiation , Cell Line , Corpus Striatum/cytology , Corpus Striatum/metabolism , Corpus Striatum/surgery , Down-Regulation , Female , Humans , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Nestin , Neurons/metabolism , Phenotype , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Substantia Nigra/cytology , Substantia Nigra/metabolism , Substantia Nigra/surgery , Transplantation, Heterologous
12.
Endocrinology ; 141(3): 1073-82, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10698183

ABSTRACT

A single injection of estradiol valerate induces a form of cystic ovary resembling some aspects of the human polycystic ovarian syndrome. Preceding the development of follicular cysts, there is an increase in intraovarian synthesis of nerve growth factor (NGF) and the low affinity NGF receptor (p75 NGFR). Selective blockade of NGF actions and p75 NGFR synthesis in the ovary restored estrous cyclicity and ovulatory capacity in estradiol valerate-treated rats, suggesting that an increase in NGF-dependent, p75 NGFR-mediated actions within the ovary contributes to the development of cystic ovarian disease. We have tested this hypothesis by grafting NGF-producing neural progenitor cells into the ovary of juvenile rats that have been induced to ovulate precociously by a single injection of PMSG. The NGF-producing cells, detected by their content of immunoreactive p75 NGFR material, were found scattered throughout the ovary with some of them infiltrating the granulosa cell compartment of large, precystic follicles. Ovarian NGF content was 2-fold higher than in the ovary of rats receiving control cells. Estrous cyclicity was disrupted, with the animals showing prolonged periods of persistent estrus, and an almost continuous background of vaginal cornified cells at other phases of the estrous cycle. Morphometric analysis revealed that the presence of NGF-producing cells neither reduced the total number of corpora lutea per ovary nor significantly increased the formation of follicular cysts. However, the ovaries receiving these cells showed an increased incidence of precystic, type III follicles, accompanied by a reduced number of healthy antral follicles, and an increased size of both healthy and atretic follicles. These changes in follicular dynamics were accompanied by a selective increase in serum androstenedione levels. The results show that an abnormally elevated production of NGF within the ovary suffices to initiate several of the structural and functional alterations associated with the development of follicular cysts in the rat ovary.


Subject(s)
Androgens/metabolism , Estrus/drug effects , Nerve Growth Factors/physiology , Ovary/physiology , Androgens/blood , Androstenedione/metabolism , Animals , Cell Transplantation/physiology , Enzyme-Linked Immunosorbent Assay , Female , Gene Transfer Techniques , Immunohistochemistry , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Ovary/drug effects , Ovary/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Radioimmunoassay , Rats , Stem Cells/drug effects , Stem Cells/metabolism , Testosterone/metabolism , Time Factors
13.
Exp Neurol ; 161(1): 67-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683274

ABSTRACT

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


Subject(s)
Cell Culture Techniques/methods , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Nerve Tissue Proteins , Neurons/cytology , Stem Cells/cytology , Blotting, Southern , Brain/cytology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Transformed , Central Nervous System/cytology , Gene Expression Regulation, Viral , Genes, gag/physiology , Genes, myc/physiology , Genetic Therapy , Glial Fibrillary Acidic Protein/analysis , Humans , Intermediate Filament Proteins/analysis , Microtubule-Associated Proteins/analysis , Nerve Degeneration , Nerve Regeneration , Nestin , Neurons/chemistry , Neurons/drug effects , Phenotype , Proliferating Cell Nuclear Antigen/analysis , Retroviridae/genetics , Stem Cells/chemistry , Stem Cells/drug effects , Transfection , Tubulin/analysis , Vimentin/analysis
14.
Gene Ther ; 6(11): 1851-66, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10602381

ABSTRACT

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


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebellum/metabolism , Gene Transfer Techniques , Hippocampus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebellum/cytology , Female , Hippocampus/cytology , Immunohistochemistry , Neurons/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells
15.
Eur J Neurosci ; 10(7): 2353-70, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9749764

ABSTRACT

Impairments in learning and memory, induced by surgical or excitotoxic lesions of the septo-hippocampal or basalo-cortical pathways, can be ameliorated by grafts of cholinergic-rich foetal basal forebrain tissue into the hippocampus and/or neocortex. However, the effects of such grafts have been only partial, which may be due to the non-specific nature of the lesioning procedures used in these studies, known to destroy both cholinergic and non-cholinergic neuronal projections. In the present study, we have explored the effects of cholinergic-rich grafts in rats subjected to selective cholinergic lesions, induced by intraventricular injections of the immunotoxin 192 IgG-saporin. This lesion, which selectively destroyed 85-95% of the cholinergic neurons in both the septal-diagonal band and nucleus basalis, produced a long-lasting, substantial impairment in both the acquisition of spatial reference memory in the Morris water maze task and delay-dependent short-term memory performance, as seen in a delayed matching-to-position test. Foetal cholinergic grafts (but not control grafts of cerebellar tissue) implanted at multiple sites into both the hippocampus and fronto-parietal neocortex, bilaterally, completely reversed the acquisition deficit in place navigation in the water maze, to an extent that greatly exceeded that previously seen in animals with non-selective lesions. Most notably, however, the impairment in short-term memory was only partially and inconsistently affected, and only at the longest delay times. The morphological analysis, performed at about 7 months after transplantation, showed that the grafts had re-established a close to normal cholinergic innervation in the initially denervated cortical and hippocampal territories. It is proposed that the differential effects of cholinergic-rich transplants on different aspects of cognitive performance may define intrinsic limitations to the functional capacity of the ectopically placed grafts, which may be due to incomplete integration of the grafted cholinergic neurons into functional regulatory circuitries normally available to the basal forebrain cholinergic system.


Subject(s)
Cerebral Cortex/surgery , Fetal Tissue Transplantation , Hippocampus/surgery , Maze Learning/physiology , Memory Disorders/surgery , Prosencephalon/embryology , Acetylcholinesterase/metabolism , Animals , Behavior, Animal/physiology , Cerebral Cortex/enzymology , Female , Hippocampus/enzymology , Memory, Short-Term/physiology , Neuropsychological Tests , Rats , Rats, Sprague-Dawley , Swimming
16.
Eur J Neurosci ; 10(1): 263-71, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9753135

ABSTRACT

Nerve growth factor (NGF) is a maintenance factor for cholinergic neurones in the brain, but its properties as a developmental survival factor are largely unknown. The low accessibility of the developing mammalian brain to experimental manipulation makes it difficult to increase NGF levels during the early phases of brain development. In the present study we have used an in utero, ex-vivo gene transfer approach to explore NGF actions during development of the cholinergic system in the rat brain. Significantly increased numbers of cholinergic neurones were found only in the mesopontine complex in animals receiving NGF-secreting transplants, whereas the cholinergic neurones in the basal forebrain and striatum were not clearly affected. The present results suggest that overexpression of NGF during development may promote the survival of distinct populations of central cholinergic neurones into adulthood.


Subject(s)
Brain Tissue Transplantation , Gene Transfer Techniques , Nerve Growth Factors/genetics , Prosencephalon/embryology , Stem Cell Transplantation , Animals , Behavior, Animal/physiology , Cell Survival/drug effects , Choline O-Acetyltransferase/analysis , Cholinergic Fibers/chemistry , Cholinergic Fibers/enzymology , Corpus Striatum/cytology , Corpus Striatum/embryology , Female , Maze Learning/physiology , Mesencephalon/cytology , Mesencephalon/embryology , Neurons/cytology , Neurons/enzymology , Neurons/ultrastructure , Pons/cytology , Pons/embryology , Pregnancy , Prenatal Exposure Delayed Effects , Prosencephalon/cytology , Rats , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/analysis
17.
Eur J Neurosci ; 10(6): 2026-36, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9753090

ABSTRACT

The objective of the present study was to explore whether grafted immortalized neural stem cells, genetically modified to secrete nerve growth factor (NGF), can ameliorate neuronal death in the adult rat striatum following transient middle cerebral artery occlusion (MCAO). One week after cell implantation in the striatum, animals were subjected to 30 min of MCAO. Striatal damage was evaluated at the cellular level after 48 h of recirculation using immunocytochemical and stereological techniques. The ischaemic insult caused an extensive degeneration of projection neurons, immunoreactive for dopamine- and adenosine 3': 5'-monophosphate-regulated phosphoprotein with a molecular weight of 32 kilodaltons (DARPP-32). 3H-Thymidine autoradiography demonstrated surviving grafted cells in the lesioned striatum in all transplanted rats. The loss of striatal projection neurons was significantly reduced (by an average of 45%) in animals with NGF-secreting grafts, whereas control cells, not producing NGF, had no effect. The neuroprotective action of NGF-secreting grafts was also observed when the total number of striatal neurons immunopositive for the neuronal marker NeuN was quantified, as well as in cresyl violet-stained sections. The present findings indicate that administration of NGF by ex vivo gene transfer and grafting of neural stem cells can ameliorate death of striatal projection neurons caused by transient focal ischaemia.


Subject(s)
Brain Ischemia/surgery , Corpus Striatum/surgery , Nerve Growth Factors/metabolism , Neurons/transplantation , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Death/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Male , Nervous System/physiopathology , Neurons/metabolism , Neurons/physiology , Rats , Rats, Wistar
18.
J Pediatr Orthop B ; 7(3): 210-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9702671

ABSTRACT

The results of three different surgical procedures for the correction of bunions on 25 adolescent feet (distal soft-tissue procedure, proximal closing-wedge osteotomy of the first metatarsal with distal soft-tissue procedure, and proximal phalangeal osteotomy) are reviewed. The results with an average follow-up of 3.5 years were excellent or good in 21 cases, and fair or poor in 4. The average correction of the metatarsophalangeal angle, the intermetatarsal angle, and the hallux valgus interphalangeus angle was, respectively, 9.88, 3.11, and 0.0 degrees for the first procedure, 21.0, 6.87, and 0.0 degrees for the second, and 11.0 0.9, and 22.5 degrees for the third. We conclude that distal soft-tissue reconstruction allows only mild to moderate correction of the metatarsophalangeal and intermetatarsal angles, and the association of a proximal osteotomy of the first metatarsal produces an statistically significant better correction (p < 0.05). Hallux valgus interphalangeus deformities are corrected only by proximal phalanx osteotomies. The selection of a particular procedure should therefore be based on an appropriate preoperative planification.


Subject(s)
Hallux Valgus/surgery , Orthopedics/methods , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Hallux Valgus/diagnostic imaging , Hallux Valgus/physiopathology , Humans , Male , Metatarsophalangeal Joint/physiopathology , Pain Measurement , Patient Satisfaction , Radiography , Range of Motion, Articular , Treatment Outcome
19.
Neuroscience ; 84(4): 1113-25, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9578399

ABSTRACT

Expression of p75 neurotrophin receptor and survival of medium-sized spiny projection neurons and cholinergic interneurons in the rat striatum were studied using immunocytochemistry at different times after transient, unilateral middle cerebral artery occlusion. Thirty minutes of middle cerebral artery occlusion caused a major loss of projection neurons, identified by their immunoreactivity to dopamine- and adenosine 3':5'-monophosphate-regulated phosphoprotein with a molecular weight of 32,000, in the lateral part of the striatum, as observed at 48 h following the insult with no further change at one week. In contrast, no reduction of the number of choline acetyltransferase-positive, cholinergic interneurons, which also expressed TrkA, was detected at either time-point. At 48 h following middle cerebral artery occlusion, expression of p75 neurotrophin receptor was observed in striatal cells which, by the use of double-label immunostaining, were identified as the cholinergic interneurons. No p75 neurotrophin receptor immunoreactivity remained in cholinergic cells after one week of reperfusion. Based on current hypotheses regarding the function of the p75 neurotrophin receptor, the transient expression of this receptor in striatal cholinergic interneurons might contribute to their high resistance to ischemic neuronal death. However, the expression of p75 neurotrophin receptor could also be a first step in a pathway leading to apoptosis, which is inhibited after the present insult due to concomitant activation of TrkA.


Subject(s)
Brain Ischemia/metabolism , Neostriatum/metabolism , Neurons/metabolism , Parasympathetic Nervous System/metabolism , Phosphoproteins , Receptors, Nerve Growth Factor/biosynthesis , Animals , Blood Pressure/physiology , Cerebral Arteries/physiology , Choline O-Acetyltransferase/biosynthesis , Dopamine and cAMP-Regulated Phosphoprotein 32 , Immunohistochemistry , Interneurons/metabolism , Male , Neostriatum/cytology , Nerve Tissue Proteins/biosynthesis , Parasympathetic Nervous System/cytology , Proto-Oncogene Proteins/biosynthesis , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, Nerve Growth Factor , Receptor, trkA , gamma-Aminobutyric Acid/physiology
20.
Proc Natl Acad Sci U S A ; 95(4): 1858-63, 1998 Feb 17.
Article in English | MEDLINE | ID: mdl-9465107

ABSTRACT

Nerve growth factor (NGF) is able to restore spatial learning and reverse forebrain cholinergic neuron atrophy when administered intracerebrally to behaviorally impaired aged rats. In the present study, behaviorally unimpaired, middle-aged rats (14-16 months old) received transplants of ex vivo transduced, clonal NGF-secreting immortalized neural progenitor cells, bilaterally in the nucleus basalis and septum. During the subsequent 9 months the aged control animals developed the expected impairment in spatial learning in the water maze task, whereas the animals with NGF-secreting grafts maintained a performance level not different from the 12-month-old control rats. The marked age-induced atrophy (-25%) of the cholinergic neurons in medial septum and nucleus basalis, seen in the aged control rats, was not present in the NGF-treated aged animals. 3H-labeled thymidine autoradiography showed that the transduced cells survived well and had become integrated into the host tissue surrounding the injection sites, and reverse transcription-PCR analysis revealed expression of the NGF transgene, at both 4 and 9 months postgrafting, in the grafted tissue. The results show that long-term supply of NGF from ex vivo transduced immortalized neural progenitor cells locally within the nucleus basalis and septum can prevent the subsequent development of age-dependent neuronal atrophy and behavioral impairments when the animals reach advanced age.


Subject(s)
Aging , Cognition/drug effects , Nerve Growth Factors/pharmacology , Neurodegenerative Diseases/prevention & control , Animals , Behavior, Animal/physiology , Cholinergic Fibers/pathology , Female , Gene Transfer Techniques , Nerve Growth Factors/administration & dosage , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/metabolism , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...