Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
J Cutan Pathol ; 51(7): 485-489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38549288

ABSTRACT

We described an unusual combination of fibroblastic connective nevus (FCTN) already present at birth with underlying vascular anomalies. Overall, the lesion appeared as a large purplish-brown mass in the groin region up to the third of the right thigh, with partial spontaneous regression during the first three months of life. The FCTN observed exhibited several unusual characteristics: it was congenital, large in size, and located in the lower limbs. Finally, it represented the first case described in which an FCTN arose in association with vascular anomalies.


Subject(s)
Nevus , Skin Neoplasms , Humans , Skin Neoplasms/pathology , Nevus/pathology , Vascular Malformations/pathology , Male , Female , Infant, Newborn , Infant
2.
Proc Natl Acad Sci U S A ; 115(4): E762-E771, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311338

ABSTRACT

Increasing evidence suggests that early neurodevelopmental defects in Huntington's disease (HD) patients could contribute to the later adult neurodegenerative phenotype. Here, by using HD-derived induced pluripotent stem cell lines, we report that early telencephalic induction and late neural identity are affected in cortical and striatal populations. We show that a large CAG expansion causes complete failure of the neuro-ectodermal acquisition, while cells carrying shorter CAGs repeats show gross abnormalities in neural rosette formation as well as disrupted cytoarchitecture in cortical organoids. Gene-expression analysis showed that control organoid overlapped with mature human fetal cortical areas, while HD organoids correlated with the immature ventricular zone/subventricular zone. We also report that defects in neuroectoderm and rosette formation could be rescued by molecular and pharmacological approaches leading to a recovery of striatal identity. These results show that mutant huntingtin precludes normal neuronal fate acquisition and highlights a possible connection between mutant huntingtin and abnormal neural development in HD.


Subject(s)
Huntington Disease/physiopathology , Neurogenesis , Cell Line , Cell Polarity , Humans , Huntington Disease/genetics , Induced Pluripotent Stem Cells , Telencephalon/cytology
3.
Gene Ther ; 20(6): 678-85, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23151521

ABSTRACT

Current therapeutic strategies for Huntington's disease (HD) are focused on symptom management of disease progression. Transcriptional dysregulation is one of the major characteristics in HD. REST is a transcriptional repressor that silences gene expression through binding to RE1/NRSE sites found in the regulatory regions of numerous neuronal genes. Dysregulation of REST and its targeted genes has been reported in different cell and mouse HD models, as well as in biopsies from human patients. In this work, we characterized transcriptional dysregulation associated with REST in two different HD mouse models and assessed the therapeutic effect of interfering with REST function by overexpressing a dominant-negative form (DN:REST). We show that delivery of DN:REST in the motor cortex restores brain-derived neurotrophic factor (BDNF) mRNA and protein levels by reducing endogenous REST occupancy at the Bdnf locus. Similarly, expression of other REST-regulated genes such as Synapsin I (Syn1), Proenkephalin (Penk1) and Cholinergic receptor muscarinic 4 (Chrm4) were restored to normal levels while non-REST-regulated genes were unaffected. This is the first study conducted to investigate REST's role in vivo in a neurodegenerative disease. Our data show that DN:REST in motor cortex reversed RESTs repressive effects on target genes. However, the lack of therapeutic effect on motor function suggests that a more widespread rescue of REST-regulated sites in the affected brain regions may be necessary.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Genetic Therapy , Huntington Disease/genetics , Repressor Proteins/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Transfer Techniques , Humans , Huntington Disease/pathology , Huntington Disease/therapy , Mice , Motor Cortex/metabolism , Neurons/metabolism , Neurons/pathology , Repressor Proteins/therapeutic use
4.
Nat Med ; 6(4): 447-50, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742153

ABSTRACT

Glioblastomas, the most frequent and malignant of primary brain tumors, have a very poor prognosis. Gene therapy of glioblastomas is limited by the short survival of viral vectors and by their difficulty in reaching glioblastoma cells infiltrating the brain parenchyma. Neural stem/progenitor cells can be engineered to produce therapeutic molecules and have the potential to overcome these limitations because they may travel along the white matter, like neoplastic cells, and engraft stably into the brain. Retrovirus-mediated transfer of the gene for interleukin-4 is an effective treatment for rat brain glioblastomas. Here, we transferred the gene for interleukin-4 into C57BL6J mouse primary neural progenitor cells and injected those cells into established syngeneic brain glioblastomas. This led to the survival of most tumor-bearing mice. We obtained similar results by implanting immortalized neural progenitor cells derived from Sprague-Dawley rats into C6 glioblastomas. We also documented by magnetic resonance imaging the progressive disappearance of large tumors, and detected 5-bromodeoxyuridine-labeled progenitor cells several weeks after the injection. These findings support a new approach for gene therapy of brain tumors, based on the grafting of neural stem cells producing therapeutic molecules.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Hematopoietic Stem Cell Transplantation , Interleukin-4/genetics , Neurons/transplantation , Animals , Brain/pathology , Brain Neoplasms/pathology , Cerebral Cortex/cytology , Glioblastoma/pathology , Humans , Interleukin-4/immunology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
5.
Neurobiol Dis ; 34(2): 320-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19236914

ABSTRACT

NS cells are a homogeneous population of neural stem cells which were previously derived from embryonic stem cells as well as from the fetal and adult brain. Our previous reports have described a 21 day long neuronal differentiation protocol able to reproducibly convert adult SVZ-derived NS (aNS) cells into a population composed of 65% mature neurons and 35% glial cells. Here we have developed a different procedure specifically applicable to ES-derived NS cells in order to fully explore their neurogenic capacity. Differently from the aNS differentiation procedure, optimized neuronal output from ES-derived NS cells requires replating of the cells on appropriate substrates followed by sequential exposure to modified media. In these conditions, ES-derived NS cells differentiate into neurons with a barely appreciable quota of astrocytes and occasional oligodendrocytes. In particular, 21 days after the beginning of the treatment, 85% of the cells has differentiated into molecularly and electrophysiologically mature neurons belonging to the GABAergic lineage. The procedure, which is applicable with no considerable differences to different ES-derived NS cell lines and to NS cells at different passages, opens to the possibility of molecular and biochemical studies on close-to-uniform stem cell derived neurons.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Embryonic Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Interneurons/cytology , Interneurons/drug effects , Interneurons/physiology , Mice , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/cytology , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Sodium Channels/drug effects , Sodium Channels/physiology , Stem Cell Transplantation/methods , gamma-Aminobutyric Acid/metabolism
6.
Science ; 293(5529): 493-8, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11408619

ABSTRACT

Huntingtin is a 350-kilodalton protein of unknown function that is mutated in Huntington's disease (HD), a neurodegenerative disorder. The mutant protein is presumed to acquire a toxic gain of function that is detrimental to striatal neurons in the brain. However, loss of a beneficial activity of wild-type huntingtin may also cause the death of striatal neurons. Here we demonstrate that wild-type huntingtin up-regulates transcription of brain-derived neurotrophic factor (BDNF), a pro-survival factor produced by cortical neurons that is necessary for survival of striatal neurons in the brain. We show that this beneficial activity of huntingtin is lost when the protein becomes mutated, resulting in decreased production of cortical BDNF. This leads to insufficient neurotrophic support for striatal neurons, which then die. Restoring wild-type huntingtin activity and increasing BDNF production may be therapeutic approaches for treating HD.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/physiology , Neurons/metabolism , Nuclear Proteins/physiology , 3T3 Cells , Animals , Apoptosis , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Corpus Striatum/cytology , Corpus Striatum/pathology , Exons , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Mutation , Nerve Degeneration , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , Transfection
7.
Neuroscience ; 152(3): 734-40, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18313855

ABSTRACT

An involvement of one particular neurotrophin, namely, the brain-derived neurotrophic factor (BDNF), has been demonstrated in the pathophysiology Huntington's disease. Type-1 cannabinoid (CB1) receptor has been postulated to upregulate BDNF gene transcription. To better understand the relationship between CB1 and BDNF levels in a situation where the striatum is degenerating, we studied, by dual label immunofluorescence, the distribution of CB1 and BDNF in cortical neurons projecting to the striatum in our rat quinolinic acid model of striatal excitotoxicity. We completed our study with quantitative analyses of BDNF protein levels and CB1 binding activity in the cortex. We show that, 2 weeks post lesion, cortical neurons contain more BDNF compared with controls and to earlier time points. Such BDNF up-regulation coincides with a higher binding activity and an increased protein expression of CB1. We suggest that after excitotoxic lesions, CB1 might, at least transiently, upregulate BDNF in the attempt to rescue striatal neurons from degeneration.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Cell Communication/physiology , Cell Survival/physiology , Cerebral Cortex/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Cytoprotection/physiology , Disease Models, Animal , Fluorescent Antibody Technique , Huntington Disease/metabolism , Huntington Disease/physiopathology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurotoxins , Protein Binding , Quinolinic Acid , Rats , Rats, Wistar , Time Factors , Up-Regulation/physiology
8.
AIDS Care ; 20(5): 571-5, 2008 May.
Article in English | MEDLINE | ID: mdl-18484327

ABSTRACT

Recent literature has shown that adherence to HAART is a multi-faceted phenomenon, which involves both behavioural and psychological features. Therefore, the results obtained so far, though promising, have not yet unambiguously identified the factors that could predict non-adherence. Since any support for strengthening the adherence should take into account the HIV+ patients' perception of both their state of health and their relational style, this study tried to identify some psychological characteristics involved in the adherence phenomenon. A self-administered battery of tests including the Attachment Style Questionnaire (ASQ) and the Multidimensional Health Locus of Control Form-C (MHLC-C) was administered to an Italian sample. Results showed significant gender differences between non-adherent and adherent subjects. Specifically, the psychological profile of non-adherent males seemed focused less on relational aspects and perceived relevance of physicians and of 'significant other people', whilst that of non-adherent females seemed more 'relationship-oriented'. This study means to encourage clinicians to plan specific, gender-focused support for enhancing adherence.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/psychology , Patient Compliance/psychology , Adolescent , Adult , Emotions/physiology , Female , HIV Infections/drug therapy , Humans , Italy , Male , Middle Aged , Pilot Projects , Sex Factors , Surveys and Questionnaires
9.
AIDS Care ; 20(4): 495-502, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18449829

ABSTRACT

In the last few years, highly active antiretroviral therapy (HAART) has resulted in a remarkable decrease in HIV-related morbidity and mortality. This "new deal" encouraged clinical research in investigating patients' manifest behaviours and their beliefs regarding their health status, which likely influence not only their treatment-linked behaviours but also their quality of life. Locus of control has been shown to be a construct that can predict and explain health-related behaviours. The Multidimensional Health Locus of Control Form C (MHLC-C) is a condition-specific locus of control scale that can be easily adapted for use with any medical or health-related condition. With the aim to enhance the knowledge about the HIV+ patients' point of view of their complex health condition, this study preliminarily investigated the psychometrics properties of the MHLC-C Italian version and its generalizability across samples defined both by being adherent or not and by gender. Two more samples of chronic patients (Cardiac Surgery and Cancer) were enrolled to better characterize the HIV+ patient's MHLC-C profile. The results showed the validity, reliability and generalizability of the 4-factor structure of MHLC-C. More interestingly, HIV+ subjects revealed a peculiar pattern of beliefs regarding their health condition that clinicians should take into account when managing patients' complex bio-psychosocial condition.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/psychology , Health Status , Internal-External Control , Patient Compliance/psychology , Adolescent , Adult , Chronic Disease , Factor Analysis, Statistical , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Surveys and Questionnaires
10.
Nat Neurosci ; 4(6): 579-86, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11369938

ABSTRACT

Previously we found that the availability of ShcA adapter is maximal in neural stem cells but that it is absent in mature neurons. Here we report that ShcC, unlike ShcA, is not present in neural stem/progenitor cells, but is expressed after cessation of their division and becomes selectively enriched in mature neurons. Analyses of its activity in differentiating neural stem/progenitor cells revealed that ShcC positively affects their viability and neuronal maturation via recruitment of the PI3K-Akt-Bad pathway and persistent activation of the MAPK pathway. We suggest that the switch from ShcA to ShcC modifies the responsiveness of neural stem/progenitor cells to extracellular stimuli, generating proliferation (with ShcA) or survival/differentiation (with ShcC).


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Stem Cells/physiology , Carrier Proteins/metabolism , Cell Death , Cell Survival , Cells, Cultured , Cloning, Molecular , Epidermal Growth Factor/pharmacology , Fetus , Green Fluorescent Proteins , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteins/physiology , Recombinant Fusion Proteins/metabolism , Shc Signaling Adaptor Proteins , Stem Cells/cytology , Telencephalon/cytology , Telencephalon/embryology , Transfection , bcl-Associated Death Protein , src Homology Domains
11.
Neuroscience ; 149(1): 38-52, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17870247

ABSTRACT

A population of mouse embryonic stem (ES)-derived neural stem cells (named NS cells) that exhibits traits reminiscent of radial glia-like cell population and that can be homogeneously expanded in monolayer while remaining stable and highly neurogenic over multiple passages has been recently discovered. This novel population has provided a unique in vitro system in which to investigate physiological events occurring as stem cells lose multipotency and terminally differentiate. Here we analysed the timing, quality and quantity of the appearance of the excitability properties of differentiating NS cells which have been long-term expanded in vitro. To this end, we studied the biophysical properties of voltage-dependent Na(+) currents as an electrophysiological readout for neuronal maturation stages of differentiating NS cells toward the generation of fully functional neurons, since the expression of neuronal voltage-gated Na(+) channels is an essential hallmark of neuronal differentiation and crucial for signal transmission in the nervous system. Using the whole cell and single-channel cell-attached variations of the patch-clamp technique we found that the Na(+) currents in NS cells showed substantial electrophysiological changes during in vitro neuronal differentiation, consisting mainly in an increase of Na(+) current density and in a shift of the steady-state activation and inactivation curves toward more negative and more positive potentials respectively. The changes in the Na(+) channel system were closely related with the ability of differentiating NS cells to generate action potentials, and could therefore be exploited as an appropriate electrophysiological marker of ES-derived NS cells undergoing functional neuronal maturation.


Subject(s)
Action Potentials/physiology , Cell Differentiation/physiology , Ion Channel Gating/physiology , Neurons/physiology , Potassium Channels, Voltage-Gated/metabolism , Stem Cells/physiology , Action Potentials/drug effects , Action Potentials/radiation effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Hippocampus/cytology , Ion Channel Gating/drug effects , Ion Channel Gating/radiation effects , Patch-Clamp Techniques/methods , Potassium Channels, Voltage-Gated/genetics , Rats , Sodium Channel Blockers/pharmacology , Stem Cells/drug effects , Tetrodotoxin/pharmacology , Time Factors
12.
Trends Neurosci ; 14(8): 338-40, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1721737

ABSTRACT

Fetal brain tissue has been shown to have clear behavioral effects when transplanted into adult lesioned brains. These results have focused attention on the cell types of the embryonic brain. Transplantation experiments using primary cells are beginning to define the plasticity of these cells and the times when they become committed to specific neuronal fates. Growth factors have been defined that regulate the proliferation of these cells in culture. Cell lines have been established that express stem cell properties and that are capable of differentiation when implanted into the developing brain. In this article we review this work on mammalian neuroepithelial stem cells and discuss how these studies might contribute to the therapeutic use of brain transplants.


Subject(s)
Brain Tissue Transplantation/physiology , Fetal Tissue Transplantation/physiology , Neurons/physiology , Stem Cells/physiology , Animals , Humans
13.
Trends Neurosci ; 21(11): 476-81, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829689

ABSTRACT

In mammalian systems, SH2-containing cytoplasmic signalling molecules are known to play an important role in determining cell responsiveness to the environment. In particular, following activation of a receptor protein tyrosine kinase (RPTK), proteins like Shc and Grb2 bind to phosphotyrosine residues of stimulated receptors, thereby activating downstream components of specific signalling pathways. The ShcA gene was identified in 1992 and was found to encode three proteins with properties of adaptor molecules coupling RPTKs to Ras. Early data obtained in non-neuronal cells have revealed that Shc and Grb2 proteins are highly expressed and activated in all cells. However, recent analyses of ShcA mRNA and protein in the developing brain revealed progressive downregulation of their expression during differentiation from neuroblasts to neurons. Conversely, the two newly identified Shc homologues (ShcB/Sli and ShcC/Rai) are highly expressed in the mature brain.Thus, variations in the intracellular levels of adaptor proteins might represent one of the mechanisms by which a differentiating cell changes its ability to respond to a given factor, allowing a cell to choose between proliferation and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Brain Chemistry/physiology , Brain/embryology , Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , src Homology Domains/physiology , Animals , Mammals , Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Shc Signaling Adaptor Proteins
14.
Trends Neurosci ; 22(8): 365-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10407422

ABSTRACT

The JAK -STAT (Janus kinase-signal transducer and activator of transcription) signalling pathway that is stimulated by cytokines has been much investigated in haematopoietic cells, but recent data indicate that this pathway is also present and active during neuronal and glial differentiation. Furthermore, it is now clear that growth factors other than the classical cytokines can act through this pathway and that physiological inhibitors of this signalling cascade exist. Thus, the JAKs, the STATs and their specific inhibitors could be molecules with important roles in the CNS.


Subject(s)
Brain/embryology , Brain/growth & development , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Transcriptional Activation/physiology , Aging/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/physiology , Embryo, Mammalian/physiology , Embryo, Nonmammalian , Embryonic and Fetal Development/physiology
15.
Trends Neurosci ; 24(3): 182-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11182459

ABSTRACT

Huntington's disease is characterized by a loss of brain striatal neurons that occurs as a consequence of an expansion of a CAG repeat in the huntingtin protein. The resulting extended polyglutamine stretch confers a deleterious gain-of-function to the protein. Analysis of the mutant protein has attracted most of the research activity in the field, however re-examination of earlier data and new results on the beneficial functions of normal huntingtin indicate that loss of the normal protein function might actually equally contribute to the pathology. Thus, complete elucidation of the physiological role(s) of huntingtin and its mode of action are essential and could lead to new therapeutic approaches.


Subject(s)
Huntington Disease/physiopathology , Mutation , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Animals , Corpus Striatum/pathology , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics
16.
J Neurosci ; 20(10): 3705-13, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10804212

ABSTRACT

Expansion of a polyglutamine sequence in the N terminus of huntingtin is the gain-of-function event that causes Huntington's disease. This mutation affects primarily the medium-size spiny neurons of the striatum. Huntingtin is expressed in many neuronal and non-neuronal cell types, implying a more general function for the wild-type protein. Here we report that wild-type huntingtin acts by protecting CNS cells from a variety of apoptotic stimuli, including serum withdrawal, death receptors, and pro-apoptotic Bcl-2 homologs. This protection may take place at the level of caspase-9 activation. The full-length protein also modulates the toxicity of the poly-Q expansion. Cells expressing full-length mutant protein are susceptible to fewer death stimuli than cells expressing truncated mutant huntingtin.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/enzymology , Nuclear Proteins/genetics , Animals , Caspase 3 , Caspase 9 , Cell Line, Transformed , Cell Survival/physiology , Cerebral Cortex/cytology , Corpus Striatum/cytology , Gene Expression Regulation, Enzymologic , In Situ Nick-End Labeling , Mutagenesis/physiology , Neurons/chemistry , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Transfection , bcl-X Protein
17.
Cell Death Differ ; 11(11): 1179-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15257302

ABSTRACT

Deficiency of the apoptosome component Apaf1 leads to accumulation of supernumerary brain cells in mouse embryos. We observed that neural precursor cells (NPCs) in Apaf1(-/-) embryos escape programmed cell death, proliferate and retain their potential to differentiate. To evaluate the circumstances of Apaf1(-/-) NPC survival and investigate their fate under neurodegenerative conditions, we established cell lines of embryonic origin (ETNA). We found that Apaf1(-/-) NPCs resist common apoptotic stimuli and neurodegenerative inducers such as amyloid-beta peptide (typical of Alzheimer's disease) and mutant G93A superoxide dismutase 1 (typical of familial amyotrophic lateral sclerosis). Similar results were obtained in Apaf1(-/-) primary cells. When death is prevented by Apaf1 deficiency, cytochrome c is released from mitochondria and rapidly degraded by the proteasome, but mitochondria remain intact. Under these conditions, neither activation by cleavage of initiator caspases nor release of alternative apoptotic inducers from mitochondria takes place. In addition, NPCs can still differentiate, as revealed by neurite outgrowth and expression of differentiation markers. Our findings imply that the mitochondrion/apoptosome pathway is the main route of proneural and neural cells to death and that its inhibition prevents them from dismantling in neurodegenerative conditions. Indeed, the ETNA cell model is ideally suited for exploring the potential of novel cell therapies for the treatment of human neurodegenerations.


Subject(s)
Apoptosis , Nerve Degeneration , Neurons/pathology , Proteins/physiology , Amyloid beta-Peptides/chemistry , Animals , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Bromodeoxyuridine/pharmacology , Caspases/metabolism , Cell Death , Cell Differentiation , Cell Proliferation , Cell Survival , Immunohistochemistry , Immunoprecipitation , Membrane Potentials , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondria/pathology , Neurodegenerative Diseases , Peptide Fragments/chemistry , Plasmids/metabolism , Time Factors , Transgenes
18.
Neuroscience ; 133(1): 105-15, 2005.
Article in English | MEDLINE | ID: mdl-15893635

ABSTRACT

Shc family of adaptor molecules has been demonstrated to play an important role during the transition from proliferating neural stem cells to postmitotic neurons. Previous studies from our group demonstrated a progressive decrease of ShcA levels occurring in coincidence with the end of embryonic neurogenesis and neuronal maturation, being ShcB and ShcC the major Shc molecules expressed in the mature brain. A growing body of evidence indicates that ShcB and ShcC are neuronal specific molecules exerting important roles in neuronal survival and phenotypic stability thus becoming potential attracting target molecules for development of drugs for interfering with brain demises. Here, we examine the expression pattern of ShcB and ShcC in neuronal populations composing the adult central and peripheral nervous system, in order to better elucidate their roles in vivo. We found a heterogeneous and peculiar presence and subcellular localization of ShcB and ShcC in specific neuronal populations, enlightening a potential specific requirement of these two molecules in the survival/maintenance of defined neuronal subtypes.


Subject(s)
Brain Chemistry/genetics , Brain Chemistry/physiology , Gene Expression Profiling , Gene Expression Regulation/physiology , Neurons/metabolism , Neuropeptides/biosynthesis , Neuropeptides/genetics , Animals , Basal Ganglia/cytology , Basal Ganglia/metabolism , Blotting, Western , Cerebellum/cytology , Cerebellum/metabolism , Eye/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Immunohistochemistry , In Situ Hybridization , Neurons/ultrastructure , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Optic Nerve/cytology , Optic Nerve/metabolism , Peripheral Nerves/metabolism , Rats , Rats, Wistar , Shc Signaling Adaptor Proteins , Spinal Cord/cytology , Spinal Cord/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3 , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , src Homology Domains
19.
Cell Death Differ ; 22(4): 690-702, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25301063

ABSTRACT

In the adult brain, neurons require local cholesterol production, which is supplied by astrocytes through apoE-containing lipoproteins. In Huntington's disease (HD), such cholesterol biosynthesis in the brain is severely reduced. Here we show that this defect, occurring in astrocytes, is detrimental for HD neurons. Astrocytes bearing the huntingtin protein containing increasing CAG repeats secreted less apoE-lipoprotein-bound cholesterol in the medium. Conditioned media from HD astrocytes and lipoprotein-depleted conditioned media from wild-type (wt) astrocytes were equally detrimental in a neurite outgrowth assay and did not support synaptic activity in HD neurons, compared with conditions of cholesterol supplementation or conditioned media from wt astrocytes. Molecular perturbation of cholesterol biosynthesis and efflux in astrocytes caused similarly altered astrocyte-neuron cross talk, whereas enhancement of glial SREBP2 and ABCA1 function reversed the aspects of neuronal dysfunction in HD. These findings indicate that astrocyte-mediated cholesterol homeostasis could be a potential therapeutic target to ameliorate neuronal dysfunction in HD.


Subject(s)
Cholesterol/biosynthesis , Huntington Disease/physiopathology , Neurons/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoproteins E/analysis , Apolipoproteins E/metabolism , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cholesterol/analysis , Cholesterol/metabolism , Cholesterol/pharmacology , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Huntingtin Protein , Lipoproteins/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Trinucleotide Repeats/genetics
20.
Hum Gene Ther ; 8(11): 1345-53, 1997 Jul 20.
Article in English | MEDLINE | ID: mdl-9295129

ABSTRACT

The growth of U-87 or C6 gliomas co-implanted in nude mice with retroviral producer cells (VPC) expressing the herpes simplex virus-thymidine kinase (HSV-tk) gene is only partially impaired by treatment with ganciclovir (GCV). The effect of GCV is even less evident when C6 and VPC are co-implanted into the rat brain. Furthermore, tumors from C6 cells carrying the HSV-tk gene are not eradicated by GCV, although they remain sensitive to GCV when replated in vitro. These limits of the HSV-tk/GCV system in glioma gene therapy may be due to insufficient gene transfer and/or insufficient delivery of GCV to glioma cells. Combination of HSV-tk and one or more cytokines may improve the antitumor efficacy. Among cytokines, interleukin-4 (IL-4) has already been shown to be active against gliomas. In nude mice, GCV treatment inhibited tumor growth more effectively after co-injection of C6 cells with a mixture of VPC transducing IL-4 and HSV-tk genes than after co-injection with either IL-4 or HSV-tk VPC only. In immunocompetent Sprague-Dawley rats, co-injection of IL-4 VPC and C6 cells was also effective in inhibiting the growth of C6 brain tumors, 38% of the animals surviving for at least 2 months. Furthermore, increased and prolonged antitumor efficacy was obtained by transducing both IL-4 and HSV-tk genes.


Subject(s)
Antimetabolites/pharmacology , Ganciclovir/pharmacology , Genetic Therapy , Glioma/therapy , Interleukin-4/genetics , Simplexvirus/enzymology , Thymidine Kinase/genetics , Viral Proteins/genetics , Animals , Female , Gene Transfer Techniques , Genetic Vectors , Interleukin-4/metabolism , Interleukin-4/therapeutic use , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Retroviridae/genetics , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL