Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Neurochem ; 116(6): 1018-27, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21175616

ABSTRACT

The subventricular zone (SVZ) is a major reservoir for stem cells in the adult mammalian brain. Neural stem cells supply the olfactory bulb with new interneurons and provide cells that migrate towards lesioned brain areas. Neuropeptide Y (NPY), one of the most abundant neuropeptides in the brain, was previously shown to induce neuroproliferation on mice SVZ cells. In the present study, performed in rats, we demonstrate the endogenous synthesis of NPY by cells in the SVZ that suggests that NPY could act as an autocrine/paracrine factor within the SVZ area. We observed that NPY promotes SVZ cell proliferation as previously reported in mice, but does not affect self-renewal of SVZ stem cells. Additionally, this study provides the first direct evidence of a chemokinetic activity of NPY on SVZ cells. Using pharmacological approaches, we demonstrate that both the mitogenic and chemokinetic properties of NPY involve Y1 receptor-mediated activation of the ERK1/2 MAP kinase pathway. Altogether, our data establish that NPY through Y1 receptors activation controls chemokinetic activity and, as for mice, is a major neuroproliferative regulator of rat SVZ cells.


Subject(s)
Cell Movement/physiology , Cerebral Ventricles/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/physiology , Neuropeptide Y/metabolism , Animals , Animals, Newborn , Arginine/analogs & derivatives , Arginine/pharmacology , Bromodeoxyuridine/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/genetics , Neuropeptide Y/pharmacology , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
2.
J Mol Cell Cardiol ; 46(5): 713-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19233193

ABSTRACT

The purpose of the present study was to investigate the potential cardioprotective effects of an original approach based on the properties of the X chromosome-linked Inhibitor of Apoptosis (XIAP), the most effective endogenous inhibitor of apoptosis. For this purpose, the C-terminal part of XIAP (BIR3 and RING domains) was fused to the protein transduction domain (PTD) of the HIV1 transactivator of transcription, which confers to fused protein the ability to cross cell membranes. This protein, so-called PTD-BIR3/RING, was administered intravenously in C57BL/6J mice subjected to 30 min coronary artery occlusion and 24 h of reperfusion. Administration of PTD-BIR3/RING at 5 min before and 30 min after the onset of reperfusion reduced infarct size vs control (23+/-2% vs 41+/-4% and 27+/-4% vs 41+/-3%, respectively, p<0.05). Similar reduction in infarct size was observed when PTD-BIR3/RING was administered prior to ischemia (28+/-1% vs 44+/-3%). In addition to inhibition of caspase-3 and -9 activities, PTD-BIR3/RING induced an inhibition of caspase-8 and several other actors of the apoptotic pathways. In conclusion, this study demonstrates that the administration of PTD-BIR3/RING reduces myocardial infarct size even when injected during reperfusion through interruption of caspase activation by pharmacologically mimicking endogenous XIAP.


Subject(s)
Molecular Mimicry , Myocardial Infarction/prevention & control , Recombinant Fusion Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Blotting, Western , Body Weight , Caspases/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Protein Structure, Tertiary , Rats , X-Linked Inhibitor of Apoptosis Protein/chemistry
3.
Proc Natl Acad Sci U S A ; 103(15): 6025-30, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16595634

ABSTRACT

Mutation in superoxide dismutase-1 (SOD1), which is a cause of ALS, alters the folding patterns of this protein. Accumulation of misfolded mutant SOD1 might activate endoplasmic reticulum (ER) stress pathways. Here we show that transgenic mice expressing ALS-linked SOD1 mutants exhibit molecular alterations indicative of a recruitment of ER's signaling machinery. We demonstrate by biochemical and morphological methods that mutant SOD1 accumulates inside the ER, where it forms insoluble high molecular weight species and interacts with the ER chaperone immunoglobulin-binding protein. These alterations are age- and region-specific, because they develop over the course of the disease and occur in the affected spinal cord but not in the nonaffected cerebellum in transgenic mutant SOD1 mice. Our results suggest a toxic mechanism for mutant SOD1 by which this ubiquitously expressed pathogenic protein could affect motor neuron survival and contribute to the selective motor neuronal degeneration in ALS.


Subject(s)
Endoplasmic Reticulum/physiology , Microsomes/enzymology , Motor Neuron Disease/genetics , Mutation , Polymorphism, Single Nucleotide , Spinal Cord/physiopathology , Superoxide Dismutase/genetics , Animals , Apoptosis , Caspase 12 , Caspases/metabolism , Endoplasmic Reticulum/pathology , Mice , Mice, Transgenic , Signal Transduction , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
4.
Neurobiol Dis ; 22(1): 177-86, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16361106

ABSTRACT

Caspases play a major role in the infarction process that follows occlusion of cerebral arteries and are important targets for stroke therapy. We have generated three fusion proteins that link various domains of the X chromosome-linked inhibitor of apoptosis (XIAP), a potent caspase inhibitor, to the protein transduction domain (PTD) of HIV-1/Tat, and have tested their efficacy after distal occlusion of the middle cerebral artery (dMCAO) in mice. PTD-XIAP failed to accumulate in brain structures after intravenous (iv) delivery, but properly transduced cortical cells when applied topically. Shorter constructs efficiently targeted the lesion after iv delivery. All proteins retained their caspase inhibitory activity and significantly reduced infarct volumes. PTD-XIAP reversed long-term impairments in the water maze test. Sequential activation of transcription factors was observed, suggesting that the effects of XIAP are mediated by both direct inhibition of apoptotic mechanisms and secondary regulation of transcription factors involved in neuronal survival.


Subject(s)
Brain Ischemia/drug therapy , Cerebral Cortex/drug effects , Recombinant Fusion Proteins/pharmacology , X-Linked Inhibitor of Apoptosis Protein/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain Ischemia/genetics , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cerebral Infarction/drug therapy , Cerebral Infarction/physiopathology , Cerebral Infarction/prevention & control , Disease Models, Animal , Gene Products, tat/genetics , Gene Products, tat/pharmacology , Gene Products, tat/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infusion Pumps , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Regulatory Elements, Transcriptional/drug effects , Regulatory Elements, Transcriptional/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/therapeutic use
5.
Proc Natl Acad Sci U S A ; 102(52): 19126-31, 2005 Dec 27.
Article in English | MEDLINE | ID: mdl-16365298

ABSTRACT

Dysfunction of mitochondrial complex I is a feature of human neurodegenerative diseases such as Leber hereditary optic neuropathy and Parkinson's disease. This mitochondrial defect is associated with a recruitment of the mitochondrial-dependent apoptotic pathway in vivo. However, in isolated brain mitochondria, complex I dysfunction caused by either pharmacological or genetic means fails to directly activate this cell death pathway. Instead, deficits of complex I stimulate intramitochondrial oxidative stress, which, in turn, increase the releasable soluble pool of cytochrome c within the mitochondrial intermembrane space. Upon mitochondrial permeabilization by the cell death agonist Bax, more cytochrome c is released to the cytosol from brain mitochondria with impaired complex I activity. Given these results, we propose a model in which defects of complex I lower the threshold for activation of mitochondrial-dependent apoptosis by Bax, thereby rendering compromised neurons more prone to degenerate. This molecular scenario may have far-reaching implications for the development of effective neuroprotective therapies for these incurable illnesses.


Subject(s)
Apoptosis Regulatory Proteins , Electron Transport Complex I/genetics , Mitochondria/pathology , Neurodegenerative Diseases/pathology , Neurons/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis , Ascorbic Acid/chemistry , Brain/metabolism , Cardiolipins/chemistry , Cardiolipins/metabolism , Cell Death , Chromatography, High Pressure Liquid , Cytochromes c/metabolism , Electron Transport Complex I/metabolism , Genetic Techniques , Hydrogen Peroxide/chemistry , Male , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Biological , Oxidative Stress , Oxygen/chemistry , Parkinson Disease/metabolism , Reactive Oxygen Species , Subcellular Fractions/metabolism , Submitochondrial Particles/pathology , Time Factors
6.
Biochem Pharmacol ; 66(8): 1643-9, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14555245

ABSTRACT

A number of studies have validated the importance of caspase activation in ischemia-induced brain damage. Caspases participate in both the initiation and execution phases of apoptosis, and play a central role in neuronal death after global cerebral ischemia. In focal ischemia, apoptosis occurs in the penumbra during the secondary phase of expansion of the lesion. However, ultrastructural and biochemical analysis have also shown signs of apoptosis in the initial lesion, or infarct core, which is traditionally considered necrotic. Specific caspase pathways are activated in the core and in the penumbra, and participate in both cytoplasmic and nuclear apoptotic events, notwithstanding their initial classification as activator or initiator caspases. This confirms previous suggestions that caspase inhibition holds tremendous neuroprotective potential in stroke and other apoptosis-related degenerative diseases. Consequently, two new approaches, aimed at treating stroke-induced brain damage by anti-apoptotic molecules, are being developed in academic and industrial laboratories. These are based, respectively, on the use of small peptide sequences corresponding to the preferred cleavage site of a caspase, and on genomic constructions derived from the fusion of endogenous anti-caspase molecules with a protein transduction domain from the human immunodeficiency virus-1. Fusion proteins containing endogenous caspases inhibitors efficiently counteract apoptosis in vitro. In in vivo models of focal cerebral ischemia, fusion proteins successfully cross the blood brain barrier and protect cells from ischemic death. This new approach by protein therapy could prove to be an interesting alternative for the reduction of the dramatic consequences of stroke, provided that the long-term efficiency of this protection in terms of functional recovery is demonstrated.


Subject(s)
Apoptosis , Brain Ischemia/pathology , Caspase Inhibitors , Brain Ischemia/complications , Brain Ischemia/enzymology , Enzyme Inhibitors/therapeutic use , Humans , Stroke/drug therapy , Stroke/etiology
7.
Mol Neurobiol ; 27(1): 33-72, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12668901

ABSTRACT

Stroke is one of the leading causes of death and severe disability in most industrialized countries. Despite the extensive research efforts of both academic and industrial laboratories during the last few decades, no changes have been brought about by the design of neuroprotective therapies. The progressive decrease of stroke-induced death and disability is entirely attributable to improvements in the identification and reduction of risk factors. Over the past few years, experimental research has led to the emergence of a wealth of information regarding the complex and interrelated processes of neuronal degeneration and death triggered by ischemia. This unprecedented insight has led to new theories on the mechanisms of ischemic damage, and has suggested new targets and strategies for therapeutic intervention designed to reduce the clinical consequences of stroke. Among current developments, three strategies seem particularly appealing namely, the limitation of initial or secondary neuronal death by inhibition of apoptotic mechanisms, the enhancement of the endogenous capacity of nervous structures to restore lost function, and the replacement of lost cells by transplantation therapy.


Subject(s)
Apoptosis/physiology , Brain Ischemia/metabolism , Stroke/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Brain Tissue Transplantation , Caspase Inhibitors , Caspases/metabolism , Humans , Necrosis , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroprotective Agents/therapeutic use , Stroke/physiopathology , Stroke/therapy
9.
Mol Cell Neurosci ; 20(4): 553-62, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12213439

ABSTRACT

Transgenic expression of mutant superoxide dismutase-1 (SOD1) produces an animal model of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. We have previously shown that the mitochondrial-dependent programmed cell death (PCD) pathway, including the redistribution of Bax, the cytosolic release of cytochrome c, and the activation of caspase-9, is recruited during neurodegeneration in spinal cords of transgenic mutant SOD1 mice. Herein, we show that the pro-PCD protein Bid is highly expressed in spinal cords of both wild-type and transgenic mutant SOD1 mice. While full-length Bid is found in the spinal cord of the two groups of mice, its cleaved form is only seen in transgenic mutant SOD1 mice, as early as the beginning of symptoms. In contrast, activated caspase-8, which is known to cleave Bid, is detected only at the end-stage of the disease. We also found that the expression of a dominant negative mutant of caspase-1 attenuates Bid cleavage as well as the mitochondrial release of cytochrome c, and the ensuing activation of caspase-9 and -3 in spinal cords of transgenic mutant SOD1 mice. These findings suggest that Bid cleavage may occur in this model by a pathway other than caspase-8 and shed light onto the molecular correlates of the previously reported beneficial effect of caspase-1 inhibition in transgenic mutant SOD1 mice.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , BH3 Interacting Domain Death Agonist Protein , Biological Transport/physiology , Carrier Proteins/chemistry , Caspase 1/genetics , Caspase 1/pharmacology , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cytochrome c Group/antagonists & inhibitors , Disease Models, Animal , Enzyme Activation/physiology , Mice , Mice, Transgenic/genetics , Mutation , Reference Values , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/pharmacology , Superoxide Dismutase-1
10.
J Biol Chem ; 277(37): 34217-22, 2002 Sep 13.
Article in English | MEDLINE | ID: mdl-12065591

ABSTRACT

Caspase-8 is the prototypic initiator of the death domain receptor pathway of apoptosis. Here, we report that caspase-8 not only triggers and amplifies the apoptotic process at cytoplasmic sites but can also act as an executioner at nuclear levels. In a murine model of acute ischemia, caspase-8 is relocated into the nucleus of apoptotic neurons, where it cleaves PARP-2, a member of the poly(ADP-ribose) polymerase family involved in DNA repair. As indicated by site-directed mutagenesis, PARP-2 cleavage occurs preferentially at the LQMD sequence mapped between the DNA binding and the catalytic domains of the protein. This is close to the cleavage sequence found in Bid, the cytoplasmic target of caspase-8. Activity assays confirm that cleavage of PARP-2 results in inactivation of its poly(ADP-ribosylation) property, proportional to the efficiency of the cleavage. Our findings add to the complexity of proteolytic caspase networks by demonstrating that caspase-8 is in turn an initiator, amplifier, and effector caspase.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Cell Nucleus/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , Animals , Biological Transport , Caspase 8 , Caspase 9 , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data
11.
J Cereb Blood Flow Metab ; 22(3): 289-98, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11891434

ABSTRACT

SUMMARY: The generally accepted concept that astrocytes are highly resistant to hypoxic/ischemic conditions has been challenged by an increasing amount of data. Considering the differences in functional implications of protoplasmic versus fibrous astrocytes, the authors have investigated the possibility that those discrepancies come from specific behaviors of the two cell types. The reactivity and fate of protoplasmic and fibrous astrocytes were observed after permanent occlusion of the medial cerebral artery in mice. A specific loss of glial fibrillary acidic protein (GFAP) immunolabeling in protoplasmic astrocytes occurred within minutes in the area with total depletion of regional CBF (rCBF) levels, whereas "classical" astrogliosis was observed in areas with remaining rCBF. Severe disturbance of cell function, as suggested by decreased GFAP content and increased permeability of the blood-brain barrier to macromolecules, was rapidly followed by necrotic cell death, as assessed by ultrastructure and by the lack of activation of the apoptotic protease caspase-3. In contrast to the response of protoplasmic astrocytes, fibrous astrocytes located at the brain surface and in deep cortical layers displayed a transient and limited hypertrophy, with no conspicuous cell death. These results point to a differential sensitivity of protoplasmic versus fibrous cortical astrocytes to blood deprivation, with a rapid demise of the former, adding to the suggestion that protoplasmic astrocytes play a crucial role in the pathogenesis of ischemic injury.


Subject(s)
Astrocytes/pathology , Cerebral Cortex/pathology , Ischemic Attack, Transient/pathology , Astrocytes/classification , Astrocytes/ultrastructure , Blood-Brain Barrier/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/ultrastructure , Cerebrovascular Circulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Kinetics , Neuroglia/pathology , Regional Blood Flow/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...