Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neural Transm (Vienna) ; 110(5): 509-15, 2003 May.
Article in English | MEDLINE | ID: mdl-12721812

ABSTRACT

Deprenyl and other propargylamines are clinically beneficial in Parkinson's disease (PD). The benefits were thought to depend on monoamine oxidase B (MAO-B) inhibition. A large body of research has now shown that the propargylamines increase neuronal survival independently of MAO-B inhibition by interfering with apoptosis signaling pathways. The propargylamines bind to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The GAPDH binding is associated with decreased synthesis of pro-apoptotic proteins like BAX, c-JUN and GAPDH but increased synthesis of anti-apoptotic proteins like BCL-2, Cu-Zn superoxide dismutase and heat shock protein 70. Anti-apoptotic propargylamines that do not inhibit MAO-B are now in PD clinical trial.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Monoamine Oxidase/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Pargyline/analogs & derivatives , Pargyline/therapeutic use , Propylamines/therapeutic use , Selegiline/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , HSP70 Heat-Shock Proteins/metabolism , Humans , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Selegiline/therapeutic use , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein
2.
Neuroscience ; 118(3): 741-54, 2003.
Article in English | MEDLINE | ID: mdl-12710981

ABSTRACT

Previous studies have demonstrated that ovarian steroids exert neuroprotective effects in a variety of in vitro and in vivo systems. The mechanisms underlying these effects remain poorly understood. In the present study, the neuroprotective effects of estradiol (E(2)) and progesterone (P) were examined in two models of apoptosis induced by growth factor insufficiency: partially nerve growth factor (NGF)-differentiated PC12 cells, after serum and NGF withdrawal; and axotomized immature rat facial motor motoneurons. E(2) and P both increased the survival of trophically withdrawn NGF-differentiated PC12 cells, at physiologically relevant concentrations. However, neither steroid had a significant effect on the survival of PC12 cells that had not been NGF treated. Exposure to NGF had no effect on the expression of estrogen receptor (ER)beta, but markedly increased the levels of ERalpha and altered the expression of the progesterone receptor (PR) from predominantly PR-B in NGF naive cells, to predominantly PR-A after NGF. The survival promoting effects of E(2) and P were blocked by the specific steroid receptor antagonists Faslodex (ICI 182780) and onapristone (ZK98299), respectively. Inhibitors of RNA (actinomycin D) or protein (cycloheximide) synthesis also abrogated the protective effects of both steroids. In immature rats, E(2) and P both significantly increased the numbers of surviving facial motor neurons at 21 days after axotomy. These data demonstrate significant protective effects of E(2) and P in two well-characterized models of apoptosis induced by trophic withdrawal and suggest that, at least in PC12 cells, the effects of the steroids are mediated via interaction with nuclear steroid receptor systems. The lack of steroid responsiveness in NGF-naive PC12 cells despite the presence of abundant ERbeta and PR-B are consistent with the view that ERalpha and PR-A may be particularly important as mediators of the neuroprotective effects of their corresponding hormonal ligands.


Subject(s)
Apoptosis/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Facial Nerve Injuries/drug therapy , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Retrograde Degeneration/drug therapy , Animals , Apoptosis/physiology , Axotomy , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Choline O-Acetyltransferase/metabolism , Culture Media, Serum-Free/pharmacology , Drug Interactions/physiology , Estradiol/therapeutic use , Estrogen Receptor alpha , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/physiopathology , Fulvestrant , Gonanes/pharmacology , Nerve Growth Factor/deficiency , Nerve Growth Factor/pharmacology , Neuroprotective Agents/therapeutic use , PC12 Cells , Progesterone/therapeutic use , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Retrograde Degeneration/metabolism , Retrograde Degeneration/prevention & control
3.
J Pharmacol Exp Ther ; 301(2): 753-64, 2002 May.
Article in English | MEDLINE | ID: mdl-11961082

ABSTRACT

(-)-Deprenyl and structurally related propargylamines increase neuronal survival independently of monoamine oxidase B (MAO-B) inhibition, in part by decreasing apoptosis. We found that deprenyl and two other propargylamines, one of which does not inhibit monoamine oxidase B, increased survival in trophically withdrawn 6-day nerve growth factor (NGF)- and 9-day NGF-differentiated PC-12 cells but not in NGF naive or 3-day NGF-differentiated PC-12 cells. Four days of prior NGF exposure were required for the propargylamine-mediated antiapoptosis. Studies using actinomycin D, cycloheximide, and camptothecin revealed that the maintenance of both transcription and translation, particularly between 2 and 6 h after trophic withdrawal, was required for propargylamine-mediated antiapoptosis. Metabolic labeling of newly synthesized proteins for two-dimensional protein gel autoradiography and scintillation counting showed that the propargylamines either increased or reduced the levels of new synthesis or induced de novo synthesis of a number of different proteins, most notably proteins in the mitochondrial and nuclear subfractions. Western blotting for whole cell or subcellular fraction lysates showed that the timing of new protein synthesis changes or subcellular redistribution of apoptosis-related proteins induced by the propargylamines were appropriate to antiapoptosis. The apoptosis-related proteins included superoxide dismutases (SOD1 and SOD2), glutathione peroxidase, c-JUN, and glyceraldehyde-3-phosphate dehydrogenase. Most notable were the prevention of apoptotic decreases in BCL-2 levels and increases in mitochondrial BAX levels. In general, (-)-deprenyl-related propargylamines appear to reduce apoptosis by altering the levels or subcellular localization of proteins that affect mitochondrial membrane permeability, scavenge oxidative radicals, or participate in specific apoptosis signaling pathways.


Subject(s)
Apoptosis/physiology , Nerve Growth Factor/metabolism , Pargyline/analogs & derivatives , Pargyline/pharmacology , Propylamines/pharmacology , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Culture Media , Culture Media, Serum-Free , PC12 Cells , Protein Synthesis Inhibitors/pharmacology , Rats
4.
Eur J Ophthalmol ; 11 Suppl 2: S12-22, 2001.
Article in English | MEDLINE | ID: mdl-11592526

ABSTRACT

PURPOSE: Recent studies in the post-mortem human retina have suggested that apoptosis contributes to retinal ganglion cell (RGC) loss in glaucoma. If apoptosis contributes significantly to glaucomatous RGC loss, and if the specific apoptosis signalling pathways for glaucomatous apoptosis can be determined, agents that interrupt or oppose the signalling have the potential to slow the progression of glaucoma. METHODS: Recent data in animal models indicate that mitochondrially-dependent apoptosis contributes to RGC loss in glaucoma. Mitochondrially-dependent apoptosis involves proteins like BAX that increase mitochondrial membrane permeability and promote apoptosis and proteins like BCL-2 that decrease mitochondrial membrane permeability and reduce apoptosis. New protein synthesis induced by the alpha-2 agonist, brimonidine, prevents decreases in the levels of BCL-2 and thereby reduces mitochondrially-dependent apoptosis. CONCLUSIONS: Brimonidine appears to maintain BCL-2 levels by supporting the activity of an intrinsic anti-apoptosis signalling system that involves phosphorylation of protein kinase B. Phosphorylated protein kinase B appears to counteract the apoptosis signalling mechanisms which operate in glaucomatous retina.


Subject(s)
Apoptosis , Glaucoma/metabolism , Protein Serine-Threonine Kinases , Retinal Diseases/metabolism , Signal Transduction , Adrenergic alpha-Agonists/pharmacology , Animals , Apoptosis/drug effects , Brimonidine Tartrate , Glaucoma/drug therapy , Glaucoma/pathology , Humans , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinoxalines/pharmacology , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Retinal Ganglion Cells/pathology
5.
Surv Ophthalmol ; 45 Suppl 3: S277-83; discussuin S295-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11377449

ABSTRACT

Apoptosis may contribute to retinal ganglion cell loss in glaucoma and glaucoma models. Recent research has suggested that mitochondrially dependent apoptosis signaling may contribute to apoptosis in a rat model of glaucoma involving chronic increases in intraocular pressure. In some forms of apoptosis, mitochondrially dependent signaling involves increases in mitochondrial membrane permeability and the mitochondrial release of factors that signal for cell degradation. Opening of a multi-protein, mitochondrial megapore is one factor that contributes to the increased permeability and some anti-apoptotic proteins, particularly BCL-2 and BCL-X(L), bind at the megapore and facilitate megapore closure and reduce increases in mitochondrial membrane permeability. Phosphorylated protein kinase B (Akt) serves as an integrator for cellular survival signals and facilitates the megapore actions of BCL-2 and BCL-X(L), which could protect retinal ganglion cells against insults that induce apoptosis. Several anti-apoptotic agents are being evaluated for use in glaucoma, including brimonidine and propargylamines, which oppose mitochondrially dependent apoptosis through pathways involving phosphorylated Akt.


Subject(s)
Cell Membrane Permeability/drug effects , Glaucoma/drug therapy , Mitochondria/drug effects , Pargyline/pharmacology , Propylamines/pharmacology , Quinoxalines/pharmacology , Animals , Apoptosis/drug effects , Brimonidine Tartrate , Glaucoma/metabolism , Humans , Intraocular Pressure , Mitochondria/metabolism , Pargyline/analogs & derivatives , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , bcl-X Protein
6.
Invest Ophthalmol Vis Sci ; 41(11): 3451-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11006238

ABSTRACT

PURPOSE: To characterize a long-term elevated intraocular pressure (IOP) glaucoma model in the rat with respect to electroretinographic (ERG) changes and the pattern and mechanism of retinal ganglion cell (RGC) death. METHODS; An approximate doubling of IOP was induced in one eye (G) of female Wistar rats (150-180 g) by cautery of 3 episcleral/limbal veins. At intervals over 3 to 4 months, measurements of IOP and ERG changes (contact-lens electrode) were made in both the G and contralateral normal (N) eyes. At the end of 3 to 4 months of elevated IOP, RGCs were fluorescently labeled with Fluorogold (retrogradely from the superior colliculus), or retinas were labeled by intravitreal injection of a mitochondrial potential indicator dye and stained for apoptotic nuclei with a DNA dye. Flatmounts of fixed, dye-labeled retinas were examined by epifluorescence, confocal, or interference contrast microscopy. RESULTS: Elevated IOP was consistently maintained for up to 4 months in G eyes, but ERG a- and b-waves showed a statistically significant decline, of 30% to 40% in amplitude, after 3 months. Loss of RGCs in G retinas was primarily focal with no statistically significant loss demonstrable outside of the focal areas when assessed by an area sampling method for counting RGCs, which totaled 2% to 3% of the entire retinal area. Mitochondrial membrane potential of cells in the RGC layer was reduced by 17.5% (P: < 0.05) in regions surrounding areas of focal loss compared with comparable locations in control N eyes. After 3.5 months' elevated IOP the G retinas showed cell nuclei at various stages of apoptosis, from initial DNA condensation to fragmentation. CONCLUSIONS: The three-vein episcleral/limbal vein occlusion model for inducing glaucomatous pathology in the rat eye gives a consistent long-term elevation of IOP. After 3 to 4 months of approximately 100% increased IOP, the ERG responses begin to decline, there is a variable focal loss of RGCs, and some of the remaining RGCs show characteristics of stress and apoptosis. These changes seem consistent with retinal damage in human glaucoma (focal field defects), and this rat model appears to mimic some features of primary open-angle glaucoma.


Subject(s)
Glaucoma, Open-Angle/complications , Intraocular Pressure , Retinal Diseases/etiology , Retinal Ganglion Cells/pathology , Stilbamidines , Animals , Cell Death , Cell Nucleus/pathology , DNA Fragmentation , Disease Models, Animal , Electroretinography , Female , Fluorescent Dyes , Glaucoma, Open-Angle/physiopathology , Membrane Potentials/physiology , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondria/physiology , N-Methylaspartate/administration & dosage , Rats , Rats, Wistar , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Time Factors
7.
Mol Pharmacol ; 57(1): 2-12, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617673

ABSTRACT

Antisense oligonucleotides against the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) are able to reduce some forms of apoptosis. In those forms, overall GAPDH levels increase and the enzyme accumulates in the nucleus. The monoamine oxidase B (MAO-B) inhibitor, (-)-deprenyl (DEP), its metabolite (-)-desmethyldeprenyl, and a tricyclic DEP analog, CGP3466, can reduce apoptosis independently of MAO-B inhibition and have been found to bind to GAPDH. We used neuronally differentiated PC12 cells to show that DEP, DES, and CGP3466 reduce apoptosis caused by serum and nerve growth factor withdrawal over the concentration range of 10(-) to 10(-13) M. We provide evidence that the DEP-like compounds bind to GAPDH in the PC12 cells and that they prevent both the apoptotic increases in GAPDH levels and nuclear accumulation of GAPDH. In vitro, the compounds enhanced the conversion of NAD(+) to NADH by GAPDH in the presence of AUUUA-rich RNA and converted GAPDH from its usual tetrameric form to a dimeric form. Using cell lysates, we found a marked increase in rates of NAD(+) to NADH conversion in early apoptosis, which was returned toward control values by the DEP-like compounds. Accordingly, the DEP-like compounds appear to decrease glycolysis by preventing the GAPDH increases in early apoptosis. GAPDH dimer may not have the capacity to contribute to apoptosis in a similar manner to the tetramer, which might account for the antiapoptotic capacity of the compounds. These actions on GAPDH, rather than MAO-B inhibition, may contribute to the improvements in Parkinson's and Huntington's diseases found with DEP treatment.


Subject(s)
Apoptosis , Blood Proteins/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Nerve Growth Factor/physiology , Amphetamines/pharmacology , Animals , Blood Proteins/deficiency , Dimerization , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Monoamine Oxidase Inhibitors/pharmacology , Nerve Growth Factor/deficiency , Oxepins/pharmacology , PC12 Cells , Protein Conformation , Rats , Selegiline/pharmacology
8.
J Neural Transm Suppl ; (60): 77-100, 2000.
Article in English | MEDLINE | ID: mdl-11205159

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a well-studied glycolytic enzyme that plays a key role in energy metabolism. GAPDH catalyzes the conversion of glyceraldehyde-3-phosphate to 1,3-bisphosphoglycerate in the glycolytic pathway. As part of the conversion, GAPDH converts NAD+ to the high-energy electron carrier NADH. GAPDH has been referred to as a "housekeeping" protein and based on the view that GAPDH gene expression remains constant under changing cellular conditions, the levels of GAPDH mRNA have frequently been used to normalize northern blots. In recent years, that view has changed since GAPDH is now known to contribute to a number of diverse cellular functions unrelated to glycolysis. Normative functions of GAPDH now include nuclear RNA export, DNA replication, DNA repair, exocytotic membrane fusion, cytoskeletal organization and phosphotransferase activity. Pathologically, GAPDH has been implicated in apoptosis, neurodegenerative disease, prostate cancer and viral pathogenesis (see Sirover (1999) for a recent review of GAPDH functions). Most recently, it has been shown that GAPDH is a target for deprenyl related compounds (Carlile et al., 2000; Kragten et al., 1998) and may contribute to the neuroprotection offered by those compounds.


Subject(s)
Apoptosis/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Neurodegenerative Diseases/enzymology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Brain/physiopathology , Humans , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Selegiline/pharmacology , Signal Transduction/drug effects
10.
Biochem Biophys Res Commun ; 261(1): 123-30, 1999 Jul 22.
Article in English | MEDLINE | ID: mdl-10405334

ABSTRACT

There is accumulating evidence that mitochondrial membrane potential (DeltaPsi(M)) is reduced in aged cells. In addition, a decrease of DeltaPsi(M) has been shown to be an early event in many forms of apoptosis. Here we use a mitochondrial potentiometric dye with in situ laser scanning confocal microscopic (LSCM) imaging to demonstrate that DeltaPsi(M) is dramatically decreased in both the p53-overexpressing, senescent EJ tumor cells and in pre-apoptotic PC12 cells compared to controls. Treatment with cyclosporin A (CSA), which facilitates closure of the mitochondrial permeability transition pore (PTP), was able to reverse the decrease in DeltaPsi(M) in pre-apoptotic PC12 cells but not in the senescent EJ-p53 cells. The capacity to prevent dissipation of DeltaPsi(M) in response to agents that facilitate PTP closure may differentiate cells entering apoptosis from those participating in senescence. Therefore, regulation of the closure of the mitochondrial PTP in the presence of decreased DeltaPsi(M) may be a decisional checkpoint in distinguishing between growth arrest pathways.


Subject(s)
Cellular Senescence , Ion Channels/physiology , Mitochondria/physiology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Biotin/analysis , Cell Differentiation/drug effects , Cell Membrane Permeability/drug effects , Cyclosporine/pharmacology , Fluorescent Dyes , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Microscopy, Confocal , Mitochondria/drug effects , Nerve Growth Factors/pharmacology , PC12 Cells , Permeability , Rats , Rhodamines , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
11.
Neuroreport ; 10(5): 1149-53, 1999 Apr 06.
Article in English | MEDLINE | ID: mdl-10321500

ABSTRACT

Increased expression and nuclear accumulation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) are early, critical events in several forms of apoptosis. In order to investigate the subcellular trafficking of GAPDH in vivo, the localization of a GAPDH-green fluorescent protein (GFP) fusion was studied in PC12, HEK 293 and COS-1 cells. In control cells, fusion protein autofluorescence was largely restricted to the cytoplasm, rather than the nuclear concentration favored by GFP alone. In contrast, as early as 30 min after an insult, nuclear fluorescence increased in all cell lines studied. The fusion protein redistribution paralleled the dynamics of endogenous GAPDH. These data suggest that some nuclear GAPDH observed during apoptosis represents protein previously resident in the cytosol. This construct provides an in vivo monitor for an early change in apoptosis.


Subject(s)
Apoptosis/physiology , Cell Nucleus/metabolism , Indicators and Reagents/metabolism , Luminescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Biological Transport/physiology , COS Cells , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , PC12 Cells , Rats , Subcellular Fractions/metabolism , Time Factors , Tissue Distribution/physiology
12.
Biochem Biophys Res Commun ; 257(2): 440-7, 1999 Apr 13.
Article in English | MEDLINE | ID: mdl-10198232

ABSTRACT

MPP+ inhibits mitochondrial complex I and alpha-ketoglutarate dehydrogenase causing necrosis or apoptosis of catecholaminergic neurons. Low glucose levels or glycolytic blockade has been shown to potentiate MPP+ toxicity. We found that MPP+ caused concentration-dependent apoptosis of neuronally differentiated PC12 cells and that glucose, but not pyruvate, supplementation reduced apoptosis. Oligomycin concentrations sufficient to inhibit ATP synthase blocked the decreased apoptosis afforded by glucose supplementation. Laser-scanning confocal microscope imaging of chloromethyl-tetramethylrosamine methyl ester fluorescence to estimate DeltaPsiM showed that MPP+ and atractyloside reduced DeltaPsiM, while cyclosporin A (CSA) and glucose supplementation reversed decreases in DeltaPsiM caused by MPP+. Oligomycin blocked the effect of glucose supplementation on DeltaPsiM. These findings show that (i) MPP+-induced and atractyloside-induced apoptosis are associated with reduced DeltaPsiM; (ii) CSA maintains DeltaPsiM and reduces MPP+-induced apoptosis; and (iii) glucose supplementation maintains DeltaPsiM, likely by glycolytic ATP-dependent proton pumping at ATP synthase and reduces MPP+-induced apoptosis.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Apoptosis/drug effects , Glucose/pharmacology , Membrane Potentials/drug effects , Mitochondria/drug effects , Proton-Translocating ATPases/metabolism , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , Animals , Atractyloside/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cyclosporine/pharmacology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Glucose/antagonists & inhibitors , Glucose/metabolism , Glycolysis/drug effects , Microscopy, Confocal , Mitochondria/enzymology , Mitochondria/physiology , Nerve Growth Factors/pharmacology , Oligomycins/pharmacology , PC12 Cells , Proton-Translocating ATPases/antagonists & inhibitors , Pyruvic Acid/pharmacology , Rats , Time Factors
13.
Neuroreport ; 10(1): 41-6, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-10094130

ABSTRACT

Metabolic hypofunction is a common finding in a number of neurodegenerative diseases, including Alzheimer's disease (AD). The strong linkage between the amyloid precursor protein (APP) and AD led us to examine whether over-expression of this protein in CNS-type cells had an effect on mitochondria. We found abnormal morphology in mitochondria of the neuroectodermal progeny of P19 cells stably transfected with human APP751. In addition, the mitochondria of APP-transfected clones had a decreased mitochondrial membrane potential. These changes were independent of Abeta toxicity and distinct from complex I inhibition. Our results have important implications for the earliest events in the pathophysiology of AD and, by extrapolation, for intervention therapies.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Mitochondria/pathology , Neuroectodermal Tumors/pathology , Animals , Humans , Membrane Potentials/physiology , Mice , Microscopy, Electron , Mitochondria/metabolism , Neuroectodermal Tumors/metabolism , Oxidation-Reduction , Tumor Cells, Cultured
14.
Parkinsonism Relat Disord ; 5(4): 221-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-18591144

ABSTRACT

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) causes catecholaminergic nerve cell loss and a syndrome similar to Parkinson's disease (PD). The metabolite of MPTP, MPP(+) (1-methyl-4-phenylpyridinium), decreases mitochondrial complex I activity similar to that in the PD nigra. Opening of a multi-protein, mitochondrial membrane pore constitutes a critical decisional event in some forms of apoptosis. We review recent findings showing that the permeability transition pore (PTP) opening caused by a decrease in the mitochondrial membrane potential (DeltaPsi(M)) contributes to MPP(+)-induced apoptosis. The reduction in DeltaPsi(M) appears to result from decreased proton pumping at complex I and therefore decreased complex I activity may also contribute to apoptosis in PD.

15.
Ann Neurol ; 44(3 Suppl 1): S134-41, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9749585

ABSTRACT

Apoptotic cell death has been shown to constitute the terminal process in some neurodegenerative diseases, notably Alzheimer's disease and Parkinson's disease (PD). A decrease in mitochondrial membrane potential (delta psiM) causing opening of the permeability transition pore (PTP) in mitochondrial membranes has been implicated as a critical effector of apoptosis in a variety of non-neural cells. Opening of the PTP leads to the release of so-called apoptosis initiation factors that induce the degradative events of apoptosis, such as nuclear chromatin condensation and DNA fragmentation. We have extended those findings to a neuronal model of apoptosis caused by trophic withdrawal, by showing that a decrease in delta psiM is an early event occurring 2 to 6 hours before the degradative events of apoptosis. A deficiency in mitochondrial complex I activity has been demonstrated in the substantia nigra of postmortem brains and several peripheral tissues obtained from PD patients. Because delta psiM is generated by the pumping of protons out across the inner mitochondrial membrane at the mitochondrial complexes, particularly complex I, we hypothesized that the decrease in complex activity could result in a decrease in delta psiM that would render PD substantia nigra neurons vulnerable to apoptosis. In preliminary studies, we have found a decrease in delta psiM in fibroblasts obtained from some PD patients. If a decrease in delta psiM consequent on decreased complex activity is an intrinsic defect in some PD patients, it would open a number of new avenues for the reduction of neuronal apoptosis in PD. The oncoprotein BCL-2 and the scavenger protein SOD-1 have been shown to reduce apoptosis by facilitating closure of the PTP. A number of agents have been shown to maintain BCL-2 and/or SOD-1 synthesis in damaged nerve cells and thereby reduce apoptosis. Other agents, such as cyclosporin A and some benzodiazepine receptor-binding agents, have been found to act directly on the PTP to reduce apoptosis. Accordingly, agents that maintain delta psiM and PTP closure may offer new and effective means of treating neurodegenerative apoptosis.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Neurodegenerative Diseases/therapy , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Energy Metabolism/physiology , Humans , Neurodegenerative Diseases/physiopathology , Neurons/pathology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Risk Factors
16.
J Neurosci ; 18(3): 932-47, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9437015

ABSTRACT

Studies in non-neural cells have suggested that a fall in mitochondrial membrane potential (DeltaPsiM) is one of the earliest events in apoptosis. It is not known whether neural apoptosis caused by nerve growth factor (NGF) and serum withdrawal involves a decrease in DeltaPsiM. We used epifluorescence and laser confocal microscopy with the mitochondrial potentiometric dyes chloromethyl-tetramethylrosamine methyl ester and 5,5',6, 6'-tetrachloro-1,1',3,3'-tetraethybenzimidazol carbocyanine iodide to estimate DeltaPsiM. PC12 cells were differentiated in media containing serum and NGF for 6 d before withdrawal of trophic support. After washing, the cells were incubated with media containing serum and NGF (M/S+N), media without serum and NGF, or media with the "trophic-like" monoamine oxidase B inhibitor, (-)-deprenyl. Mitochondria in cells without trophic support underwent a progressive shift to lower DeltaPsiM values that was significant by 3 hr after washing. The percentages of cells with nuclear chromatin condensation or nuclear DNA fragmentation were not significantly increased above those for cells in M/S+N until 6 hr after washing. Replacement of cells into M/S+N or treatment with (-)-deprenyl markedly reduced the proportion of mitochondria with decreased DeltaPsiM. Measurements of cytoplasmic peroxyl radical levels with 2',7'-dihydrodichlorofluorescein fluorescence and intramitochondrial Ca2+ with dihydro-rhodamine-2-acetylmethyl ester indicated that cytoplasmic peroxyl radical levels were not increased until after 6 hr, whereas increases in intramitochondrial Ca2+ paralleled the decreases in DeltaPsiM. (-)-Deprenyl appeared to alter the relationship between intramitochondrial Ca2+ levels and DeltaPsiM, possibly through its reported capacity to increase the synthesis of proteins such as BCL-2.


Subject(s)
Apoptosis/physiology , Blood Proteins/pharmacology , Mitochondria/metabolism , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Selegiline/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Nucleus/drug effects , Cell Nucleus/physiology , Chromatin/physiology , Cytoplasm/metabolism , DNA Fragmentation , Free Radicals/metabolism , Image Processing, Computer-Assisted , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microscopy, Fluorescence , Mitochondria/drug effects , PC12 Cells , Peroxides/metabolism , Rats
17.
Neuroscience ; 76(4): 1121-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9027872

ABSTRACT

Central nervous system neurons and glia arise from undifferentiated embryonic neuroepithelial cells. Such progenitor cells from the human fetal forebrain can be propagated in vitro for extended periods, when grown on non-adhesive substrates in medium containing epidermal growth factor and insulin-like growth factor-1. These actively-dividing cells can be induced to differentiate into a variety of histochemically-characterized neurons and glia consistent with their forebrain origin. Electrophysiological recording indicates that differentiated neurons derived from these progenitors mature slowly, and display a range of glutamate- and GABA-mediated conductances characteristic of normal mammalian forebrain neurons. Our observations support a role for these trophic factors in normal development of the human brain. The methods described here may provide abundant normal, untransformed human forebrain neurons and glia for research and therapeutic applications.


Subject(s)
Brain/cytology , Stem Cells/physiology , Animals , Brain/embryology , Cell Differentiation/physiology , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , DNA/biosynthesis , Electrophysiology , Humans , Immunohistochemistry , Myelin Sheath/physiology , Neuroglia/physiology , Oligodendroglia/physiology , Rats , Stem Cells/metabolism
19.
J Neural Transm Suppl ; 49: 245-68, 1997.
Article in English | MEDLINE | ID: mdl-9266433

ABSTRACT

Apoptotic, rather than necrotic, nerve cell death now appears as likely to underlie a number of common neurological conditions including stroke, Alzheimer's disease, Parkinson's disease, hereditary retinal dystrophies and Amyotrophic Lateral Sclerosis. Apoptotic neuronal death is a delayed, multistep process and therefore offers a therapeutic opportunity if one or more of these steps can be interrupted or reversed. Research is beginning to show how specific macromolecules play a role in determining the apoptotic death process. We are particularly interested in the critical nature of gradual mitochondrial failure in the apoptotic process and propose that a maintenance of mitochondrial function through the pharmacological modulation of gene expression offers an opportunity for the effective treatment of some types of neurological dysfunction. Our research into the development of small diffusible molecules that reduce apoptosis has grown from studies of the irreversible MAO-B inhibitor (-)-deprenyl. (-)-Deprenyl can reduce neuronal death independently of MAO-B inhibition even after neurons have sustained seemingly lethal damage. (-)-Deprenyl can also influence the process outgrowth of some glial and neuronal populations and can reduce the concentrations of oxidative radicals in damaged cells at concentrations too small to inhibit MAO. In accord with earlier work of others, we showed that (-)-deprenyl alters the expression of a number of mRNAs or of proteins in nerve and glial cells and that the alterations in gene expression/protein synthesis are the result of a selective action on transcription. The alterations in gene expression/protein synthesis are accompanied by a decrease in DNA fragmentation characteristic of apoptosis and the death of responsive cells. The onco-proteins Bcl-2 and Bax and the scavenger proteins Cu/Zn superoxide dismutase (SOD1) and Mn superoxide dismutase (SOD-2) are among the 40-50 proteins whose synthesis is altered by (-)-deprenyl. Since mitochondrial membrane potential correlates with mitochondrial ATP production, we have used confocal laser imaging techniques in living cells to show that the transcriptional changes induced by (-)-deprenyl result in a maintenance of mitochondrial membrane potential, a decrease in intramitochondrial calcium and a decrease in cytoplasmic oxidative radical levels. We therefore propose that (-)-deprenyl acts on gene expression to maintain mitochondrial function and decrease cytoplasmic oxidative radical levels and thereby reduces apoptosis. An understanding of the molecular steps by which (-)-deprenyl selectively alters transcription may lead to the development of new therapies for neurodegenerative diseases.


Subject(s)
Apoptosis , Brain Diseases/drug therapy , Brain Diseases/pathology , Transcription, Genetic/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Brain Diseases/metabolism , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/pathology , Humans , Mitochondria/metabolism , Models, Neurological , Necrosis , Neurons/pathology , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein
20.
Neurology ; 47(6 Suppl 3): S171-83, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8959986

ABSTRACT

(-)-Deprenyl has been used to irreversibly inhibit monoamine oxidase B (MAO-B) in Parkinson's disease (PD) and Alzheimer's disease (AD) as a possible means of improving dopaminergic neurotransmission or of reducing neuronal necrosis caused by oxidative radical damage. Recent research in tissue culture and animal models has shown that (-)-deprenyl can reduce neuronal apoptosis caused by a variety of agents, in a variety of neuronal subtypes through a mechanism(s) that does not require MAO-B inhibition. Studies using general P450 blockers have shown that one of the principal metabolites of (-)-deprenyl, (-)-desmethyldeprenyl, mediates the antiapoptotic action. Other research has shown that (-)-deprenyl can induce altered expression of a number of genes in preapoptotic neurons both in vitro and in vivo, including the genes for superoxide dismutase (SOD) 1 and 2, BCL-2 and BCL-XL, nitric oxide synthase, c-JUN, and nicotinamide adenine dinucleotide dehydrogenase. Antiapoptosis by (-)-deprenyl is associated with a prevention of a progressive reduction of mitochondrial membrane potential in preapoptotic neurons, which has been shown to occur early in apoptosis and is likely an initiating factor. The above changes in gene expression appear to reduce oxidative radical damage to mitochondria and maintain mitochondrial permeability, thereby blocking mitochondrial "signals" that initiate apoptosis. In situ evidence suggests that apoptosis contributes to neuronal death in a number of neurodegenerative diseases. If apoptosis is critical to the progression of one or more human neurodegenerative diseases, then transcriptionally active agents such as (-)-desmethyldeprenyl may be of value in treating the diseases. The kinetics of (-)-deprenyl metabolism, however, and its biodistribution after oral administration, make it unlikely that the antiapoptotic action has played a major role in benefits found for the drug in PD and AD to date.


Subject(s)
Gene Expression , Monoamine Oxidase/metabolism , Nerve Degeneration , Selegiline/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...