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










Publication year range
1.
Methods Mol Biol ; 1780: 497-523, 2018.
Article in English | MEDLINE | ID: mdl-29856033

ABSTRACT

Advances in molecular biology and genetics have been used to elucidate the fundamental genetic mechanisms underlying central nervous system (CNS) diseases, yet disease-modifying therapies are currently unavailable for most CNS conditions. Antisense oligonucleotides (ASOs) are synthetic single stranded chains of nucleic acids that bind to a specific sequence on ribonucleic acid (RNA) and regulate posttranscriptional gene expression. Decreased gene expression with ASOs might be able to reduce production of the disease-causing protein underlying dominantly inherited neurodegenerative disorders. Huntington's disease (HD), which is caused by a CAG repeat expansion in exon 1 of the huntingtin (HTT) gene and leads to the pathogenic expansion of a polyglutamine (PolyQ ) tract in the N terminus of the huntingtin protein (Htt), is a prime candidate for ASO therapy.State-of-the art translational science techniques can be applied to the development of an ASO targeting HTT RNA, allowing for a data-driven, stepwise progression through the drug development process. A deep and wide-ranging understanding of the basic, preclinical, clinical, and epidemiologic components of drug development will improve the likelihood of success. This includes characterizing the natural history of the disease, including evolution of biomarkers indexing the underlying pathology; using predictive preclinical models to assess the putative gain-of-function of mutant Htt protein and any loss-of-function of the wild-type protein; characterizing toxicokinetic and pharmacodynamic effects of ASOs in predictive animal models; developing sensitive and reliable biomarkers to monitor target engagement and effects on pathology that translate from animal models to patients with HD; establishing a drug delivery method that ensures reliable distribution to relevant CNS tissue; and designing clinical trials that move expeditiously from proof of concept to proof of efficacy. This review focuses on the translational science techniques that allow for efficient and informed development of an ASO for the treatment of HD.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/therapy , Oligonucleotides, Antisense/therapeutic use , Targeted Gene Repair/methods , Translational Research, Biomedical/methods , Animals , Brain/pathology , Disease Models, Animal , Drug Development , Drug Evaluation, Preclinical/methods , Humans , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Macaca fascicularis , Mice , Mutation , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , RNA Precursors/genetics , Rats , Treatment Outcome
2.
Neurobiol Aging ; 33(1): 205.e1-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20947216

ABSTRACT

Activity-dependent modulation of neuronal gene expression promotes neuronal survival and plasticity, and neuronal network activity is perturbed in aging and Alzheimer's disease (AD). Here we show that cerebral cortical neurons respond to chronic suppression of excitability by downregulating the expression of genes and their encoded proteins involved in inhibitory transmission (GABAergic and somatostatin) and Ca(2+) signaling; alterations in pathways involved in lipid metabolism and energy management are also features of silenced neuronal networks. A molecular fingerprint strikingly similar to that of diminished network activity occurs in the human brain during aging and in AD, and opposite changes occur in response to activation of N-methyl-D-aspartate (NMDA) and brain-derived neurotrophic factor (BDNF) receptors in cultured cortical neurons and in mice in response to an enriched environment or electroconvulsive shock. Our findings suggest that reduced inhibitory neurotransmission during aging and in AD may be the result of compensatory responses that, paradoxically, render the neurons vulnerable to Ca(2+)-mediated degeneration.


Subject(s)
Aging/genetics , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Gene Expression , Interneurons/physiology , Nerve Net/physiopathology , Animals , Calcium Signaling/genetics , Cell Survival/genetics , Cells, Cultured , Cerebral Cortex/cytology , Electroshock , Energy Metabolism/genetics , Environment , Humans , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , N-Methylaspartate/physiology , Neuronal Plasticity/genetics , Receptor, trkB/physiology , Synaptic Transmission
3.
Age (Dordr) ; 33(2): 155-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20842535

ABSTRACT

Adiponectin exerts multiple regulatory functions in the body and in the hypothalamus primarily through activation of its two receptors, adiponectin receptor1 and adiponectin receptor 2. Recent studies have shown that adiponectin receptors are widely expressed in other areas of the brain including the hippocampus. However, the functions of adiponectin in brain regions other than the hypothalamus are not clear. Here, we report that adiponectin can protect cultured hippocampal neurons against kainic acid-induced (KA) cytotoxicity. Adiponectin reduced the level of reactive oxygen species, attenuated apoptotic cell death, and also suppressed activation of caspase-3 induced by KA. Pretreatment of hippocampal primary neurons with an AMPK inhibitor, compound C, abolished adiponectin-induced neuronal protection. The AMPK activator, 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside, attenuated KA-induced caspase-3 activity. These findings suggest that the AMPK pathway is critically involved in adiponectin-induced neuroprotection and may mediate the antioxidative and anti-apoptotic properties of adiponectin.


Subject(s)
Adiponectin/pharmacology , Adiponectin/physiology , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cell Death/drug effects , Cells, Cultured , Enzyme Activation , Hippocampus/metabolism , Kainic Acid/toxicity , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
4.
J Alzheimers Dis ; 24(1): 15-24, 2011.
Article in English | MEDLINE | ID: mdl-21187584

ABSTRACT

In this article, we propose that impaired efficiency of glutamatergic synaptic transmission and a compensatory reduction in inhibitory neurotransmission, a process called homeostatic disinhibition, occurs in the aging brain and more dramatically in Alzheimer's disease (AD). Homeostatic disinhibition may help understand certain features of the aging brain and AD, including: 1) the increased risk for epileptic seizures, especially in the early phase of the disease; 2) the reduced ability to generate γ-oscillations; and 3) the increase in neuronal activity as measured by functional MRI. Homeostatic disinhibition may be the major mechanism that activates cognitive reserve. Modulating neuronal activity may therefore be a viable therapeutic strategy in AD that can complement existing anti-amyloid strategies. Specifically, enhancing endogenous glutamatergic synaptic transmission through increased co-agonist signaling or through positive allosteric modulation of metabotropic glutamatergic receptors appears as an attractive strategy. Alternatively, further reduction of GABAergic signaling may work as well, although care has to be taken to prevent epileptic seizures.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Homeostasis/physiology , Neural Inhibition/physiology , Aging/pathology , Alzheimer Disease/pathology , Animals , Brain/pathology , Glutamic Acid/metabolism , Humans , Synaptic Transmission/physiology
5.
Antioxid Redox Signal ; 14(7): 1261-73, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-20626318

ABSTRACT

The calcium ion (Ca(2+)) is the main second messenger that helps to transmit depolarization status and synaptic activity to the biochemical machinery of a neuron. These features make Ca(2+) regulation a critical process in neurons, which have developed extensive and intricate Ca(2+) signaling pathways. High intensity Ca(2+) signaling necessitates high ATP consumption to restore basal (low) intracellular Ca(2+) levels after Ca(2+) influx through plasma membrane receptor and voltage-dependent ion channels. Ca(2+) influx may also lead to increased generation of mitochondrial reactive oxygen species (ROS). Impaired abilities of neurons to maintain cellular energy levels and to suppress ROS may impact Ca(2+) signaling during aging and in neurodegenerative disease processes. This review focuses on mitochondrial and endoplasmic reticulum Ca(2+) homeostasis and how they relate to synaptic Ca(2+) signaling processes, neuronal energy metabolism, and ROS generation. Also, the contribution of altered Ca(2+) signaling to neurodegeneration during aging will be considered. Advances in understanding the molecular regulation of Ca(2+) homeostasis and how it is perturbed in neurological disorders may lead to therapeutic strategies that modulate neuronal Ca(2+) signaling to enhance function and counteract disease processes.


Subject(s)
Aging/metabolism , Calcium/metabolism , Neurons/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Calcium Signaling , Endoplasmic Reticulum/metabolism , Energy Metabolism , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological
6.
Nat Struct Mol Biol ; 17(6): 732-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473314

ABSTRACT

Amyloid precursor protein (APP) regulates neuronal synapse function, and its cleavage product Abeta is linked to Alzheimer's disease. Here, we present evidence that the RNA-binding proteins (RBPs) heterogeneous nuclear ribonucleoprotein (hnRNP) C and fragile X mental retardation protein (FMRP) associate with the same APP mRNA coding region element, and they influence APP translation competitively and in opposite directions. Silencing hnRNP C increased FMRP binding to APP mRNA and repressed APP translation, whereas silencing FMRP enhanced hnRNP C binding and promoted translation. Repression of APP translation was linked to colocalization of FMRP and tagged APP RNA within processing bodies; this colocalization was abrogated by hnRNP C overexpression or FMRP silencing. Our findings indicate that FMRP represses translation by recruiting APP mRNA to processing bodies, whereas hnRNP C promotes APP translation by displacing FMRP, thereby relieving the translational block.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Fragile X Mental Retardation Protein/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , 3' Untranslated Regions , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Base Sequence , Binding, Competitive , Cell Line , Cytoplasmic Structures/metabolism , Fragile X Mental Retardation Protein/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Mice , Mice, Knockout , Models, Biological , Molecular Sequence Data , Protein Biosynthesis , RNA, Small Interfering/genetics , Recombinant Proteins/genetics
7.
Neuromolecular Med ; 12(1): 1-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20232515

ABSTRACT

The generation of amyloid beta-peptide (A beta) by enzymatic cleavages of the beta-amyloid precursor protein (APP) has been at the center of Alzheimer's disease (AD) research. While the basic process of beta- and gamma-secretase-mediated generation of A beta is text book knowledge, new aspects of A beta and other cleavage products have emerged in recent years. Also our understanding of the enzymes involved in APP proteolysis has increased dramatically. All of these discoveries contribute to a more complete understanding of APP processing and the physiologic and pathologic roles of its secreted and intracellular protein products. Understanding APP processing is important for any therapeutic strategy aimed at reducing A beta levels in AD. In this review, we provide a concise description of the current state of understanding the enzymes involved in APP processing, the cleavage products generated by different processing patterns, and the potential functions of those cleavage products.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Molecular Sequence Data , Signal Transduction/physiology
8.
Neuromolecular Med ; 12(1): 44-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19885650

ABSTRACT

The amyloid beta-peptide theory of Alzheimer's Disease has helped to advance our understanding of the disease tremendously. A new area of research focuses on the changes in neuronal network activity that take place and may contribute to the clinical and pathological picture of Alzheimer's Disease. An apparent symptom of altered neuronal network activity in Alzheimer's Disease is an increased frequency in epileptic seizures that is observed both in human patients and in mouse models of Alzheimer's Disease. A root cause for altered network activity may be amyloid beta itself by its ability to alter synaptic (glutamatergic) transmission and to impair the induction of long-term potentiation. It is on this aspect of Alzheimer's Disease research that the current issue of NeuroMolecular Medicine will focus. Reviews will discuss the basic research and clinical aspects of the issue such as the effects of amyloid beta on synaptic transmission and neuronal networks, as well as the changes in functional MRI activation patterns observed in early stages of Alzheimer's Disease and the frequency and relevance of epileptic seizures in Alzheimer's Disease patients.


Subject(s)
Alzheimer Disease/physiopathology , Nerve Net/physiopathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Humans , Nerve Net/physiology , Neurons/metabolism , Neurons/pathology
9.
Neuromolecular Med ; 11(1): 28-42, 2009.
Article in English | MEDLINE | ID: mdl-19288225

ABSTRACT

Neurons require large amounts of energy to support their survival and function, and are therefore susceptible to excitotoxicity, a form of cell death involving bioenergetic stress that may occur in several neurological disorders including stroke and Alzheimer's disease. Here we studied the roles of NAD(+) bioenergetic state, and the NAD(+)-dependent enzymes SIRT1 and PARP-1, in excitotoxic neuronal death in cultured neurons and in a mouse model of focal ischemic stroke. Excitotoxic activation of NMDA receptors induced a rapid decrease of cellular NAD(P)H levels and mitochondrial membrane potential. Decreased NAD(+) levels and poly (ADP-ribose) polymer (PAR) accumulation in nuclei were relatively early events (<4 h) that preceded the appearance of propidium iodide- and TUNEL-positive cells (markers of necrotic cell death and DNA strand breakage, respectively) which became evident by 6 h. Nicotinamide, an NAD(+) precursor and an inhibitor of SIRT1 and PARP1, inhibited SIRT1 deacetylase activity without affecting SIRT1 protein levels. NAD(+) levels were preserved and PAR accumulation and neuronal death induced by excitotoxic insults were attenuated in nicotinamide-treated cells. Treatment of neurons with the SIRT1 activator resveratrol did not protect them from glutamate/NMDA-induced NAD(+) depletion and death. In a mouse model of focal cerebral ischemic stroke, NAD(+) levels were decreased in both the contralateral and ipsilateral cortex 6 h after the onset of ischemia. Stroke resulted in dynamic changes of SIRT1 protein and activity levels which varied among brain regions. Administration of nicotinamide (200 mg/kg, i.p.) up to 1 h after the onset of ischemia elevated brain NAD(+) levels and reduced ischemic infarct size. Our findings demonstrate that the NAD(+) bioenergetic state is critical in determining whether neurons live or die in excitotoxic and ischemic conditions, and suggest a potential therapeutic benefit in stroke of agents that preserve cellular NAD(+) levels. Our data further suggest that, SIRT1 is linked to bioenergetic state and stress responses in neurons, and that under conditions of reduced cellular energy levels SIRT1 enzyme activity may consume sufficient NAD(+) to nullify any cell survival-promoting effects of its deacetylase action on protein substrates.


Subject(s)
Brain Ischemia/prevention & control , NAD/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Niacinamide/metabolism , Sirtuins/metabolism , Animals , Antioxidants/metabolism , Brain Ischemia/metabolism , Cell Death/physiology , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurotoxins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism , Resveratrol , Sirtuin 1 , Stilbenes/metabolism
10.
J Neurochem ; 109(2): 644-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19226367

ABSTRACT

In order to determine the sequence of cellular processes in glutamate toxicity, we simultaneously recorded O(2) consumption, cytosolic Ca(2+) concentration ([Ca(2+)](i)), and mitochondrial membrane potential (mDeltapsi) in single cortical neurons. Oxygen consumption was measured using an amperometric self-referencing platinum electrode adjacent to neurons in which [Ca(2+)](i) and mDeltapsi were monitored with Fluo-4 and TMRE(+), respectively, using a spinning disk laser confocal microscope. Excitotoxic doses of glutamate caused an elevation of [Ca(2+)](i) followed seconds afterwards by an increase in O(2) consumption which reached a maximum level within 1-5 min. A modest increase in mDeltapsi occurred during this time period, and then, shortly before maximal O(2) consumption was reached, the mDeltapsi, as indicated by TMRE(+) fluorescence, dissipated. Maximal O(2) consumption lasted up to 5 min and then declined together with mDeltapsi and ATP levels, while [Ca(2+)](i) further increased. mDeltapsi and [Ca(2+)](i) returned to baseline levels when neurons were treated with an NMDA receptor antagonist shortly after the [Ca(2+)](i) increased. Our unprecedented spatial and time resolution revealed that this sequence of events is identical in all neurons, albeit with considerable variability in magnitude and kinetics of changes in O(2) consumption, [Ca(2+)](i), and mDeltapsi. The data obtained using this new method are consistent with a model where Ca(2+) influx causes ATP depletion, despite maximal mitochondrial respiration, minutes after glutamate receptor activation.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Glutamic Acid/toxicity , Membrane Potential, Mitochondrial/physiology , Mitochondrial Membranes/metabolism , Neurons/metabolism , Oxygen Consumption/physiology , Animals , Cations, Divalent/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Neurons/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley
11.
Exp Neurol ; 215(2): 236-42, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19013457

ABSTRACT

Brain iron insufficiency has been implicated in several neurological disorders. The dopamine system is consistently altered in studies of iron deficiency in rodent models. Changes in striatal dopamine D(2) receptors are directly proportional to the degree of iron deficiency. In light of the unknown mechanism for the iron deficiency-dopamine connection and because of the known interplay between adenosinergic and dopaminergic systems in the striatum we examined the effects of iron deficiency on the adenosine system. We first attempted to assess whether there is a functional change in the levels of adenosine receptors in response to this low iron. Mice made iron-deficient by diet had an increase in the density of striatal adenosine A(2A) (A(2A)R) but not A(1) receptor (A(1)R) compared to mice on a normal diet. Between two inbred murine strains, which had 2-fold differences in their striatal iron concentrations under normal dietary conditions, the strain with the lower striatal iron had the highest striatal A(2A)R density. Treatment of SH-SY5Y (human neuroblastoma) cells with an iron chelator resulted in increased density of A(2A)R. In these cells, A(2A)R agonist-induced cyclic AMP production was enhanced in response to iron chelation, also demonstrating a functional upregulation of A(2A)R. A significant correlation (r(2)=0.79) was found between a primary marker of cellular iron status (transferrin receptor (TfR)) and A(2A)R protein density. In conclusion, the A(2A)R is increased across different iron-insufficient conditions. The relation between A(2A)R and cellular iron status may be an important pathway by which adenosine may alter the function of the dopaminergic system.


Subject(s)
Corpus Striatum/metabolism , Iron Deficiencies , Neuroblastoma/metabolism , Receptors, Adenosine A2/metabolism , Up-Regulation/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Iron, Dietary/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenethylamines/pharmacology , Receptors, Adenosine A2/genetics , Receptors, Transferrin/agonists , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Time Factors , Up-Regulation/drug effects
12.
Neuron ; 60(5): 748-66, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19081372

ABSTRACT

Mitochondrial electron transport generates the ATP that is essential for the excitability and survival of neurons, and the protein phosphorylation reactions that mediate synaptic signaling and related long-term changes in neuronal structure and function. Mitochondria are highly dynamic organelles that divide, fuse, and move purposefully within axons and dendrites. Major functions of mitochondria in neurons include the regulation of Ca(2+) and redox signaling, developmental and synaptic plasticity, and the arbitration of cell survival and death. The importance of mitochondria in neurons is evident in the neurological phenotypes in rare diseases caused by mutations in mitochondrial genes. Mitochondria-mediated oxidative stress, perturbed Ca(2+) homeostasis, and apoptosis may also contribute to the pathogenesis of prominent neurological diseases including Alzheimer's, Parkinson's, and Huntington's diseases; stroke; amyotrophic lateral sclerosis; and psychiatric disorders. Advances in understanding the molecular and cell biology of mitochondria are leading to novel approaches for the prevention and treatment of neurological disorders.


Subject(s)
Mitochondria/physiology , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Neuronal Plasticity/physiology , Animals , Electron Transport Complex I/metabolism , Humans , Mitochondrial Proteins/metabolism , Models, Biological
13.
Nucleic Acids Res ; 36(15): 5111-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18682529

ABSTRACT

XRCC1 is a critical scaffold protein that orchestrates efficient single-strand break repair (SSBR). Recent data has found an association of XRCC1 with proteins causally linked to human spinocerebellar ataxias-aprataxin and tyrosyl-DNA phosphodiesterase 1-implicating SSBR in protection against neuronal cell loss and neurodegenerative disease. We demonstrate herein that shRNA lentiviral-mediated XRCC1 knockdown in human SH-SY5Y neuroblastoma cells results in a largely selective increase in sensitivity of the nondividing (i.e. terminally differentiated) cell population to the redox-cycling agents, menadione and paraquat; this reduced survival was accompanied by an accumulation of DNA strand breaks. Using hypoxanthine-xanthine oxidase as the oxidizing method, XRCC1 deficiency affected both dividing and nondividing SH-SY5Y cells, with a greater effect on survival seen in the former case, suggesting that the spectrum of oxidative DNA damage created dictates the specific contribution of XRCC1 to cellular resistance. Primary XRCC1 heterozygous mouse cerebellar granule cells exhibit increased strand break accumulation and reduced survival due to increased apoptosis following menadione treatment. Moreover, knockdown of XRCC1 in primary human fetal brain neurons leads to enhanced sensitivity to menadione, as indicated by increased levels of DNA strand breaks relative to control cells. The cumulative results implicate XRCC1, and more broadly SSBR, in the protection of nondividing neuronal cells from the genotoxic consequences of oxidative stress.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , DNA-Binding Proteins/physiology , Neurons/metabolism , Oxidative Stress , Animals , Brain/cytology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival , DNA-Binding Proteins/antagonists & inhibitors , Humans , Mice , Neurons/cytology , X-ray Repair Cross Complementing Protein 1
14.
Ann N Y Acad Sci ; 1123: 197-212, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18375592

ABSTRACT

The mitochondrial permeability transition (MPT) pore complex is a key participant in the machinery that controls mitochondrial fate and, consequently, cell fate. The quest for the pore identity has been ongoing for several decades and yet the main structure remains unknown. Established "dogma" proposes that the core of the MPT pore is composed of an association of voltage-dependent anion channel (VDAC) and adenine nucleotide translocase (ANT). Recent genetic knockout experiments contradict this commonly accepted interpretation and provide a basis for substantial revision of the MPT pore identity. There is now sufficient evidence to exclude VDAC and ANT as the main pore structural components. Regarding MPT pore regulation, the role of cyclophilin D is confirmed and ANT may still serve some regulatory function, although the involvement of hexokinase II and creatine kinase remains unresolved. When cell protection signaling pathways are activated, we have found that the Bcl-2 family members relay the signal from glycogen synthase kinase-3 beta onto a target at or in close proximity to the pore. Our experimental findings in intact cardiac myocytes and neurons indicate that the current "dogma" related to the role of Ca2+ in MPT induction requires reevaluation. Emerging evidence suggests that after injury-producing stresses, reactive oxygen species (but not Ca2+) are largely responsible for the pore induction. In this article we discuss the current state of knowledge and provide new data related to the MPT pore structure and regulation.


Subject(s)
Heart/physiology , Mitochondrial Membrane Transport Proteins/physiology , Animals , Apoptosis , Peptidyl-Prolyl Isomerase F , Cyclophilins/physiology , Humans , Intracellular Membranes/physiology , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membranes/physiology , Mitochondrial Permeability Transition Pore , Models, Biological , Models, Cardiovascular , Permeability , Proto-Oncogene Proteins c-bcl-2/physiology
15.
J Neurochem ; 102(4): 1232-41, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17542809

ABSTRACT

Ammonium trichloro(dioxoethylene-0,0') tellurate (AS101) is a non-toxic organotellurium compound with pleiotropic activities. It was recently shown to induce production of the neurotrophic factor glial cell line-derived neurotrophic factor and to rescue neuronal-like PC-12 cells from neurotrophic factor deprivation-induced apoptosis. In this study, we show that AS101 improves functional outcome and reduces brain damage in a mouse model of focal ischemic stroke. Both pre-stroke and post-stroke intraperitoneal treatments with AS101 reduced infarct size and edema and improved the neurological function of the animals. AS101 treatments reduced both apoptotic and inflammatory caspase activities, and also inhibited protein tyrosine nitration suggesting that AS101 suppresses oxidative stress. Studies of cultured neurons showed that AS101 confers protection against apoptosis induced by either glucose deprivation or the lipid peroxidation product 4-hydroxynonenal. Moreover, AS101 treatment reduced glutamate-induced intracellular calcium elevation, a major contributor to neuronal death in stroke. As AS101 has an excellent safety profile in humans, our pre-clinical data suggest a potential therapeutic benefit of AS101 in patients suffering from stroke and other neurodegenerative conditions.


Subject(s)
Ethylenes/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose/deficiency , Hypoxia/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Rats , Time Factors
16.
Brain Pathol ; 16(4): 266-72, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17107595

ABSTRACT

UNLABELLED: Autosomal dominant Emery-Dreifuss muscular dystrophy is caused by mutations in the LMNA gene that code for the nuclear membrane protein lamin A/C. We investigated skeletal muscle fibers from several muscles for cytoplasmic degenerative changes in three patients with genetically confirmed Emery-Dreifuss muscular dystrophy. Methods included quantitative light and electron microscopy and PCR-based mutational analysis. RESULTS: The degenerative pathway was characterized by the gradual replacement of individual myofibers by connective tissue. Early stages of degeneration typically involved only a segment of the cross-sectional area of a myofiber. Intermediate stages consisted of myofiber shrinkage due to "shedding" of peripheral cytoplasmic portions into the endomysial space, and fragmentation of the myofibers by interposed collagen fibrils. Empty basement membrane sheaths surrounded by abundant deposits of extracellular matrix marked the end stage of the degenerative process. The nuclear number-to-cytoplasmic area in myofibers of one patient increased with increasing cross-sectional area, suggesting that satellite cell fusion with myofibers may have compensated for myofiber shrinkage. The pattern of degeneration described herein differs from muscular dystrophies with plasma membrane defects (dystrophinopathy, dysferlinopathy) and explains the frequently found absence of highly elevated serum creatine kinase levels in autosomal dominant Emery-Dreifuss muscular dystrophy.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Emery-Dreifuss/pathology , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Adult , Child , Child, Preschool , Creatine Kinase/blood , DNA Mutational Analysis , Humans , Lamin Type A/genetics , Male , Microscopy, Electron, Transmission , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation
17.
Mov Disord ; 21(10): 1667-73, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16941469

ABSTRACT

In spinocerebellar ataxias (SCAs), up to 30% of patients complain of restless legs syndrome (RLS). In primary RLS, a putative role of the dopaminergic system has been postulated. To assess dopaminergic function in SCA1, 2, and 3, dopamine D(2) receptor binding potential (BP) was assessed by [(11)C]raclopride positron emission tomography in 10 SCA patients, 4 of whom suffered from RLS as demonstrated by polysomnography. BP was compared to 9 age-matched control subjects. In 2 SCA patients, striatal BP was clearly reduced (<2 SD below the mean of controls). However, there were no significant group differences between SCA and controls, largely owing to a significantly higher variance of striatal BP in SCA. BP was negatively correlated with disease duration. The fit suggests an increased BP in early stages, followed by a moderate decline in all quantified regions (caudate, dorsal putamen, ventral striatum) presumably reflecting a progressive loss of D(2) receptors. RLS in SCA was not accompanied by a significant reduction of D(2) receptor availability in the striatum. This missing correlation may point to an extrastriatal origin of RLS.


Subject(s)
Carbon Radioisotopes , Corpus Striatum/diagnostic imaging , Machado-Joseph Disease/diagnostic imaging , Positron-Emission Tomography , Raclopride , Receptors, Dopamine D2/physiology , Restless Legs Syndrome/diagnostic imaging , Spinocerebellar Ataxias/diagnostic imaging , Adult , Ataxin-1 , Ataxin-3 , Ataxins , Corpus Striatum/physiopathology , Female , Genotype , Humans , Machado-Joseph Disease/genetics , Machado-Joseph Disease/physiopathology , Male , Middle Aged , Nerve Tissue Proteins/genetics , Neurologic Examination , Nuclear Proteins/genetics , Occipital Lobe/diagnostic imaging , Occipital Lobe/physiopathology , Polysomnography , Repressor Proteins/genetics , Restless Legs Syndrome/physiopathology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology
18.
Ageing Res Rev ; 5(2): 165-78, 2006 May.
Article in English | MEDLINE | ID: mdl-16682262

ABSTRACT

Throughout life, organisms and their cells are subjected to various stressors which they must respond to adaptively in order to avoid disease and death. Accordingly, cells possess a variety of stress-responsive signaling pathways that are coupled to kinase cascades and transcription factors that induce the expression of genes that encode cytoprotective proteins such as protein chaperones (PC), growth factors and antioxidant enzymes. Emerging findings suggest that many of the environmental factors that improve health and so prolong lifespan (for example, dietary restriction, exercise and cognitive stimulation) exert their beneficial effects through a hormesis-like mechanism. Here we describe data supporting the hormesis hypothesis of disease resistance and longevity, with a focus on findings from studies of the nervous system in this laboratory.


Subject(s)
Adaptation, Physiological/physiology , Aging/metabolism , Nervous System/metabolism , Aging/physiology , Animals , Caloric Restriction , Free Radicals/metabolism , Humans , Models, Biological , Nervous System/physiopathology , Reactive Oxygen Species/metabolism
19.
Nat Med ; 12(6): 621-3, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16680150

ABSTRACT

Mice transgenic for antisense Notch and normal mice treated with inhibitors of the Notch-activating enzyme gamma-secretase showed reduced damage to brain cells and improved functional outcome in a model of focal ischemic stroke. Notch endangers neurons by modulating pathways that increase their vulnerability to apoptosis, and by activating microglial cells and stimulating the infiltration of proinflammatory leukocytes. These findings suggest that Notch signaling may be a therapeutic target for treatment of stroke and related neurodegenerative conditions.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Endopeptidases/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Stroke/pathology , Amyloid Precursor Protein Secretases , Animals , Apoptosis , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain/cytology , Brain/metabolism , Brain Ischemia/metabolism , Brain Ischemia/therapy , Cells, Cultured , Endopeptidases/genetics , Enzyme Inhibitors/metabolism , Humans , Leukocytes/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Neurons/cytology , Neurons/metabolism , Peptides/genetics , Peptides/metabolism , Rats , Receptor, Notch1/genetics , Reperfusion Injury , Stroke/metabolism , Stroke/therapy , Treatment Outcome
20.
Mol Interv ; 5(5): 278-81, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16249523

ABSTRACT

Par-4, discovered in a screen for genes whose expression is increased in prostate tumor cells undergoing apoptosis, participates in physiological and pathological nerve cell death. A new study, however, provides evidence for an unexpected role for Par-4 in regulating synaptic transmission in the brain: Par-4 binds to the D2 dopamine receptor (D2DR) and modulates its activity. Mice in which the function of Par-4 is disrupted exhibit impaired dopaminergic neurotransmission, resulting in a depression-like syndrome. Several other cell death-related proteins also appear to function in regulating synaptic plasticity, suggesting that a better understanding of the functions of these proteins may lead to novel therapeutic approaches for a psychiatric and neurodegenerative disorders.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Dopamine , Neuronal Plasticity , Synapses/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Humans , Receptors, Dopamine D2/agonists , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...