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










Publication year range
1.
Dis Model Mech ; 4(5): 634-48, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21596710

ABSTRACT

A growing body of evidence supports the 'calcium hypothesis' of Alzheimer's disease (AD), which postulates that a variety of insults might disrupt the homeostatic regulation of neuronal calcium (Ca(2+)) in the brain, resulting in the progressive symptoms that typify the disease. However, despite ongoing efforts to develop new methods for testing therapeutic compounds that might be beneficial in AD, no single bioassay permits both rapid screening and in vivo validation of candidate drugs that target specific components of the Ca(2+) regulatory machinery. To address this issue, we have integrated four distinct model systems that provide complementary information about a trial compound: the human neuroblastoma MC65 line, which provides an in vitro model of amyloid toxicity; a transgenic Drosophila model, which develops age-dependent pathologies associated with AD; the 3×TgAD transgenic mouse, which recapitulates many of the neuropathological features that typify AD; and the embryonic nervous system of Manduca, which provides a novel in vivo assay for the acute effects of amyloid peptides on neuronal motility. To demonstrate the value of this 'translational suite' of bioassays, we focused on a set of clinically approved dihydropyridines (DHPs), a class of well-defined inhibitors of L-type calcium channels that have been suggested to be neuroprotective in AD. Among the DHPs tested in this study, we found that isradipine reduced the neurotoxic consequences of ß-amyloid accumulation in all four model systems without inducing deleterious side effects. Our results provide new evidence in support of the Ca(2+) hypothesis of AD, and indicate that isradipine represents a promising drug for translation into clinical trials. In addition, these studies also demonstrate that this continuum of bioassays (representing different levels of complexity) provides an effective means of evaluating other candidate compounds that target specific components of the Ca(2+) regulatory machinery and that therefore might be beneficial in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Disease Models, Animal , Isradipine/therapeutic use , Translational Research, Biomedical , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Biological Assay , Calcium Channels, L-Type/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Drosophila/drug effects , Humans , Isradipine/administration & dosage , Isradipine/pharmacology , Manduca/drug effects , Manduca/embryology , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Neurons/ultrastructure , Protective Agents/pharmacology
2.
Mol Imaging Biol ; 12(3): 240-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19949987

ABSTRACT

PURPOSE: The purpose of this study is to detect myelin-specific T cells, key pathological mediators in early multiple sclerosis, and the corresponding animal model, experimental autoimmune encephalomyelitis (EAE), in the mouse spinal cord. PROCEDURES: T cells were labeled with the iron-based, magnetic resonance (MR) contrast reagent, Feridex, and the transfection reagent, protamine sulfate, resulting in approximately 100% iron-labeling efficiency. Feridex-labeling did not alter the induction of EAE by T cells, and recipients were imaged by a 12-T MR instrument. RESULTS: Focal hypointense lesions were resolvable to gray or white matter of the lumbar spinal cord in T(2)-weighted images of the recipients of Feridex-labeled T cells. Lesions corresponded to histological evidence of inflammatory lesions and iron-labeled cells in eight-of-eight mice. In contrast, hypointense lesions were not observed eight-of-eight recipients of unlabeled T cells. CONCLUSIONS: These results demonstrate and provide methodologies for labeling, detecting, and extracting MRI-detectable foci of iron-labeled cells.


Subject(s)
Autoimmunity/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Magnetic Resonance Imaging , Myelin Sheath/immunology , Spinal Cord/immunology , T-Lymphocytes/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/diagnosis , Iron/metabolism , Mice , Myelin Sheath/pathology , Protamines/metabolism , Spinal Cord/pathology , Staining and Labeling
3.
J Neuroimmunol ; 204(1-2): 110-7, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18722022

ABSTRACT

Xenotransplantation of rat bone marrow cells (BMC) into immunodeficient (SCID) mice generates chimeric mice susceptible to paralytic autoimmune CNS inflammation. Herein, we identified a disease relevant subset of transplantable BMC lacking expression of CD11b/c and CD49d. Moreover, disease susceptibility was enhanced in the presence of non-myelin specific T-cells. Only the CD11b/c negative population of BM retained the capability to populate the blood, spleen and spinal cord of recipients and matured after transplant to express CD11b/c. These results indicate non-myelin T cells in combination with integrin negative BM represent pre-pathogenic determinants of an enhanced disease susceptibility to myelin reactive T cells.


Subject(s)
Bone Marrow Transplantation/methods , Encephalomyelitis, Autoimmune, Experimental/surgery , Integrins/deficiency , Transplantation, Heterologous/methods , Animals , Bone Marrow Cells , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cell Transplantation/methods , Chimera , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunophenotyping/methods , Integrin alpha1/metabolism , Mice , Mice, SCID , Rats , Rats, Inbred Lew , Severity of Illness Index
4.
Nat Protoc ; 3(6): 941-7, 2008.
Article in English | MEDLINE | ID: mdl-18536642

ABSTRACT

A general protocol is described to improve the specificity for imaging superoxide formation in live cells via fluorescence microscopy with either hydroethidine (HE) or its mitochondrially targeted derivative Mito-HE (MitoSOX Red). Two different excitation wavelengths are used to distinguish the superoxide-dependent hydroxylation of Mito-HE (385-405 nm) from the nonspecific formation of ethidium (480-520 nm). Furthermore, the dual wavelength imaging in live cells can be combined with immunocolocalization, which allows superoxide formation to be compared simultaneously in cocultures of two types of genetically manipulated cells in the same microscopic field. The combination of these approaches can greatly improve the specificity for imaging superoxide formation in cultured cells and tissues.


Subject(s)
Microscopy, Fluorescence/methods , Mitochondria/metabolism , Superoxides/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Animals, Genetically Modified , Astrocytes/metabolism , Coculture Techniques , Fluorescent Dyes/metabolism , Humans , Hydroxylation , Mutation , Oxidation-Reduction , Phenanthridines/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
5.
J Neurosci ; 28(16): 4115-22, 2008 Apr 16.
Article in English | MEDLINE | ID: mdl-18417691

ABSTRACT

Mitochondrial dysfunction and oxidative stress contribute to motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Recent reports indicate that astrocytes expressing the mutations of superoxide dismutase-1 (SOD1) may contribute to motor neuron injury in ALS. Here, we provide evidence that mitochondrial dysfunction in SOD1(G93A) rat astrocytes causes astrocytes to induce apoptosis of motor neurons. Mitochondria from SOD1(G93A) rat astrocytes displayed a defective respiratory function, including decreased oxygen consumption, lack of ADP-dependent respiratory control, and decreased membrane potential. Protein 3-nitrotyrosine was detected immunochemically in mitochondrial proteins from SOD1(G93A) astrocytes, suggesting that mitochondrial defects were associated with nitroxidative damage. Furthermore, superoxide radical formation in mitochondria was increased in SOD1(G93A) astrocytes. Similar defects were found in mitochondria isolated from the spinal cord of SOD1(G93A) rats, and pretreatment of animals with the spin trap 5,5-dimethyl-1-pyrroline N-oxide restored mitochondrial function, forming adducts with mitochondrial proteins in vivo. As shown previously, SOD1(G93A) astrocytes induced death of motor neurons in cocultures, compared with nontransgenic ones. This behavior was recapitulated when nontransgenic astrocytes were treated with mitochondrial inhibitors. Remarkably, motor neuron loss was prevented by preincubation of SOD1(G93A) astrocytes with antioxidants and nitric oxide synthase inhibitors. In particular, low concentrations (approximately 10 nm) of two mitochondrial-targeted antioxidants, ubiquinone and carboxy-proxyl nitroxide, each covalently coupled to a triphenylphosphonium cation (Mito-Q and Mito-CP, respectively), prevented mitochondrial dysfunction, reduced superoxide production in SOD1(G93A) astrocytes, and restored motor neuron survival. Together, our results indicate that mitochondrial dysfunction in astrocytes critically influences motor neuron survival and support the potential pharmacological utility of mitochondrial-targeted antioxidants in ALS treatment.


Subject(s)
Antioxidants/administration & dosage , Astrocytes/enzymology , Mitochondria/enzymology , Motor Neurons/enzymology , Nerve Degeneration/enzymology , Superoxide Dismutase/genetics , Amino Acid Substitution/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Animals, Genetically Modified , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Drug Delivery Systems/methods , Mitochondria/drug effects , Mitochondria/metabolism , Motor Neurons/drug effects , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/physiology
6.
J Neurosci ; 27(29): 7777-85, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17634371

ABSTRACT

Nerve growth factor (NGF) can induce apoptosis by signaling through the p75 neurotrophin receptor (p75(NTR)) in several nerve cell populations. Cultured embryonic motor neurons expressing p75(NTR) are not vulnerable to NGF unless they are exposed to an exogenous flux of nitric oxide (*NO). In the present study, we show that p75(NTR)-mediated apoptosis in motor neurons involved neutral sphingomyelinase activation, increased mitochondrial superoxide production, and cytochrome c release to the cytosol. The mitochondria-targeted antioxidants mitoQ and mitoCP prevented neuronal loss, further evidencing the role of mitochondria in NGF-induced apoptosis. In motor neurons overexpressing the amyotrophic lateral sclerosis (ALS)-linked superoxide dismutase 1(G93A) (SOD1(G93A)) mutation, NGF induced apoptosis even in the absence of an external source of *NO. The increased susceptibility of SOD1(G93A) motor neurons to NGF was associated to decreased nuclear factor erythroid 2-related factor 2 (Nrf2) expression and downregulation of the enzymes involved in glutathione biosynthesis. In agreement, depletion of glutathione in nontransgenic motor neurons reproduced the effect of SOD1(G93A) expression, increasing their sensitivity to NGF. In contrast, rising antioxidant defenses by Nrf2 activation prevented NGF-induced apoptosis. Together, our data indicate that p75(NTR)-mediated motor neuron apoptosis involves ceramide-dependent increased mitochondrial superoxide production. This apoptotic pathway is facilitated by the expression of ALS-linked SOD1 mutations and critically modulated by Nrf2 activity.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Motor Neurons/physiology , NF-E2-Related Factor 2/metabolism , Receptor, Nerve Growth Factor/metabolism , Spinal Cord/cytology , Animals , Animals, Genetically Modified , Apoptosis/drug effects , Cells, Cultured , Cytochromes c/metabolism , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Mitochondria/drug effects , Motor Neurons/drug effects , Nerve Growth Factor/pharmacology , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/pharmacology
7.
J Biol Chem ; 282(9): 6324-37, 2007 03 02.
Article in English | MEDLINE | ID: mdl-17200124

ABSTRACT

Although peroxynitrite stimulates apoptosis in many cell types, whether peroxynitrite acts directly as an oxidant or the induction of apoptosis is because of the radicals derived from peroxynitrite decomposition remains unknown. Before undergoing apoptosis because of trophic factor deprivation, primary motor neuron cultures become immunoreactive for nitrotyrosine. We show here using tyrosine-containing peptides that free radical processes mediated by peroxynitrite decomposition products were required for triggering apoptosis in primary motor neurons and in PC12 cells cultures. The same concentrations of tyrosine-containing peptides required to prevent the nitration and apoptosis of motor neurons induced by trophic factor deprivation and of PC12 cells induced by peroxynitrite also prevented peroxynitrite-mediated nitration of motor neurons, brain homogenates, and PC12 cells. The heat shock protein 90 chaperone was nitrated in both trophic factor-deprived motor neurons and PC12 cells incubated with peroxynitrite. Tyrosine-containing peptides did not affect the induction of PC12 cell death by hydrogen peroxide. Tyrosine-containing peptides should protect by scavenging peroxynitrite-derived radicals and not by direct reactions with peroxynitrite as they neither increase the rate of peroxynitrite decomposition nor decrease the bimolecular peroxynitrite-mediated oxidation of thiols. These results reveal an important role for free radical-mediated nitration of tyrosine residues, in apoptosis induced by endogenously produced and exogenously added peroxynitrite; moreover, tyrosine-containing peptides may offer a novel strategy to neutralize the toxic effects of peroxynitrite.


Subject(s)
Apoptosis/drug effects , Motor Neurons/pathology , Peptides/pharmacology , Peroxynitrous Acid/pharmacology , Tyrosine , Animals , Free Radical Scavengers/pharmacology , Free Radicals , PC12 Cells , Rats , Tyrosine/analogs & derivatives
8.
Free Radic Biol Med ; 41(11): 1632-44, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145551

ABSTRACT

Nerve growth factor (NGF) overexpression and increased production of peroxynitrite occur in several neurodegenerative diseases. We investigated whether NGF could undergo posttranslational oxidative or nitrative modifications that would modulate its biological activity. Compared to native NGF, peroxynitrite-treated NGF showed an exceptional ability to induce p75(NTR)-dependent motor neuron apoptosis at physiologically relevant concentrations. Whereas native NGF requires an external source of nitric oxide (NO) to induce motor neuron death, peroxynitrite-treated NGF induced motor neuron apoptosis in the absence of exogenous NO. Nevertheless, NO potentiated the apoptotic activity of peroxynitrite-modified NGF. Blocking antibodies to p75(NTR) or downregulation of p75(NTR) expression by antisense treatment prevented motor neuron apoptosis induced by peroxynitrite-treated NGF. We investigated what oxidative modifications were responsible for inducing a toxic gain of function and found that peroxynitrite induced tyrosine nitration in a dose-dependent manner. Moreover, peroxynitrite triggered the formation of stable high-molecular-weight oligomers of NGF. Preventing tyrosine nitration by urate abolished the effect of peroxynitrite on NGF apoptotic activity. These results indicate that the oxidation of NGF by peroxynitrite enhances NGF apoptotic activity through p75(NTR) 10,000-fold. To our knowledge, this is the first known posttranslational modification that transforms a neurotrophin into an apoptotic agent.


Subject(s)
Apoptosis/physiology , Motor Neurons/metabolism , Nerve Growth Factor/chemistry , Peroxynitrous Acid/pharmacology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Electrophoretic Mobility Shift Assay , Mass Spectrometry , Motor Neurons/drug effects , Motor Neurons/pathology , Nerve Growth Factor/metabolism , Oligonucleotides, Antisense/pharmacology , Rats , Receptor, Nerve Growth Factor/antagonists & inhibitors , Receptor, Nerve Growth Factor/drug effects , Receptor, Nerve Growth Factor/metabolism , Tyrosine/metabolism
9.
Proc Natl Acad Sci U S A ; 103(41): 15038-43, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17015830

ABSTRACT

The putative oxidation of hydroethidine (HE) has become a widely used fluorescent assay for the detection of superoxide in cultured cells. By covalently joining HE to a hexyl triphenylphosphonium cation (Mito-HE), the HE moiety can be targeted to mitochondria. However, the specificity of HE and Mito-HE for superoxide in vivo is limited by autooxidation as well as by nonsuperoxide-dependent cellular processes that can oxidize HE probes to ethidium (Etd). Recently, superoxide was shown to react with HE to generate 2-hydroxyethidium [Zhao, H., Kalivendi, S., Zhang, H., Joseph, J., Nithipatikom, K., Vasquez-Vivar, J. & Kalyanaraman, B. (2003) Free Radic. Biol. Med. 34, 1359-1368]. However, 2-hydroxyethidium is difficult to distinguish from Etd by conventional fluorescence techniques exciting at 510 nm. While investigating the oxidation of Mito-HE by superoxide, we found that the superoxide product of both HE and Mito-HE could be selectively excited at 396 nm with minimal interference from other nonspecific oxidation products. The oxidation of Mito-HE monitored at 396 nm by antimycin-stimulated mitochondria was 30% slower than at 510 nm, indicating that superoxide production may be overestimated at 510 nm by even a traditional superoxide-stimulating mitochondrial inhibitor. The rate-limiting step for oxidation by superoxide was 4x10(6) M-1.s-1, which is proposed to involve the formation of a radical from Mito-HE. The rapid reaction with a second superoxide anion through radical-radical coupling may explain how Mito-HE and HE can compete for superoxide in vivo with intracellular superoxide dismutases. Monitoring oxidation at both 396 and 510 nm of excitation wavelengths can facilitate the more selective detection of superoxide in vivo.


Subject(s)
Ethidium , Fluorescent Dyes , Phenanthridines , Superoxides/metabolism , Animals , Animals, Newborn , Cells, Cultured , Chromatography, High Pressure Liquid , Microscopy, Confocal , Mitochondria/chemistry , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization , Superoxides/chemistry
10.
Methods Enzymol ; 396: 207-14, 2005.
Article in English | MEDLINE | ID: mdl-16291234

ABSTRACT

We report a simple method for the synthesis of peroxynitrite from nitrite and hydrogen peroxide that can generate hundreds of milliliters of 180 mM peroxynitrite within 1 h from start to finish. It can be scaled down to make small quantities of isotope-labeled peroxynitrite. The method requires only a syringe pump and tubing connectors and is feasible for any biochemical laboratory. Unreacted hydrogen peroxide is eliminated with manganese dioxide, using an improved preparation compared to commercially available manganese dioxide. A number of contaminants were detected by mass spectrometry in peroxynitrite solutions cleaned with commercially purchased manganese dioxide. Nitrite contamination of the peroxynitrite solution is less than 2% as determined using the Griess method. The residual contaminants are principally 0.28 M sodium chloride and 0.1 M sodium hydroxide, which pose few problems when peroxynitrite is diluted for use in biological experiments.


Subject(s)
Hydrogen Peroxide/chemistry , Nitrites/chemistry , Peroxynitrous Acid/chemical synthesis , Manganese Compounds/chemistry , Mass Spectrometry , Oxides/chemistry
11.
Arch Biochem Biophys ; 423(1): 213-7, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14871483

ABSTRACT

Urate is an efficient antioxidant and has recently emerged as a competitive inhibitor of tyrosine nitration by peroxynitrite. In vivo and in vitro studies demonstrate the large extent to which urate prevents nitration and establish the biological importance of the reaction between urate and peroxynitrite. The existing lack of characterization of this reaction has led us to focus our studies upon the mechanism of urate oxidation and the products formed. An oxidation product has been previously isolated and mass spectrometry revealed a mass of 146, which spontaneously fragmented into several other ion peaks without use of MS/MS mode. Here, we propose the novel oxidation product to be triuret (H(2)NCONHCONHCONH(2)). Triuret accurately reproduced the peculiar mass spectrum. Identification of the oxidation product helps to develop the mechanism of peroxynitrite-mediated oxidation of urate and can help explain urate's potential as both an antioxidant for tyrosine nitration while paradoxically acting as a pro-oxidant for lipids and sulfhydryls.


Subject(s)
Peroxynitrous Acid/metabolism , Urea/analogs & derivatives , Urea/chemistry , Uric Acid/metabolism , Mass Spectrometry , Oxidation-Reduction , Urea/isolation & purification , Urea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...