Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Science ; 342(6156): 373-7, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24136970

ABSTRACT

The conservation of sleep across all animal species suggests that sleep serves a vital function. We here report that sleep has a critical function in ensuring metabolic homeostasis. Using real-time assessments of tetramethylammonium diffusion and two-photon imaging in live mice, we show that natural sleep or anesthesia are associated with a 60% increase in the interstitial space, resulting in a striking increase in convective exchange of cerebrospinal fluid with interstitial fluid. In turn, convective fluxes of interstitial fluid increased the rate of ß-amyloid clearance during sleep. Thus, the restorative function of sleep may be a consequence of the enhanced removal of potentially neurotoxic waste products that accumulate in the awake central nervous system.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Neurodegenerative Diseases/metabolism , Sleep/physiology , Adrenergic Antagonists/administration & dosage , Animals , Brain/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Cerebrospinal Fluid/metabolism , Diffusion , Electroencephalography , Extracellular Space , Intracellular Space , Male , Mice , Mice, Inbred C57BL , Quaternary Ammonium Compounds/chemistry , Receptors, Adrenergic/metabolism , Wakefulness/physiology
3.
Sci Rep ; 3: 2582, 2013.
Article in English | MEDLINE | ID: mdl-24002448

ABSTRACT

In the brain, a paravascular space exists between vascular cells and astroglial end-foot processes, creating a continuous sheath surrounding blood vessels. Using in vivo two-photon imaging we demonstrate that the paravascular circulation facilitates selective transport of small lipophilic molecules, rapid interstitial fluid movement and widespread glial calcium signaling. Depressurizing the paravascular system leads to unselective lipid diffusion, intracellular lipid accumulation and pathological signaling in astrocytes. As the central nervous system is devoid of lymphatic vessels, the paravascular space may serve as a lymphatic equivalent that represents a separate highway for the transport of lipids and signaling molecules.


Subject(s)
Astrocytes/metabolism , Brain/blood supply , Brain/metabolism , Lipid Metabolism , Microcirculation , Signal Transduction , Animals , Biological Transport , Calcium/metabolism , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Molecular Imaging/methods
4.
J Transl Med ; 11: 107, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23635358

ABSTRACT

BACKGROUND: Neurodegenerative diseases such as Alzheimer's are associated with the aggregation of endogenous peptides and proteins that contribute to neuronal dysfunction and loss. The glymphatic system, a brain-wide perivascular pathway along which cerebrospinal fluid (CSF) and interstitial fluid (ISF) rapidly exchange, has recently been identified as a key contributor to the clearance of interstitial solutes from the brain, including amyloid ß. These findings suggest that measuring changes in glymphatic pathway function may be an important prognostic for evaluating neurodegenerative disease susceptibility or progression. However, no clinically acceptable approach to evaluate glymphatic pathway function in humans has yet been developed. METHODS: Time-sequenced ex vivo fluorescence imaging of coronal rat and mouse brain slices was performed at 30-180 min following intrathecal infusion of CSF tracer (Texas Red- dextran-3, MW 3 kD; FITC- dextran-500, MW 500 kD) into the cisterna magna or lumbar spine. Tracer influx into different brain regions (cortex, white matter, subcortical structures, and hippocampus) in rat was quantified to map the movement of CSF tracer following infusion along both routes, and to determine whether glymphatic pathway function could be evaluated after lumbar intrathecal infusion. RESULTS: Following lumbar intrathecal infusions, small molecular weight TR-d3 entered the brain along perivascular pathways and exchanged broadly with the brain ISF, consistent with the initial characterization of the glymphatic pathway in mice. Large molecular weight FITC-d500 remained confined to the perivascular spaces. Lumbar intrathecal infusions exhibited a reduced and delayed peak parenchymal fluorescence intensity compared to intracisternal infusions. CONCLUSION: Lumbar intrathecal contrast delivery is a clinically useful approach that could be used in conjunction with dynamic contrast enhanced MRI nuclear imaging to assess glymphatic pathway function in humans.


Subject(s)
Brain/blood supply , Cerebrospinal Fluid/metabolism , Injections, Spinal , Molecular Probes , Animals , Extracellular Fluid/metabolism , Female , Intracranial Pressure , Lumbar Vertebrae/metabolism , Male , Mice, Inbred C57BL , Molecular Weight , Rats, Sprague-Dawley
5.
J Clin Invest ; 122(4): 1377-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22406537

ABSTRACT

In Alzheimer disease (AD), amyloid ß peptide (Aß) accumulates in plaques in the brain. Receptor for advanced glycation end products (RAGE) mediates Aß-induced perturbations in cerebral vessels, neurons, and microglia in AD. Here, we identified a high-affinity RAGE-specific inhibitor (FPS-ZM1) that blocked Aß binding to the V domain of RAGE and inhibited Aß40- and Aß42-induced cellular stress in RAGE-expressing cells in vitro and in the mouse brain in vivo. FPS-ZM1 was nontoxic to mice and readily crossed the blood-brain barrier (BBB). In aged APPsw/0 mice overexpressing human Aß-precursor protein, a transgenic mouse model of AD with established Aß pathology, FPS-ZM1 inhibited RAGE-mediated influx of circulating Aß40 and Aß42 into the brain. In brain, FPS-ZM1 bound exclusively to RAGE, which inhibited ß-secretase activity and Aß production and suppressed microglia activation and the neuroinflammatory response. Blockade of RAGE actions at the BBB and in the brain reduced Aß40 and Aß42 levels in brain markedly and normalized cognitive performance and cerebral blood flow responses in aged APPsw/0 mice. Our data suggest that FPS-ZM1 is a potent multimodal RAGE blocker that effectively controls progression of Aß-mediated brain disorder and that it may have the potential to be a disease-modifying agent for AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Benzamides/therapeutic use , Disease Models, Animal , Neuroprotective Agents/therapeutic use , Peptide Fragments/metabolism , Receptors, Immunologic/antagonists & inhibitors , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Animals , Benzamides/pharmacology , Benzamides/toxicity , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , CHO Cells/drug effects , Cerebrovascular Circulation/drug effects , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Humans , Male , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Peptide Fragments/genetics , Psychomotor Performance/drug effects , Receptor for Advanced Glycation End Products , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries
6.
Neurosurgery ; 66(1): 165-71; discussion 171-2, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023547

ABSTRACT

OBJECTIVE: Activated protein C (APC) is neuroprotective in stroke models and promotes postischemic neovascularization and neurogenesis. We used a controlled cortical impact (CCI) in mice to determine the effects of APC on neuroprotection and angiogenesis and neurogenesis after traumatic brain injury (TBI). METHODS: Mice were given (1) single-dose APC (0.8 mg/kg intraperitoneally) 15 minutes after injury, (2) multidose APC (0.8 mg/kg intraperitoneally) 15 minutes and 6 to 48 hours after injury, or (3) vehicle. We then assessed the effects of APC on posttraumatic motor function with the rotarod and wire grip and beam balance tasks, and we determined the lesion volumes and studied the formation of new blood vessels and markers of neurogenesis. RESULTS: Mice treated with single-dose or multidose APC, compared with vehicle, showed significantly improved motor function on all tests. In the single-dose and multidose APC treatment groups, at 7 days after treatment, lesion volume was significantly decreased by 30% and 50%, respectively. Multidose APC, but not single-dose APC, increased new blood vessel formation as shown by CD105(+)/Ki-67(+) double immunostaining by nearly 2-fold at 7 days. Multidose APC also promoted posttraumatic proliferation of neuroblasts in the subventricular zone (SVZ) and their migration from the SVZ to the perilesional area. CONCLUSION: Activated protein C improves functional outcome and is neuroprotective after TBI. It also promotes angiogenesis and survival and migration of neuroblasts from the SVZ to the perilesional area, but the exact role of these brain repair mechanisms remains to be determined. The present findings suggest that APC therapy may hold a significant therapeutic potential for TBI.


Subject(s)
Brain Injuries , Cerebral Cortex/pathology , Neovascularization, Physiologic/drug effects , Neurogenesis/drug effects , Neuroprotective Agents , Protein C , Analysis of Variance , Animals , Brain Injuries/drug therapy , Brain Injuries/pathology , Brain Injuries/physiopathology , Bromodeoxyuridine/metabolism , Cell Count/methods , Cerebral Cortex/drug effects , Disease Models, Animal , Doublecortin Domain Proteins , Enzyme Activation/drug effects , Enzyme Activation/physiology , Hand Strength/physiology , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Neuropeptides/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein C/metabolism , Protein C/pharmacology , Protein C/therapeutic use , Psychomotor Performance/drug effects , Rotarod Performance Test , Vascular Cell Adhesion Molecule-1/metabolism
7.
J Clin Invest ; 119(11): 3437-49, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19841542

ABSTRACT

Activated protein C (APC) is a signaling protease with anticoagulant activity. Here, we have used mice expressing a mutation in superoxide dismutase-1 (SOD1) that is linked to amyotrophic lateral sclerosis (ALS) to show that administration of APC or APC analogs with reduced anticoagulant activity after disease onset slows disease progression and extends survival. A proteolytically inactive form of APC with reduced anticoagulant activity provided no benefit. APC crossed the blood-spinal cord barrier in mice via endothelial protein C receptor. When administered after disease onset, APC eliminated leakage of hemoglobin-derived products across the blood-spinal cord barrier and delayed microglial activation. In microvessels, motor neurons, and microglial cells from SOD1-mutant mice and in cultured neuronal cells, APC transcriptionally downregulated SOD1. Inhibition of SOD1 synthesis in neuronal cells by APC required protease-activated receptor-1 (PAR1) and PAR3, which inhibited nuclear transport of the Sp1 transcription factor. Diminished mutant SOD1 synthesis by selective gene excision within endothelial cells did not alter disease progression, which suggests that diminished mutant SOD1 synthesis in other cells, including motor neurons and microglia, caused the APC-mediated slowing of disease. The delayed disease progression in mice after APC administration suggests that this approach may be of benefit to patients with familial, and possibly sporadic, ALS.


Subject(s)
Fibrinolytic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Microglia/enzymology , Motor Neurons/enzymology , Protein C/pharmacology , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Blood-Brain Barrier/metabolism , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Endothelium/metabolism , Fibrinolytic Agents/therapeutic use , Male , Mice , Microglia/cytology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein C/therapeutic use , Receptors, Cell Surface/metabolism , Receptors, Proteinase-Activated/metabolism , Sp1 Transcription Factor/metabolism , Spinal Cord/blood supply , Spinal Cord/enzymology , Superoxide Dismutase/genetics
8.
Stroke ; 40(5): 1864-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19057019

ABSTRACT

BACKGROUND AND PURPOSE: Activated protein C (APC), a protease with anticoagulant and cytoprotective activities, protects neurons and endothelium from ischemic injury. Drotrecogin-alfa activated, a hyperanticoagulant form of human recombinant APC, is currently being studied in patients with ischemic stroke. How changes in APC anticoagulant activity influence APC's neuroprotection and risk for bleeding is not clear. METHODS: We used neuronal and brain endothelial cell injury models and middle cerebral artery occlusion in mice to compare efficacy and safety of drotrecogin-alfa activated and human 3K3A-APC, an APC nonanticoagulant mutant. RESULTS: Drotrecogin-alfa activated and 3K3A-APC exhibited 148% and 10% of plasma-derived APC's anticoagulant activity and differ in the carbohydrate content. 3K3A-APC protected mouse neurons from N-methyl-d-aspartate-induced apoptosis and human brain endothelial cell from oxygen-glucose deprivation with 1.8- and 3.1-fold greater efficacy than drotrecogin-alfa activated. Given 5 minutes before transient middle cerebral artery occlusion, 3K3A-APC and drotrecogin-alfa activated (0.5 and 2 mg/kg intravenously) reduced comparably and dose-dependently the infarction lesion up to 85%. 3K3A-APC, but not drotrecogin-alfa activated, improved neurological score dose-dependently (P<0.05). 3K3A-APC did not cause bleeding. In contrast, drotrecogin-alfa activated dose-dependently increased hemoglobin content in postischemic brain. After permanent middle cerebral artery occlusion, 3K3A-APC multidose therapy (1 mg/kg intravenously at 12 hours and 1, 3, 5, and 7 days) improved functional recovery and reduced infarction by 60% with no risk for bleeding, whereas drotrecogin-alfa activated increased hemoglobin deposition in the postischemic brain and showed relatively modest neuroprotection. CONCLUSIONS: Nonanticoagulant 3K3A-APC exhibits greater neuroprotective efficacy with no risk for bleeding compared with drotrecogin-alfa activated, a hyperanticoagulant form of APC.


Subject(s)
Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/epidemiology , Fibrinolytic Agents/pharmacology , Neuroprotective Agents , Protein C/genetics , Protein C/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Excitatory Amino Acid Agonists/pharmacology , Female , Humans , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Mutation/genetics , Mutation/physiology , N-Acetylneuraminic Acid/metabolism , N-Methylaspartate/pharmacology , Neurons/drug effects , Partial Thromboplastin Time , Polysaccharides/metabolism , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Risk
9.
J Neurosci ; 28(48): 12788-97, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036971

ABSTRACT

Activated protein C (APC) is a serine protease with anticoagulant and direct cytoprotective activities. Early postischemic APC application activates the cellular protein C pathway in brain endothelium and neurons, which is neuroprotective. Whether late APC administration after a transient ischemic attack is neuroprotective and whether APC influences brain repair is not known. Here, we determined safety and efficacy of late APC and tissue-plasminogen activator (tPA) administrations in a mouse model of transient brain ischemia. tPA given at 6 h after onset of ischemia killed all mice within 2 d, whereas APC given at 6 or 24 h after ischemia onset improved significantly functional outcome and reduced spread of the ischemic lesion. At 7 d postischemia, APC multiple dosing (0.8 mg/kg, i.p.) at 6-72 or 72-144 h enhanced comparably cerebral perfusion in the ischemic border by approximately 40% as shown by in vivo lectin-FITC angiography, blocked blood-brain barrier leakage of serum proteins, and increased the number of endothelial replicating cells by 4.5- to 4.7-fold. APC multidosing at 6-72 h or 72-144 h increased proliferation of neuronal progenitor cells in the subventricular zone (SVZ) by 40-50% and migration of newly formed neuroblasts from the SVZ toward the ischemic border by approximately twofold. The effects of APC on neovascularization and neurogenesis were mediated by protease-activated receptor 1 and were independent of the reduction by APC of infarction volume. Our data show that delayed APC administration is neuroprotective and mediates brain repair (i.e., neovascularization and neurogenesis), suggesting a significant extension of the therapeutic window for APC intervention in postischemic brain.


Subject(s)
Brain Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Neurogenesis/drug effects , Protein C/pharmacology , Receptor, PAR-1/genetics , Recovery of Function/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/enzymology , Brain/metabolism , Brain/physiopathology , Brain Infarction/drug therapy , Brain Infarction/enzymology , Brain Infarction/physiopathology , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cerebral Arteries/enzymology , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Disease Models, Animal , Enzyme Activation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/physiology , Nerve Regeneration/physiology , Neurogenesis/physiology , Protein C/metabolism , Recovery of Function/physiology , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology , Tissue Plasminogen Activator/toxicity , Treatment Outcome
11.
FASEB J ; 20(13): 2313-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17077308

ABSTRACT

Recent studies suggest that moderate red wine consumption reduces the incidence of Alzheimer's disease (AD) clinical dementia. Using Tg2576 mice, which model AD-type amyloid beta-protein (Abeta) neuropathology, we tested whether moderate consumption of the red wine Cabernet Sauvignon modulates AD-type neuropathology and cognitive deterioration. The wine used in the study was generated using Cabernet Sauvignon grapes from Fresno, California, and was delivered to Tg2576 in a final concentration of approximately 6% ethanol. We found that Cabernet Sauvignon significantly attenuated AD-type deterioration of spatial memory function and Abeta neuropathology in Tg2576 mice relative to control Tg2576 mice that were treated with either a comparable amount of ethanol or water alone. Chemical analysis showed the Cabernet Sauvignon used in this study contains a very low content of resveratrol (0.2 mg/L), 10-fold lower than the minimal effective concentration shown to promote Abeta clearance in vitro. Our studies suggest Cabernet Sauvignon exerts a beneficial effect by promoting nonamyloidogenic processing of amyloid precursor protein, which ultimately prevents the generation of Abeta peptides. This study supports epidemiological evidence indicating that moderate wine consumption, within the range recommended by the FDA dietary guidelines of one drink per day for women and two for men, may help reduce the relative risk for AD clinical dementia.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Cognition Disorders/prevention & control , Wine , Alanine Transaminase/blood , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartate Aminotransferases/blood , Disease Models, Animal , Female , Flavonoids/isolation & purification , Flavonoids/therapeutic use , Mice , Mice, Transgenic , Phenols/isolation & purification , Phenols/therapeutic use , Polyphenols , Protein Processing, Post-Translational , Wine/analysis , Wine/toxicity
12.
Nat Med ; 12(11): 1278-85, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17072311

ABSTRACT

Brain hemorrhage is a serious complication of tissue plasminogen activator (tPA) therapy for ischemic stroke. Here we report that activated protein C (APC), a plasma serine protease with systemic anticoagulant, anti-inflammatory and antiapoptotic activities, and direct vasculoprotective and neuroprotective activities, blocks tPA-mediated brain hemorrhage after transient brain ischemia and embolic stroke in rodents. We show that APC inhibits a pro-hemorrhagic tPA-induced, NF-kappaB-dependent matrix metalloproteinase-9 pathway in ischemic brain endothelium in vivo and in vitro by acting through protease-activated receptor 1. The present findings suggest that APC may improve thrombolytic therapy for stroke, in part, by reducing tPA-mediated hemorrhage.


Subject(s)
Intracranial Hemorrhages/prevention & control , Protein C/pharmacology , Tissue Plasminogen Activator/pharmacology , Animals , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , NF-kappa B/metabolism , Promoter Regions, Genetic , Recombinant Proteins/pharmacology
13.
J Biol Chem ; 281(31): 21745-21754, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16751189

ABSTRACT

Nicotinamide adenine dinucleotide (NAD)+-dependent sirtuins have been identified to be key regulators in the lifespan extending effects of calorie restriction (CR) in a number of species. In this study we report for the first time that promotion of the NAD+-dependent sirtuin, SIRT1-mediated deacetylase activity, may be a mechanism by which CR influences Alzheimer disease (AD)-type amyloid neuropathology. Most importantly, we report that the predicted attenuation of beta-amyloid content in the brain during CR can be reproduced in mouse neurons in vitro by manipulating cellular SIRT1 expression/activity through mechanisms involving the regulation of the serine/threonine Rho kinase ROCK1, known in part for its role in the inhibition of the non-amyloidogenic alpha-secretase processing of the amyloid precursor protein. Conversely, we found that the expression of constitutively active ROCK1 in vitro cultures significantly prevented SIRT1-mediated response, suggesting that alpha-secretase activity is required for SIRT1-mediated prevention of AD-type amyloid neuropathology. Consistently we found that the expression of exogenous human (h) SIRT1 in the brain of hSIRT1 transgenics also resulted in decreased ROCK1 expression and elevated alpha-secretase activity in vivo. These results demonstrate for the first time a role for SIRT1 activation in the brain as a novel mechanism through which CR may influence AD amyloid neuropathology. The study provides a potentially novel pharmacological strategy for AD prevention and/or treatment.


Subject(s)
Alzheimer Disease/diet therapy , Caloric Restriction , Neurons/enzymology , Sirtuins/metabolism , Alzheimer Disease/prevention & control , Amyloid/analysis , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Endopeptidases/metabolism , Enzyme Activation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurons/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Sirtuin 1 , rho-Associated Kinases
14.
BMC Neurosci ; 7: 29, 2006 Apr 03.
Article in English | MEDLINE | ID: mdl-16584562

ABSTRACT

BACKGROUND: The cause of neuronal death in amyotrophic lateral sclerosis (ALS) is uncertain but mitochondrial dysfunction may play an important role. Ketones promote mitochondrial energy production and membrane stabilization. RESULTS: SOD1-G93A transgenic ALS mice were fed a ketogenic diet (KD) based on known formulations for humans. Motor performance, longevity, and motor neuron counts were measured in treated and disease controls. Because mitochondrial dysfunction plays a central role in neuronal cell death in ALS, we also studied the effect that the principal ketone body, D-beta-3 hydroxybutyrate (DBH), has on mitochondrial ATP generation and neuroprotection. Blood ketones were > 3.5 times higher in KD fed animals compared to controls. KD fed mice lost 50% of baseline motor performance 25 days later than disease controls. KD animals weighed 4.6 g more than disease control animals at study endpoint; the interaction between diet and change in weight was significant (p = 0.047). In spinal cord sections obtained at the study endpoint, there were more motor neurons in KD fed animals (p = 0.030). DBH prevented rotenone mediated inhibition of mitochondrial complex I but not malonate inhibition of complex II. Rotenone neurotoxicity in SMI-32 immunopositive motor neurons was also inhibited by DBH. CONCLUSION: This is the first study showing that diet, specifically a KD, alters the progression of the clinical and biological manifestations of the G93A SOD1 transgenic mouse model of ALS. These effects may be due to the ability of ketone bodies to promote ATP synthesis and bypass inhibition of complex I in the mitochondrial respiratory chain.


Subject(s)
Amyotrophic Lateral Sclerosis/diet therapy , Amyotrophic Lateral Sclerosis/metabolism , Diet , Ketone Bodies/biosynthesis , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Survival/drug effects , Disease Progression , Hydroxybutyrate Dehydrogenase/pharmacology , Ketone Bodies/blood , Male , Mice , Mice, Transgenic , Mitochondria/drug effects , Motor Neurons/drug effects , Superoxide Dismutase/genetics
15.
J Neurosurg ; 101(4): 669-75, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15481724

ABSTRACT

OBJECT: The authors evaluated the neuroprotective effect of 5,10,15,20-tetrakis(N-methyl-4'-pyridyl)porphyrinato-iron(III) (FeTMPyP), a peroxynitrite decomposition catalyst, and 1,5-isoquinolinediol (ISO), a poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitor, alone and in combination in rats with focal cerebral ischemia induced by middle cerebral artery occlusion (MCAO). METHODS: Male Sprague-Dawley rats were subjected to 2 hours of MCAO followed by 22 hours of reperfusion. Cerebral infarction and neurological deficits were estimated after ischemia. Intraperitoneal injections of FeTMPyP (1 and 2 mg/kg) and ISO (0.05 and 0.1 mg/kg) were administered alone or in combination in ischemic animals. The PARP activity in vehicle- and drug-treated groups was estimated using anti-poly(ADP-ribose) antibody in immunofluorescence and immunoblotting studies. Two hours of MCAO and 22 hours of reperfusion produced significant cerebral infarction and neurological deficits. Treatment with FeTMPyP (1 and 2 mg/kg) and ISO (0.05 and 0.1 mg/kg) produced a significant reduction in cerebral infarction and neurological deficits. Combination therapy (2 mg/kg FeTMPyP and 0.1 mg/kg ISO) enhanced the inhibition of ischemic volume (77.81+/-0.86%) compared with monotherapies (FeTMPyP 54.07+/-5.6% and ISO 53.06+/-3.88%). Immunoblotting and immunofluorescence studies showed PARP activation after ischemia, which was reduced by drug treatment. CONCLUSIONS: Neuroprotection observed with FeTMPyP and ISO alone and in combination may be attributed to inhibition of the peroxynitrite-PARP cascade of cerebral ischemia/reperfusion injury.


Subject(s)
Brain Ischemia/prevention & control , Metalloporphyrins/pharmacology , Quinolines/pharmacology , Animals , Brain Ischemia/veterinary , Cerebral Arterial Diseases/complications , Cerebral Arterial Diseases/veterinary , Drug Therapy, Combination , Isoquinolines , Male , Poly(ADP-ribose) Polymerase Inhibitors , Rats , Rats, Sprague-Dawley
16.
Brain Res Bull ; 62(5): 425-33, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-15168908

ABSTRACT

In the present study, neuroprotective effect of 4-amino-1,8-napthalimide (4-ANI), a poly(ADP-ribose) polymerase (PARP) inhibitor was investigated in middle cerebral artery occlusion (MCAo)-induced focal ischemia. Sprague-Dawley rats were subjected to 2 h of middle cerebral artery occlusion followed by 22 h of reperfusion. After 22 h of reperfusion rats were evaluated for cerebral infarction, neurological deficits, brain NAD levels, and in situ terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling (TUNEL). Focal ischemia produced significant infarct volume (201 +/- 14 mm3), neurological scores (2 +/- 0.5) and 28 +/- 4.5% brain NAD depletion. Ischemia was associated with increased in TUNEL positive cells in brain sections indicating DNA fragmentation. 4-ANI treatment (1 and 3 mg/kg, i.p.) significantly decreased infarct volume to 35 +/- 7% and 70 +/- 6%, respectively. Neurological functions were also significantly improved at these doses. 4-ANI (3 mg/kg) completely reversed brain NAD depletion and significantly reduced the increase in the number of TUNEL positive cells. Nevertheless, 4-ANI treatment did not alter cerebral blood flow and blood pressure. Our study suggests 4-ANI is a potent neuroprotective agent in focal cerebral ischemia and its neuroprotective effects may be attributed to reduction of NAD depletion and DNA fragmentation.


Subject(s)
1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Enzyme Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Quinolones/therapeutic use , Animals , Brain Chemistry/drug effects , Brain Ischemia/etiology , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/complications , Male , NAD/analysis , Naphthalimides , Poly(ADP-ribose) Polymerases/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy
17.
Br J Pharmacol ; 142(5): 899-911, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15197101

ABSTRACT

Free radicals have been implicated in cerebral ischemia reperfusion (IR) injury. Massive production of nitric oxide and superoxide results in continuous formation of peroxynitrite even several hours after IR insult. This can produce DNA strand nicks, hydroxylation and/or nitration of cytosolic components of neuron, leading to neuronal death. Peroxynitrite decomposition catalysts 5,10,15,20-tetrakis(N-methyl-4'-pyridyl)porphyrinato iron (III) (FeTMPyP) and 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron (III) (FeTPPS) have been demonstrated to protect neurons in in vitro cultures; however, their neuroprotective efficacy in cerebral IR injury has not been explored. In the present study, we investigated the efficacy and the therapeutic time window of FeTMPyP and FeTPPS in focal cerebral ischemia (FCI). FCI was induced according to the middle cerebral artery occlusion (MCAO) method. After 2 h of MCAO and 70 h of reperfusion, the extent of neurological deficits, infarct and edema volume were measured in Sprague-Dawley rats. FeTMPyP and FeTPPS were administered at different time points 2, 6, 9 and 12 h post MCAO. FeTMPyP and FeTPPS (3 mg kg(-1), i.v.) treatment at 2 and 6 h post MCAO produced significant reduction in infarct volume, edema volume and neurological deficits. However, treatment at latter time points did not produce significant neuroprotection. Significant reduction of peroxynitrite in blood and nitrotyrosine in brain sections was observed on FeTMPyP and FeTPPS treatment. As delayed treatment of FeTMPyP and FeTPPS produced neuroprotection, we tested whether treatment had any influence over the apoptotic neuronal death. DNA fragmentation and in situ nick end-labeling assays showed that FeTMPyP and FeTPPS treatment reduced IR injury-induced DNA fragmentation. In conclusion, peroxynitrite decomposition catalysts (FeTMPyP and FeTPPS) produced prominent neuroprotection even if administered 6 h post MCAO and the neuroprotective effect is at least in part due to the reduction of peroxynitrite and apoptosis.


Subject(s)
Brain Ischemia/drug therapy , Metalloporphyrins/therapeutic use , Neuroprotective Agents/therapeutic use , Peroxynitrous Acid/metabolism , Animals , Apoptosis/drug effects , Brain/pathology , Brain Edema/pathology , Brain Ischemia/pathology , Catalysis , DNA Fragmentation/drug effects , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Kinetics , Male , Microscopy, Electron , Middle Cerebral Artery/physiology , Nervous System Diseases/psychology , Rats , Rats, Sprague-Dawley , Tyrosine/metabolism
18.
Life Sci ; 74(8): 969-85, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14672754

ABSTRACT

Free radical induced neuronal damage is implicated in cerebral ischemia reperfusion (IR) injury and antioxidants are reported to have neuroprotective activity. Several in vitro and in vivo studies have proved the antioxidant potential of curcumin and its metabolites. Hence, in the present study the neuroprotective potential of curcumin was investigated in middle cerebral artery occlusion (MCAO) induced focal cerebral IR injury. 2 h of MCAO and 22 h of reperfusion resulted in the infarct volume of 210.39 +/- 31.25 mm3. Administration of curcumin 100 and 300 mg/kg, i.p. 30 min. after MCAO produced 37.23 +/- 5.10% and 46.39 +/- 10.23% (p < 0.05) reduction in infarct volume, respectively. Ischemia induced cerebral edema was reduced in a dose dependent manner. Curcumin at 300 mg/kg, i.p. produced 50.96 +/- 6.04% reduction in edema (p < 0.05) volume. Increase in lipid peroxidation after MCAO in ipsilateral and contralateral hemisphere of brain was observed, which was reduced by curcumin (300 mg/kg, i.p.)-treatment. Decrease in superoxide dismutase and glutathione peroxidase activity was observed in ipsilateral hemisphere of MCAO animal. Curcumin-treatment (300 mg/kg, i.p.) prevented IR injury mediated fall in glutathione peroxide activity. Peroxynitrite measured using rhodamine123 fluorescence and anti-nitrotyrosine immunofluorescence indicated increased peroxynitrite formation after IR insult. Curcumin-treatment reduced peroxynitrite formation and hence the extent of tyrosine nitration in the cytosolic proteins. These results suggest the neuroprotective potential of curcumin in cerebral ischemia and is mediated through its antioxidant activity.


Subject(s)
Brain Ischemia/drug therapy , Curcumin/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Body Temperature/drug effects , Brain/pathology , Brain Edema/drug therapy , Brain Edema/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Circulation/drug effects , Fluorescent Antibody Technique , Glutathione Peroxidase/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Lipid Peroxidation/drug effects , Male , Peroxynitrous Acid/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Tyrosine/metabolism
19.
Eur J Pharmacol ; 453(2-3): 335-44, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12398922

ABSTRACT

The present study was undertaken to investigate the effect of testosterone on the alpha-adrenoceptor-mediated contractile responses in ventral lobe of rat prostate. Contractile responses to various alpha-adrenoceptor agonists (phenylephrine, A61603 (N-[5-(4,5-dihydro-1H-imidazol-2-yl)-2-hydroxy-5,6,7,8-tetrahydronaphthalen-1-yl] methanesulphonamide), clonidine, guanfacine, ST587 ((2,3-dihydro-benzo[1,4]dioxin-2-ylmethyl)-[2-(2,6-dimethoxy-phenoxy)-ethyl]-amine) and xylazine) were tested in prostate strips obtained from control and testosterone (3 mg/kg, s.c. 5 days a week for 15 days-10 doses total)-treated rats. Dose-response curves for alpha-adrenoceptor agonists in testosterone-treated animals showed a leftward shift, indicating increased sensitivity of tissue to alpha-adrenoceptor agonists. To find the mechanism of increased sensitivity, K(A) value and receptor reserve of phenylephrine were estimated. Neither the K(A) value nor the receptor reserve of phenylephrine was altered in testosterone-treated rats. The concentration-occupancy curve for A61603 was shifted leftward and the K(A) value for A61603 decreased about four-fold. The K(B) value of 2-(2,6-dimethoxyphenoxyethyl) aminomethyl-1,4-benzodioxane (WB4101) was not altered, however, the K(B) value for prazosin was decreased approximately 5.5-fold. These findings indicate that the testosterone-mediated increase in sensitivity of prostate to alpha-adrenoceptor agonists is due to alterations in the alpha(1)-adrenoceptor pool.


Subject(s)
Prostate/drug effects , Receptors, Adrenergic, alpha/drug effects , Testosterone/pharmacology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Injections, Subcutaneous , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Phenylephrine/pharmacology , Prostate/physiology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha/metabolism , Testosterone/physiology , Tetrahydronaphthalenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...