Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Pharm Nanotechnol ; 8(3): 207-224, 2020.
Article in English | MEDLINE | ID: mdl-32416685

ABSTRACT

BACKGROUND: Carvedilol (CD), a non-selective beta-blocker, is indicated for the management of mild to moderate congestive heart failure. After oral administration, CD is rapidly absorbed with an absolute bioavailability of 18-25% because of low solubility and extensive first-pass metabolism. OBJECTIVE: The present investigation focused on enhanced oral delivery of CD using supersaturated self-emulsifying drug delivery (SEDDS) system. METHODS: Optimized SEDDS consisted of a blend of Oleic acid and Labrafil-M2125 as an oil-phase, Cremophor-RH40, polyethylene glycol-400 and HPMC-E5 as a surfactant, co-surfactant and supersaturation promoter respectively. Formulations were characterized for physical characteristics, invitro release in simulated and biorelevant dissolution media, intestinal permeability and bioavailability studies in Wistar rats. Differential scanning calorimetry (DSC) and X-ray diffraction (XRD) analysis, scanning electron microscopy (SEM) studies were used to confirm the crystalline nature and shape of the optimized formulation. RESULTS: DSC and XRD, SEM studies showed that the drug was in amorphous form, and droplets were spherical in shape. Dissolution studies clearly showed distinct CD release in compendial and biorelevant dissolution media. The results from permeability and in-vivo studies depicted 2.2-folds and 3.2-folds increase in permeability and bioavailability, respectively from supersaturated SEDDS in comparison with control. CONCLUSION: The results conclusively confirmed that the SEDDS formulation could be considered as a new alternative delivery vehicle for the oral supply of CD. Lay Summary: Carvedilol (CD) is a non-selective antihypertensive drug with poor oral bioavailability. Previously, various lipid delivery systems were reported with enhanced oral delivery. We developed suprsaturable SEDDS formulation with immediate onset of action. SEDDS formulation was developed and optimized as per the established protocols. The optimized SEDDS formulation was stable over three months and converted to solid and supersaturated SEDDS. The results from permeability and in-vivo studies demonstrated an enhancement in permeability and bioavailability from supersaturated SEDDS in comparison with control. The results conclusively confirmed that the SEDDS formulation could be considered as a new alternative delivery vehicle for the oral administration of CD.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Carvedilol/pharmacokinetics , Drug Delivery Systems/methods , Emulsifying Agents/pharmacokinetics , Intestinal Absorption/drug effects , Administration, Oral , Adrenergic beta-Antagonists/administration & dosage , Animals , Carvedilol/administration & dosage , Emulsifying Agents/administration & dosage , Intestinal Absorption/physiology , Male , Organ Culture Techniques , Rats , Rats, Wistar , Solubility
2.
J Neurotrauma ; 36(11): 1869-1875, 2019 06.
Article in English | MEDLINE | ID: mdl-30704365

ABSTRACT

Traumatic brain injury (TBI) has many long-term consequences, including impairment in memory and changes in mood. Glycogen synthase kinase 3ß (GSK-3ß) in its phosphorylated form (p-GSK-3ß) is considered to be a major contributor to memory problems that occur post-TBI. We have developed an antisense that targets the GSK-3ß (GAO) gene. Using a model of closed-head concussive TBI, we subjected mice to TBI and injected GAO or a random antisense (RAO) 15 min post-injury. One week post-injury, mice were tested in object recognition with 24 h delay. At 4 weeks post- injury, mice were tested with a T-maze foot shock avoidance memory test and a second object recognition test with 24 h delay using different objects. Mice that received GAO show improved memory in both object recognition and T-maze compared with RAO- treated mice that were subjected to TBI. Next, we verified that GAO blocked the surge in phosphorylated GSK-3ß post-TBI. Mice were subjected to TBI and injected with antisense 15 min post-TBI with GAO or RAO. Mice were euthanized at 4 and 72 h post-TBI. Analysis of p-ser9GSK-3ß, p-tyr216GSK-3ß, and phospho-tau (p-tau)404 showed that mice that received a TBI+RAO had significantly higher p-ser9GSK-3ß, p-tyr216GSK-3ß, and p-tau404 levels than the mice that received TBI+GAO and the Sham+RAO mice. The current finding suggests that inhibiting GSK-3ß increase after TBI with an antisense directed at GSK-3ß prevents learning and memory impairments.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/enzymology , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/etiology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Animals , Maze Learning/drug effects , Mice , Oligonucleotides, Antisense/pharmacology
3.
Eur J Pharmacol ; 845: 65-73, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30579934

ABSTRACT

3,4-Dihydroxyphenylacetaldehyde (DOPAL), the monoamine oxidase (MAO) metabolite of dopamine, plays a role in pathogenesis of Parkinson disease, inducing α-synuclein aggregation. DOPAL generates discrete α-synuclein aggregates. Inhibiting this aggregation could provide therapy for slowing Parkinson disease progression. Primary and secondary amines form adducts with aldehydes. Rasagiline and aminoindan contain these amine groups. DOPAL-induced α-synuclein aggregates were resolved in the presence and absence of rasagiline or aminoindan using quantitative Western blotting. DOPAL levels in incubation mixtures, containing increased rasagiline or aminoindan concentrations, were determined by high pressure liquid chromatography (HPLC). Schiff base adducts between DOPAL and rasagiline or aminoindan were determined using mass spectrometry. A neuroprotective effect of rasagiline and aminoindan against DOPAL-induced toxicity was demonstrated using PC-12 cells. Rasagiline and aminoindan significantly reduced aggregation of α-synuclein of all sizes in test tube and PC-12 cells experiments. Dimethylaminoindan did not reduce aggregation. DOPAL levels in incubation mixtures were reduced with increasing rasagiline or aminoindan concentrations but not with dimethylaminoindan. Schiff base adducts between DOPAL and either rasagiline or aminoindan were demonstrated by mass spectrometry. A neuroprotective effect against DOPAL-induced toxicity in PC-12 cells was demonstrated for both rasagiline and aminoindan. Inhibiting DOPAL-induced α-synuclein aggregation through amine adducts provides a therapeutic approach for slowing Parkinson disease progression.


Subject(s)
3,4-Dihydroxyphenylacetic Acid/analogs & derivatives , Aldehydes/pharmacology , Indans/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , alpha-Synuclein/metabolism , 3,4-Dihydroxyphenylacetic Acid/toxicity , Aldehydes/therapeutic use , Animals , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , PC12 Cells , Rats
4.
J Alzheimers Dis ; 54(4): 1339-1348, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27589526

ABSTRACT

Glycogen synthase kinase (GSK)-3ß is a multifunctional protein that has been implicated in the pathological characteristics of Alzheimer's disease (AD), including the heightened levels of neurofibrillary tangles, amyloid-beta (Aß), and neurodegeneration. We have previously shown that an antisense oligonucleotide directed at the Tyr 216 site on GSK-3ß (GAO) when injected centrally can decrease GSK-3ß levels, improve learning and memory, and decrease oxidative stress. In addition, we showed that GAO can cross the blood-brain barrier. Herein the impact of peripherally administered GAO in both the non-transgenic SAMP8 and transgenic Tg2576 (APPswe) models of AD were examined respective to learning and memory. Brain tissues were then evaluated for expression changes in the phosphorylated-Tyr 216 residue, which leads to GSK-3ß activation, and the phosphorylated-Ser9 residue, which reduces GSK-3ß activity. SAMP8 GAO-treated mice showed improved acquisition and retention using aversive T-maze, and improved declarative memory as measured by the novel object recognition (NOR) test. Expression of the phosphorylated-Tyr 216 was decreased and the phosphorylated-Ser9 was increased in GAO-treated SAMP8 mice. Tg2576 GAO-treated mice improved acquisition and retention in both the T-maze and NOR tests, with an increased phosphorylated-Ser9 GSK-3ß expression. Results demonstrate that peripheral administration of GAO improves learning and memory, corresponding with alterations in GSK-3ß phosphorylation state. This study supports peripherally administered GAO as a viable means to mediate GSK-3ß activity within the brain and a possible treatment for AD.


Subject(s)
Alzheimer Disease/drug therapy , Cellular Senescence/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Maze Learning/drug effects , Memory/drug effects , Oligonucleotides, Antisense/administration & dosage , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Animals , Cellular Senescence/physiology , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Transgenic
5.
J Alzheimers Dis ; 46(2): 535-48, 2015.
Article in English | MEDLINE | ID: mdl-25854933

ABSTRACT

The senescence-accelerated mouse (SAMP8) strain exhibits an age-related decrease in memory accompanied by an increase in hippocampal amyloid-ß protein precursor (AßPP) and amyloid-ß peptide (Aß). We have shown that administration of an antisense oligonucleotide against the Aß region of AßPP (AßPP antisense) reverses the memory deficits. The purpose of this study was to determine the effect of peripheral (IV) administration of AßPP antisense on hippocampal gene expression. The AßPP antisense reversed the memory deficits and altered expression of 944 hippocampal genes. Pathway analysis showed significant gene expression changes in nine pathways. These include the MAPK signaling pathway (p = 0.0078) and the phosphatidylinositol signaling pathway (p = 0.043), which we have previously shown to be altered in SAMP8 mice. The changes in these pathways contributed to significant changes in the neurotropin (p = 0.0083) and insulin signaling (p = 0.015) pathways, which are known to be important in learning and memory. Changes in these pathways were accompanied by phosphorylation changes in the downstream target proteins p70S6K, GSK3ß, ERK, and CREB. These changes in hippocampal gene expression and protein phosphorylation may suggest specific new targets for antisense therapy aimed at improving memory.


Subject(s)
Amyloid beta-Peptides/chemistry , Hippocampus/drug effects , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory/drug effects , Oligonucleotides, Antisense/administration & dosage , Animals , Disease Models, Animal , Gene Expression , Mice , Phosphorylation , Signal Transduction
6.
Neurobiol Aging ; 35(1): 159-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23969180

ABSTRACT

The senescence-accelerated mouse (SAMP8) strain exhibits decreased learning and memory and increased amyloid beta (Aß) peptide accumulation at 12 months. To detect differences in gene expression in SAMP8 mice, we used a control mouse that was a 50% cross between SAMP8 and CD-1 mice and which showed no memory deficits (50% SAMs). We then compared gene expression in the hippocampus of 4- and 12-month-old SAMP8 and control mice using Affymetrix gene arrays. At 12 months, but not at 4 months, pathway analysis revealed significant differences in the long term potentiation (6 genes), phosphatidylinositol signaling (6 genes), and endocytosis (10 genes) pathways. The changes in long term potentiation included mitogen-activated protein kinase (MAPK) signaling (N-ras, cAMP responsive element binding protein [CREB], protein phosphatase inhibitor 1) and Ca-dependent signaling (inositol triphosphate [ITP] receptors 1 and 2 and phospholipase C). Changes in phosphatidylinositol signaling genes suggested altered signaling through phosphatidylinositol-3-kinase, and Western blotting revealed phosphorylation changes in serine/threonine protein kinase AKT and 70S6K. Changes in the endocytosis pathway involved genes related to clathrin-mediated endocytosis (dynamin and clathrin). Endocytosis is required for receptor recycling, is involved in Aß metabolism, and is regulated by phosphatidylinositol signaling. In summary, these studies demonstrate altered gene expression in 3 SAMP8 hippocampal pathways associated with memory formation and consolidation. These pathways might provide new therapeutic targets in addition to targeting Aß metabolism itself.


Subject(s)
Aging/genetics , Endocytosis/genetics , Endocytosis/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression , Hippocampus , Long-Term Potentiation/genetics , Memory Disorders/genetics , Phosphatidylinositols/genetics , Signal Transduction/genetics , Aging/physiology , Amyloid beta-Peptides/metabolism , Animals , Gene Expression Regulation, Developmental/physiology , Hippocampus/metabolism , Hippocampus/physiology , Learning/physiology , Long-Term Potentiation/physiology , Memory/physiology , Mice , Mice, Inbred Strains , Phosphatidylinositols/physiology , Signal Transduction/physiology
7.
Free Radic Biol Med ; 65: 1-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23777706

ABSTRACT

Amyloid ß-peptide (Aß) plays a central role in the pathophysiology of Alzheimer's disease (AD) through the induction of oxidative stress. This peptide is produced by proteolytic cleavage of amyloid precursor protein (APP) by the action of ß- and γ-secretases. Previous studies demonstrated that reduction of Aß, using an antisense oligonucleotide (AO) directed against the Aß region of APP, reduced oxidative stress-mediated damage and prevented or reverted cognitive deficits in senescence-accelerated prone mice (SAMP8), a useful animal model for investigating the events related to Aß pathology and possibly to the early phase of AD. In the current study, aged SAMP8 were treated by AO directed against PS-1, a component of the γ-secretase complex, and tested for learning and memory in T-maze foot shock avoidance and novel object recognition. Brain tissue was collected to identify the decrease of oxidative stress and to evaluate the proteins that are differently expressed and oxidized after the reduction in free radical levels induced by Aß. We used both expression proteomics and redox proteomics approaches. In brain of AO-treated mice a decrease of oxidative stress markers was found, and the proteins identified by proteomics as expressed differently or nitrated are involved in processes known to be impaired in AD. Our results suggest that the treatment with AO directed against PS-1 in old SAMP8 mice reverses learning and memory deficits and reduces Aß-mediated oxidative stress with restoration to the normal condition and identifies possible pharmacological targets to combat this devastating dementing disease.


Subject(s)
Hippocampus/drug effects , Maze Learning/drug effects , Oligonucleotides, Antisense/pharmacology , Oxidative Stress/drug effects , Presenilin-1/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Blotting, Western , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Hippocampus/metabolism , Immunoprecipitation , Mass Spectrometry , Mice , Proteomics
8.
Curr Pharm Des ; 18(8): 1123-30, 2012.
Article in English | MEDLINE | ID: mdl-22288401

ABSTRACT

The senescence accelerate mouse P8 (SAMP8) is an excellent model of early learning and memory problems. A number of studies have shown that it has cholinergic deficits, oxidative damage, alterations in membrane lipids and circadian rhythm disturbances. The brains of the SAMP8 overproduce amyloid precursor protein (APP), amyloid-beta protein and have an increased physphorylation of tau. An antisense to APP has been developed that reverses the cognitive deficits and oxidative damage. This antisense represents a potential treatment for Alzheimer's disease.


Subject(s)
Alzheimer Disease/physiopathology , Disease Models, Animal , Oligonucleotides, Antisense/pharmacology , Aging , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/physiopathology , Circadian Rhythm , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Drug Design , Humans , Membrane Lipids/metabolism , Mice , Oxidative Stress/drug effects
9.
Biochim Biophys Acta ; 1822(5): 650-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22142563

ABSTRACT

The senescence accelerated mouse (SAMP8) is a spontaneous animal model of overproduction of amyloid precursor protein (APP) and oxidative damage. It develops early memory disturbances and changes in the blood-brain barrier resulting in decreased efflux of amyloid-ß protein from the brain. It has a marked increase in oxidative stress in the brain. Pharmacological treatments that reduce oxidative stress improve memory. Treatments that reduce amyloid-ß (antisense to APP and antibodies to amyloid-ß) not only improve memory but reduce oxidative stress. Early changes in lipid peroxidative damage favor mitochondrial dysfunction as being a trigger for amyloid-ß overproduction in this genetically susceptible mouse strain. This sets in motion a cycle where the increased amyloid-beta further damages mitochondria. We suggest that this should be termed the Inflammatory-Amyloid Cycle and may well be similar to the mechanisms responsible for the pathophysiology of Alzheimer's disease. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.


Subject(s)
Aging/physiology , Alzheimer Disease/metabolism , Disease Models, Animal , Oxidative Stress , Aging/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Hormesis , Melatonin/physiology , Mice , Mitochondria/metabolism
10.
Int J Androl ; 34(1): 55-68, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20403060

ABSTRACT

Testosterone regulates energy metabolism and skeletal muscle mass in males, but the molecular mechanisms are not fully understood. This study investigated the response of skeletal muscle to castration and testosterone replacement in 8-week-old male mice. Using microarray analyses of mRNA levels in gastrocnemius muscle, 91 genes were found to be negatively regulated by testosterone and 68 genes were positively regulated. The mRNA levels of the insulin signalling suppressor molecule Grb10 and the glycogen synthesis inhibitors, protein phosphatase inhibitor-1 and phosphorylase kinase-γ, were negatively regulated by testosterone. The insulin-sensitive glucose and amino acid transporters, Glut3 and SAT2, the lipodystrophy gene, Lpin1 and protein targeting to glycogen were positively regulated. These changes would be expected to increase nutrient availability and sensing within skeletal muscle, increase metabolic rate and carbohydrate utilization and promote glycogen accumulation. The observed positive regulation of atrogin-1 (Fbxo32) by testosterone could be explained by the phosphorylation of Akt and Foxo3a, as determined by Western blotting. Testosterone prevented the castration-induced increase in interleukin-1α, the decrease in interferon-γ and the atrophy of the levator ani muscle, which were all correlated with testosterone-regulated gene expression. These findings identify specific mechanisms by which testosterone may regulate skeletal muscle glucose and protein metabolism.


Subject(s)
Gene Expression Regulation , Glucose/metabolism , Muscle, Skeletal/metabolism , Proteins/metabolism , Testosterone/administration & dosage , Acetyltransferases/genetics , Animals , GRB10 Adaptor Protein/genetics , Gene Expression , Gene Expression Profiling , Glucose Transporter Type 3/genetics , Interferon-gamma/genetics , Interleukin-1alpha/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Muscle Proteins/genetics , Nuclear Proteins/genetics , Orchiectomy , Phosphatidate Phosphatase , Phosphorylase Kinase/genetics , RNA, Messenger/analysis , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , Testosterone/blood
11.
J Alzheimers Dis ; 23(4): 599-605, 2011.
Article in English | MEDLINE | ID: mdl-21098986

ABSTRACT

The blood-brain barrier (BBB) influences brain levels of amyloid-ß (Aß) by transporting Aß out of the brain (efflux) and by the reabsorption of cerebrospinal fluid (CSF) into the blood stream (bulk flow). In Alzheimer's disease (AD) and normal aging, unknown factors impair Aß efflux and bulk flow in aging and in AD. These impairments have been proposed as mechanisms by which the Aß burden in brain can increase. Impairment in Aß efflux occurs in animal models of AD, including the aged SAMP8 mouse. Here, we show that CSF reabsorption is also reduced by about 50% in SAMP8 mice (p < 0.05). We then determined whether an antisense directed at the Aß region of the amyloid-ß protein precursor (AßPP) and previously shown to decrease brain levels of AßPP and to reverse the cognitive impairments of the SAMP8 mouse was able to reverse these impairments. We found that the antisense restored both the CSF reabsorption, more than doubling the rate of efflux, and the saturable efflux of Aß. These findings suggest that AßPP/Aß itself contributes to the impairments in bulk flow and saturable efflux of Aß and that reduction of AßPP/Aß levels can restore normal function of the BBB.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Blood-Brain Barrier/metabolism , Disease Models, Animal , Oligodeoxyribonucleotides, Antisense/metabolism , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/pathology , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Protein Transport/physiology
12.
J Gerontol A Biol Sci Med Sci ; 64(10): 1025-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19531769

ABSTRACT

Amyloid beta protein (Abeta) in Alzheimer's disease induces oxidative stress through several mechanisms, including stimulation of nitric oxide synthase (NOS) activity. We examined NOS activity and expression in the senescence-accelerated mouse P8 (SAMP8) line. The SAMP8 strain develops with aging cognitive impairments, increases in Abeta, and oxidative stress, all reversed by amyloid precursor protein antisense or Abeta antibody treatment. We found here that hippocampal NOS activity in 12-month-old SAMP8 mice was nearly double that of 2-month-old SAMP8 or CD-1 mice, but with no change in NOS isoenzyme mRNA and protein levels. Antisense or antibody treatment further increased NOS activity in aged SAMP8 mice. Antisense treatment increased inducible NOS (iNOS) mRNA levels, decreased neuronal NOS mRNA and protein levels, but did not affect endothelial NOS (eNOS) or iNOS protein or eNOS mRNA levels. These results suggest a complex relation between Abeta and NOS in the SAMP8 that is largely mediated through posttranslational mechanisms.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/immunology , Antibodies/therapeutic use , Antisense Elements (Genetics)/therapeutic use , Immunologic Factors/therapeutic use , Nitric Oxide Synthase/metabolism , Age Factors , Alzheimer Disease/etiology , Alzheimer Disease/therapy , Animals , Disease Models, Animal , Hippocampus/enzymology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Nitric Oxide Synthase/genetics , RNA, Messenger/metabolism
13.
J Alzheimers Dis ; 17(3): 553-70, 2009.
Article in English | MEDLINE | ID: mdl-19433890

ABSTRACT

Decreased clearance is the main reason amyloid-beta protein (Abeta) is increased in the brains of patients with Alzheimer's disease (AD). The neurovascular hypothesis states that this decreased clearance is caused by impairment of low density lipoprotein receptor related protein-1 (LRP-1), the major brain-to-blood transporter of Abeta at the blood-brain barrier (BBB). As deletion of the LRP-1 gene is a lethal mutation, we tested the neurovascular hypothesis by developing a cocktail of phosphorothioate antisenses directed against LRP-1 mRNA. We found these antisenses in comparison to random antisense selectively decreased LRP-1 expression, reduced BBB clearance of Abeta42, increased brain levels of Abeta42, and impaired learning ability and recognition memory in mice. These results support dysfunction of LRP-1 at the BBB as a mechanism by which brain levels of Abeta could increase and AD would be promoted.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/drug effects , Brain/metabolism , Cognition Disorders/chemically induced , Lipoproteins, LDL/metabolism , Oligonucleotides, Antisense/pharmacology , Thionucleotides/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Animals , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Exploratory Behavior/drug effects , Lipoproteins, LDL/genetics , Male , Maze Learning/drug effects , Mice , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Transport/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recognition, Psychology/drug effects , Regression Analysis , Time Factors , Tissue Distribution
14.
J Exp Biol ; 212(Pt 4): 494-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19181896

ABSTRACT

Senescence-accelerated mice (SAMP8) serve as a model for Alzheimer's disease (AD) as they exhibit early loss of memory and increased amyloid precursor protein (APP) expression. APP is a ubiquitous membrane protein that is physiologically processed by site-specific proteolysis firstly by alpha- or beta-secretases, releasing a large fragment called APP(S) that contains most of the extracellular sequences of APP, a small extracellular stub, the transmembrane region and the cytoplasmic tail of APP (;AICD'-APP intracellular domain). These are subsequently cleaved by gamma-secretase at multiple sites in the transmembrane region, releasing small peptides, Abeta(1-40) and Abeta(1-42), the major components of AD-associated amyloid fibrils. gamma-secretase is a high-molecular-mass complex composed of presenilin-1 (PS1), nicastrin, APH-1 and Pen-2. As PS1 has been shown to play a critical role in facilitating gamma-secretase activity, and mutations in this protein are associated with familial AD (FAD), we have cloned it from SAMP8 mouse hippocampus and compared its sequence with those of other species. Furthermore, changes in the expression of PS1 with age in the hippocampal tissue of SAMP8 were studied. The results showed that the SAMP8 PS1 cDNA sequence is identical to that of normal mice. However, its expression in the hippocampus of SAMP8 exhibited an increase, while CD-1 mice, a strain that does not exhibit premature memory loss, showed no change with age. An increased amount or mutation(s) in PS1, which alters the stoichiometric balance of the gamma-secretase complex, may be the cause of aberrant or increased processing of APP, resulting in Abeta accumulation leading to loss of memory.


Subject(s)
Aging , Amyloid beta-Protein Precursor/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Age Factors , Alzheimer Disease/genetics , Amino Acid Sequence , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Gene Expression Regulation , Humans , Memory/physiology , Mice , Molecular Sequence Data , Mutation , Rats
15.
J Cereb Blood Flow Metab ; 29(2): 411-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19002200

ABSTRACT

By isolating for the first time ever a peptide transporter from the blood-brain barrier (BBB) and developing an antisense that selectively targets the brain-to-blood efflux component, we were able to deliver a therapeutic concentration of the neurotrophic peptide pituitary adenylate cyclase-activating polypeptide (PACAP) 27 to brain in animal models of Alzheimer's and stroke. Efflux pumps at the BBB are major causes of BBB impermeability to peptides. PACAP is neuroprotective in vitro in femtomole amounts, but brain uptake of PACAP27 is limited by an efflux component of peptide transport system-6 (PTS-6). Here, we characterized, isolated, and sequenced this component of PTS-6, identifying it as beta-F1 ATPase, and colocalized it with PACAP27 on BBB endothelial cells. Antisenses targeting the BBB inhibited PACAP27 efflux, thus increasing brain uptake of PACAP27. Treatment with antisense+PACAP27 improved cognition in a mouse model of Alzheimer's disease and reduced infarct size after cerebral ischemia. This represents the first isolation from BBB tissue of a peptide transporter and shows that inhibition of peptide efflux pumps is a potential strategy for drug delivery to brain.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Endothelial Cells/enzymology , Membrane Transport Proteins/isolation & purification , Membrane Transport Proteins/metabolism , Oligonucleotides, Antisense/genetics , Stroke/enzymology , Adenosine Triphosphatases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Disease Models, Animal , Genetic Therapy , Male , Membrane Transport Proteins/genetics , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Protein Binding , Stroke/genetics , Stroke/pathology , Stroke/therapy
16.
Acta Neuropathol ; 115(2): 193-203, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17965867

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by the selective loss of dopamine (DA) neurons and the presence of alpha-synuclein (AS) aggregates as Lewy bodies (LBs) in the remaining substantia nigra (SN) neurons. A continuing puzzle in studying PD pathogenesis is that although AS is expressed throughout the brain, LBs and selective dopaminergic cell loss lead to characteristic clinical signs of PD, suggesting that there is a link between AS aggregation and DA metabolism. One potential candidate for this link is the monoamine oxidase (MAO) metabolite of DA, 3,4-dihydroxyphenylacetaldehyde (DOPAL), as neither DA nor DA metabolites other than DOPAL are toxic to SN neurons at physiological concentrations. We tested DOPAL-induced AS aggregation in a cell-free system, in vitro in DA neuron cultures and in vivo with stereotactic injections into the SN of Sprague-Dawley rats by Western blots, fluorescent confocal microscopy and immunohistochemistry. We demonstrate that DOPAL in physiologically relevant concentrations, triggers AS aggregation in the cell-free system, and in cell cultures resulting in the formation of potentially toxic AS oligomers and aggregates. Furthermore, DOPAL injection into the SN of Sprague-Dawley rats resulted in DA neuron loss and the accumulation of high molecular weight oligomers of AS detected by Western blot. Our findings support the hypothesis that DA metabolism via DOPAL can cause both DA neuron loss and AS aggregation observed in PD.


Subject(s)
3,4-Dihydroxyphenylacetic Acid/metabolism , Brain/metabolism , Dopamine/metabolism , Lewy Bodies/metabolism , alpha-Synuclein/metabolism , Animals , Blotting, Western , Brain/pathology , Cells, Cultured , Immunohistochemistry , Lewy Bodies/pathology , Microscopy, Confocal , Monoamine Oxidase/metabolism , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley
17.
Peptides ; 27(4): 784-96, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16229925

ABSTRACT

Antisense potentially can manipulate target gene expression in the brain if it can cross the blood-brain barrier (BBB). We designed three (10mer, 17mer, and 19mer) phosphorothioated antisenses (PS-ODNs) directed against the precursor molecule of methionine enkephalin (Met-Enk), an opiate peptide which suppresses voluntary ethanol drinking. We measured the ability of the antisenses to cross the BBB, accumulate in the brain and CSF, decrease levels of Met-Enk in brain and blood, and affect voluntary ethanol drinking. Each antisense readily crossed the BBB, with 0.07-0.16% of the i.v. dose accumulating per gram of brain. Capillary depletion and CSF sampling each confirmed that the antisenses entered the CNS. Gel electrophoresis of radioactivity recovered from brain and serum showed intact antisense and a higher molecular weight form likely representing antisense bound to protein, but no degradation products. Each antisense molecule and a cocktail of all three reduced Met-Enk levels in brain and serum. Met-Enk levels in the brain were reduced more rapidly and for a longer duration than Met-Enk levels in the serum, indicating a degree of selective targeting to the CNS. Additionally, administration of the cocktail was more effective in reducing Met-Enk levels than any of the individual antisenses. Each antisense increased voluntary ethanol drinking by about 20% and the cocktail increased it by about 80%. Taken together, these results used pharmacokinetic, immunochemical, and behavioral methods to show that PS-ODN antisenses that readily cross the BBB can decrease brain levels of Met-Enk and increase voluntary ethanol drinking.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Drinking Behavior/physiology , Enkephalin, Methionine/deficiency , Enkephalins/genetics , Ethanol/administration & dosage , Protein Precursors/genetics , RNA, Antisense/metabolism , Animals , Biological Transport , Enkephalin, Methionine/metabolism , Enkephalins/biosynthesis , Gene Targeting , Injections, Intravenous , Male , Protein Precursors/biosynthesis , RNA, Antisense/genetics
18.
Life Sci ; 77(19): 2361-8, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-15946698

ABSTRACT

HIV-1 within the CNS produces a neuroAIDS syndrome and may act as a reservoir for reinfection of the peripheral tissues. Study of how HIV-1 crosses the blood-brain barrier (BBB) has been hampered by the lack of nonprimate animal models. However, BBB transport of HIV-1 does not involve any of the known steps conferring species specificity, including binding to CD4 receptors. In vivo and in vitro studies show that HIV-1 and its glycoprotein coat, gp120, are taken up and transported across the BBB of the mouse. Here, we compared the ability of gp120 and HIV-1 to be taken up by isolated brain microvessels (IBM) freshly isolated from mice, from post-mortem human brain, and from mice that had been treated in a manner analogous to the human material (mouse post-mortem). Freshly isolated mouse IBM took up more gp120 and HIV-1 than the human or mouse post-mortem cells. We found no difference between the ability of mouse post-mortem and human IBM to take up either gp120 or HIV-1. Wheatgerm agglutinin has been previously shown to stimulate gp120 and HIV-1 uptake by the BBB; here, it stimulated the uptake of gp120 and of HIV-1 by both mouse post-mortem and human IBM, although stimulated uptake was greatest for fresh mouse IBM. These results show that the mouse can be used to study the initial phases of HIV-1 uptake by the BBB.


Subject(s)
Blood-Brain Barrier/physiology , HIV Infections/virology , HIV-1 , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/pharmacology , Animals , Brain/virology , Capillaries/virology , Cell Line , Disulfides/pharmacology , HIV Envelope Protein gp120/metabolism , Humans , Mice , Species Specificity , Wheat Germ Agglutinins
19.
Peptides ; 26(5): 759-65, 2005 May.
Article in English | MEDLINE | ID: mdl-15808906

ABSTRACT

Orexin-A is a peptide produced in the lateral hypothalamus/perifornical area, which stimulates feeding. The production of orexin-A is determined by the metabolic state of the animal. We have previously shown that nitric oxide (NO) plays an important role as a mediator of feeding induced by a variety of neuropeptides. This raises the question of whether orexin-A's effects are NO dependent. Here, we first determined that intracerebroventricular administration of 25 ng of orexin-A significantly increased food intake in satiated mice. We next examined the effects of Nomega-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, on orexin-A-induced increase in food intake. L-NAME (50 mg/kg; SC) significantly blocked the orexin-A-induced increase in food intake. Orexin-A administration increased the levels of nitric oxide synthase in the hypothalamus. To further verify the importance of NO in the orexin-A-induced increase in food intake, we compared the ability of orexin-A to increase food intake in neuronal nitric oxide synthase knockout (NOS-KO) mice and their wild-type controls. Orexin-A failed to increase food intake in the NOS-KO mice, whereas it did increase food intake in the wild-type controls. This supports the hypothesis that nitric oxide is a central regulator of food consumption.


Subject(s)
Eating/physiology , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Neuropeptides/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Eating/drug effects , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuropeptides/administration & dosage , Neuropeptides/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Orexins
20.
Alcohol Clin Exp Res ; 28(5): 792-6, 2004 May.
Article in English | MEDLINE | ID: mdl-15166656

ABSTRACT

BACKGROUND: Opiate peptides are involved in the physical dependence on ethanol. Levels of methionine enkephalin (MEnk), for example, are affected by ethanol. No study on the effect of ethanol on endomorphin, the endogenous ligand for the mu-opiate receptor, has yet been conducted. METHODS: We examined the effect of ethanol ingestion on serum endomorphin (EM)-1 and MEnk levels. We also determined the effect of antisense directed at MEnk on serum levels of EM-1 and MEnk. RESULTS: Serum EM-1 levels steadily decreased about 20% during 56 days of ethanol ingestion in liquid feed, whereas a similar decrease in serum MEnk levels was not statistically significant. Serum MEnk levels decreased about 20% by 48 hr after antisense injection and then returned to baseline, whereas serum EM-1 levels increased by about 80% and remained elevated for about 2 weeks. In mice not treated with antisense or alcohol, there was no correlation between the serum levels of EM-1 and MEnk. CONCLUSIONS: These results show that serum levels of EM-1 are decreased by physical dependence on ethanol and that this effect is not directly mediated through MEnk.


Subject(s)
Alcoholism/blood , Enkephalin, Methionine/blood , Ethanol/administration & dosage , Oligopeptides/blood , Animals , Mice , Self Administration , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...