Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Neuroscience ; 259: 94-100, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24316473

ABSTRACT

Despite the enormous public health impact of Alzheimer's disease (AD), no disease-modifying treatment has yet been proven to be efficacious in humans. A rate-limiting step in the discovery of potential therapies for humans is the absence of efficient non-invasive methods of evaluating drugs in animal models of disease. Magnetic resonance spectroscopy (MRS) provides a non-invasive way to evaluate the animals at baseline, at the end of treatment, and serially to better understand treatment effects. In this study, MRS was assessed as potential outcome measure for detecting disease modification in a transgenic mouse model of AD. Passive immunization with two different antibodies, which have been previously shown to reduce plaque accumulation in transgenic AD mice, was used as intervention. Treatment effects were detected by MRS, and the most striking finding was attenuation of myo-inositol (mIns) increases in APP-PS1 mice with both treatments. Additionally, a dose-dependent effect was observed with one of the treatments for mIns. MRS appears to be a valid in vivo measure of anti-Aß therapeutic efficacy in pre-clinical studies. Because it is noninvasive, and can detect treatment effects, use of MRS-based endpoints could substantially accelerate drug discovery.


Subject(s)
Alzheimer Disease , Aspartic Acid/analogs & derivatives , Brain/metabolism , Immunization, Passive/methods , Magnetic Resonance Spectroscopy , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid/metabolism , Choline , Disease Models, Animal , Humans , Inositol , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Presenilin-1/genetics , Statistics, Nonparametric
2.
Neuroimage ; 54(1): 113-22, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20728546

ABSTRACT

Our laboratory and others have reported the ability to detect individual Alzheimer's disease (AD) amyloid plaques in transgenic mouse brain in vivo by magnetic resonance imaging (MRI). Since amyloid plaques contain iron, most MRI studies attempting to detect plaques in AD transgenic mouse brain have employed techniques that exploit the paramagnetic effect of iron and have had mixed results. In the present study, using five-way anatomic spatial coregistration of MR images with three different histological techniques, properties of amyloid plaques in AD transgenic mouse brain were revealed that may explain their variable visibility in gradient- and spin-echo MR images. The results demonstrate differences in the visibility of plaques in the cortex and hippocampus, compared to plaques in the thalamus, by the different MRI sequences. All plaques were equally detectable by T(2)SE, while only thalamic plaques were reliably detectable by T(2)*GE pulse sequences. Histology revealed that cortical/hippocampal plaques have low levels of iron while thalamic plaques have very high levels. However, the paramagnetic effect of iron does not appear to be the sole factor leading to the rapid decay of transverse magnetization (short T(2)) in cortical/hippocampal plaques. Accordingly, MRI methods that rely less on iron magnetic susceptibility effect may be more successful for eventual human AD plaque MR imaging, particularly since human AD plaques more closely resemble the cortical and hippocampal plaques of AD transgenic mice than thalamic plaques.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Cerebral Cortex/anatomy & histology , Cerebral Cortex/pathology , Hippocampus/anatomy & histology , Hippocampus/pathology , Humans , Iron/metabolism , Magnetic Resonance Imaging/methods , Mice , Mice, Transgenic , Organ Specificity , Thalamus/anatomy & histology , Thalamus/pathology
3.
Neuroreport ; 12(15): 3197-200, 2001 Oct 29.
Article in English | MEDLINE | ID: mdl-11711855

ABSTRACT

Much research is now focused on a potential vaccine for Alzheimer's disease (AD). Current studies involve administering the amyloid beta peptide (Abeta) in Freund's complete adjuvant, which cannot be used in humans. Our studies show that the immune complex of Abeta is taken up by a receptor-mediated process at the blood-brain barrier (BBB). The success of immunization for AD, therefore, may be critically dependent on circulating Abeta levels which are lower in AD patients compared to AD transgenic mice. Moreover, we have found that modifying the antibody with polyamine increases its BBB permeability and may provide a better approach to passive immunization for Alzheimer's disease.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/pharmacokinetics , Blood-Brain Barrier/drug effects , Carrier Proteins/drug effects , Immunization/methods , Peptide Fragments/pharmacokinetics , Receptors, Peptide/drug effects , Vaccines/pharmacokinetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/pharmacology , Blood-Brain Barrier/immunology , Brain/drug effects , Brain/immunology , Brain/metabolism , Carrier Proteins/immunology , Dose-Response Relationship, Drug , Humans , Iodine Radioisotopes/pharmacokinetics , Mice , Peptide Fragments/chemistry , Peptide Fragments/immunology , Polyamines/chemistry , Polyamines/pharmacokinetics , Receptors, Peptide/immunology , Vaccines/chemical synthesis
4.
Neurobiol Dis ; 8(4): 555-67, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11493021

ABSTRACT

The permeability of albumin, insulin, and human A beta 1--40 at the blood-brain barrier (BBB) was determined in the normal adult mouse (B6/SJL) and in the double transgenic Alzheimer mouse (APP, PS1) by using an I.V. bolus injection technique to quantify the permeability coefficient-surface area (PS) product for each protein after correction for the residual plasma volume (V(p)) occupied by the protein in the blood vessels of different brain regions using a second aliquot of the same protein radiolabeled with a different isotope of iodine ((125)I vs (131)I) as a vascular space marker. This technology for quantifying BBB permeability of proteins was adapted from the rat to the mouse and involved catheterizing the femoral artery and vein of the mouse instead of the brachial artery and vein as for the rat. Because of the smaller blood volume in the mouse, serial sampling (20 microl) of blood from the femoral artery of the mouse was performed and directly TCA precipitated to generate a whole blood washout curve for the intact protein. When similar blood sampling techniques were used in the rat, the PS values for albumin and insulin at the BBB were similar in these two species. In the double transgenic mouse, the V(p) values for albumin were significantly increased 1.4- to 1.6-fold in five of six brain regions compared to the normal adult mouse, which indicated increased adherence of albumin to vessel walls. As a result, the PS values were significantly decreased, from 1.4- to 3.2-fold, which likely reflected decreased transport of albumin by passive diffusion. In contrast, insulin, which is taken up into the brain by a receptor-mediated transport mechanism at the BBB, showed no significant difference in the V(p) values but a significant increase in the PS values in four of six brain regions. This suggests a compensatory mechanism in the Alzheimer's transgenic brain whereby there is an increased permeability to insulin at the BBB. Surprisingly, there was no significant difference in the V(p) or PS values for human A beta 1--40 at the BBB in the double transgenic Alzheimer mouse at 24, 32, or 52 weeks of age, when there is both significant A beta levels in the plasma and amyloid burden in the brains of these animals. These data suggest that there is not an alteration in permeability to human A beta 1--40 at the BBB with increasing amyloid burden in the double transgenic Alzheimer mouse. Although these observations suggest structural alterations at the BBB, they do not support the concept of extensive BBB damage with substantial increases in BBB permeability in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Blood-Brain Barrier/physiology , Membrane Proteins/genetics , Serum Albumin/pharmacokinetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacokinetics , Animals , Disease Models, Animal , Hypoglycemic Agents/pharmacokinetics , Insulin/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/pharmacokinetics , Presenilin-1 , Rats , Rats, Sprague-Dawley , Species Specificity
5.
Ann Neurol ; 48(6): 943-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11117554

ABSTRACT

Continuous subcutaneous administration of polyamine-modified catalase that has increased permeability at the blood-brain barrier showed both a highly significant delay in onset and an increase in survival in a transgenic mouse model of familial amyotrophic lateral sclerosis having a point mutation in the gene encoding copper/zinc superoxide dismutase. These results suggest that hydrogen peroxide-mediated oxidative stress with subsequent free radical damage involving nitric oxide and possibly hydroxyl radicals in motor neurons may be the culprit in familial amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Catalase/physiology , Hydrogen Peroxide/metabolism , Nitric Oxide/metabolism , Putrescine/pharmacology , Amyotrophic Lateral Sclerosis/genetics , Animals , Blood-Brain Barrier , Catalase/metabolism , Mice , Mice, Transgenic , Putrescine/analogs & derivatives , Survival Analysis
6.
Neuroscience ; 101(4): 939-44, 2000.
Article in English | MEDLINE | ID: mdl-11113343

ABSTRACT

The development of transgenic mice has created new opportunities for the generation of animal models of human neurodegenerative diseases where previously there was no animal counterpart. The first successful transgenic mouse model of Alzheimer's disease expressed increased levels of mutant human amyloid precursor protein, exhibiting neuritic-type amyloid deposits and behavioral deficits at six to nine months of age. More recently, it was shown that transgenic mice expressing both mutant human amyloid precursor protein and presenilin 1 exhibit neuritic-type amyloid deposits and behavioral deficits in as little as 12 weeks. This accelerated Alzheimer phenotype greatly reduces the time necessary to conduct preclinical drug trials, as well as animal housing costs. The purpose of this study was to quantify the deposition of amyloid in five regions of the cortex and two regions of the hippocampus of transgenic mice expressing amyloid precursor protein (K670N, M671L) and presenilin 1 (M146L) mutations at various ages, using quantitative methods of confocal laser scanning microscopy and image analysis. Amyloid burden, expressed as the percentage area occupied by thioflavin S-positive amyloid deposits, increased an average of 179-fold from 12 to 54 weeks of age (0.02+/-0.01% to 3.57+/-0.29%, mean+/-S.E.M., respectively) in five regions of the cortex and two of the hippocampus. This was a function of increases in both deposit number and size. This transgenic mouse provides an ideal animal model for evaluating the efficacy of potential therapeutic agents aimed at reducing amyloid deposition, such as inhibitors of amyloid fibril formation or secretase inhibitors.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Brain/metabolism , Brain/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Image Processing, Computer-Assisted , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Microscopy, Confocal , Presenilin-1
7.
Nat Biotechnol ; 18(8): 868-72, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10932157

ABSTRACT

The only definitive diagnosis for Alzheimer disease (AD) at present is postmortem observation of neuritic plaques and neurofibrillary tangles in brain sections. Radiolabeled amyloid-beta peptide (Abeta), which has been shown to label neuritic plaques in vitro, therefore could provide a diagnostic tool if it also labels neuritic plaques in vivo following intravenous injection. In this study, we show that the permeability of Abeta at the blood-brain barrier can be increased by at least twofold through covalent modification with the naturally occurring polyamine, putrescine. We also show that, following intravenous injection, radiolabeled, putrescine-modified Abeta labels amyloid deposits in vivo in a transgenic mouse model of AD, as well as in vitro in human AD brain sections. This technology, when applied to humans, may be used to detect plaques in vivo, allowing early diagnosis of the disease and therapeutic intervention before cognitive decline occurs.


Subject(s)
Alzheimer Disease/diagnosis , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Humans , Male , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Rats , Rats, Sprague-Dawley
8.
Diabetes ; 49(7): 1219-23, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10909981

ABSTRACT

Leptin exerts important effects on the regulation of food intake and energy expenditure by acting in the brain. Leptin is secreted by adipocytes into the bloodstream and must gain access to specific regions in the brain involved in regulating energy balance. Its action is mediated by interaction with a receptor that is mainly expressed in the hypothalamus but is also present in other cerebral areas. To reach these target areas, leptin most likely needs to cross the blood-brain barrier (BBB). In this study, we compared the permeability of leptin at the BBB in homozygous lean (FA/FA), high-fat diet-induced (HFD) obese rats (FA/FA rats on a highfat diet), and genetically obese fa/fa Zucker rats by quantifying the permeability coefficient surface area (PS) product after correction for the residual plasma volume (Vp) occupied by leptin in the vessel bed of different brain regions. The intravenous bolus injection technique was used in the cannulated brachial vein and artery using leptin radioiodinated with 2 isotopes of iodine (125I and 131I) to separately determine the PS and Vp values. The PS for leptin at the BBB in lean FA/FA rats ranged from 11.0 +/- 1.6 at the cortex to 14.8 +/- 1.4 x 10(-6) ml x g(-1) x ml(-1) at the posterior hypothalamus. The PS for leptin in HFD obese FA/FA and obese fa/fa rats ranged from 3.0- to 4.0-fold lower than in lean FA/FA rats. The Vp values were not significantly different among the 3 groups studied. SDS-PAGE analysis of the radioiodinated leptin after 60 min of uptake revealed intact protein in the 8 different brain regions. Plasma leptin levels were significantly higher in both obese rat groups compared with those in lean FA/FA rats. Leptin levels in cerebrospinal fluid were not significantly different among the 3 groups of rats. These findings strongly suggest that the leptin receptor (OB-R) in the BBB can be easily saturated. Saturation of the BBB OB-R in obese individuals would explain the defect in leptin transport into the brain described in this study.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Leptin/metabolism , Obesity/physiopathology , Animals , Body Weight , Dietary Fats , Homozygote , Iodine Radioisotopes , Leptin/blood , Leptin/cerebrospinal fluid , Obesity/blood , Obesity/genetics , Organ Specificity , Radioisotope Dilution Technique , Rats , Rats, Zucker , Thinness
9.
Exp Neurol ; 159(1): 191-203, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10486187

ABSTRACT

Free radical-mediated oxidative damage has been proposed to be an underlying mechanism in several neurodegenerative disorders. Previous investigations in our laboratory have shown that putrescine-modified catalase (PUT-CAT) has increased permeability at the blood-brain (BBB) and blood-nerve barriers with retained enzymatic activity after parenteral administration when compared to native catalase (CAT). The goals of the present study were to examine the plasma stability, spinal cord BBB permeability, nervous system biodistribution, and spinal cord enzyme activity of CAT and PUT-CAT after parenteral administration in the adult rat. TCA precipitation and chromatographic analyses revealed that CAT and PUT-CAT were found intact in the plasma and in the central nervous system (CNS) after iv, ip, or sc bolus injections. The highest percentages of intact CAT or PUT-CAT proteins were found in the plasma after iv administration, and similar percentages of intact CAT or PUT-CAT were found in the CNS following all three types of administration. Increases of 2.4- to 4.7-fold in permeability at the BBB and similar increases in the levels of intact PUT-CAT were found in different brain regions compared to the levels of CAT. A 2.4-fold higher level of intact PUT-CAT compared to that of CAT (P < 0.05) was found in the spinal cord 60 min after a sc bolus injection. CAT enzyme activity in the spinal cord was 50% higher (P < 0.05) in rats treated with PUT-CAT continuously for 1 week by subcutaneously implanted, osmotic pumps than the activity found in rats treated with PBS. These results provide evidence that intact, enzymatically active PUT-CAT is efficiently delivered to the nervous system following iv, ip, and sc administration and suggest that sc administration of PUT-CAT may be effective in treating neurodegenerative disorders in which the underlying mechanisms involve the action of free radicals and oxidative damage.


Subject(s)
Blood-Brain Barrier , Catalase/pharmacokinetics , Putrescine/pharmacokinetics , Age Factors , Animals , Antioxidants/pharmacokinetics , Catalase/blood , Central Nervous System/blood supply , Central Nervous System/enzymology , Injections, Intraperitoneal , Injections, Intravenous , Injections, Subcutaneous , Iodine Radioisotopes/pharmacokinetics , Nerve Degeneration/drug therapy , Putrescine/analogs & derivatives , Putrescine/blood , Rats , Rats, Sprague-Dawley , Tissue Distribution
10.
Exp Neurol ; 159(1): 204-16, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10486188

ABSTRACT

Dominant mutations in the copper/zinc superoxide dismutase (SOD1) gene have been observed in 15-20% of familial amyotrophic lateral sclerosis (FALS) cases. The mechanism by which SOD1 mutations result in motor neuron degeneration in FALS mice partly involves oxidative damage and an increased peroxidase activity of the mutant SOD1. A new therapeutic approach designed to eliminate the substrate of this peroxidase activity was examined in two lines of transgenic mice expressing the FALS-linked mutation glycine to alanine (G93A). We investigated the ability of putrescine-modified catalase (PUT-CAT), an antioxidant enzyme that removes hydrogen peroxide and has increased permeability at the blood-brain barrier, to modify the time course of the SOD1 mutation-induced motor neuron disease in these FALS mice. Continuous, subcutaneous administration of PUT-CAT significantly delayed the age at which onset of clinical disease occurred (indicated by loss of splay and/or tremors of hindlimbs) in a high-expressor line of FALS transgenic mice. Intraperitoneal injection of PUT-CAT given two times per week also significantly delayed the onset of clinical disease in a low-expressor line of FALS mice. PUT-CAT also significantly delayed the age at which clinical weakness developed (quantified by measuring the shortening of stride length) in both lines of FALS animals. No significant changes were observed in the survival times of the high-expressor FALS mice in any of the treatment groups. However, a trend toward a prolongation of survival was observed in the PUT-CAT-treated low-expressor FALS mice. These results support the role of free radical-mediated damage in the cascade of events leading to motor neurodegeneration in FALS and indicate that PUT-CAT interacts with a critical step in this cascade to delay the onset of clinical disease as well as the development of clinical weakness in FALS transgenic mice.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Catalase/pharmacology , Putrescine/pharmacology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Animals , Antioxidants/pharmacology , Blood-Brain Barrier , Buffers , Catalase/metabolism , Disease Models, Animal , Female , Free Radicals/metabolism , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/enzymology , Muscle Weakness/drug therapy , Muscle Weakness/genetics , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Phosphates , Putrescine/analogs & derivatives , Spinal Cord/cytology , Superoxide Dismutase/genetics , Survival Analysis
11.
Neurobiol Dis ; 6(3): 190-9, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10408808

ABSTRACT

Since amyloid beta-protein (A beta) is the primary component of both vascular and parenchymal amyloid deposits in Alzheimer's disease, information regarding its permeability at the blood-brain barrier (BBB) will help elucidate the contribution of circulating A beta to vascular and parenchymal A beta deposition in this disease and in brain aging. The permeability of the D- and L-enantiomers of A beta 1-40 and L-A beta 1-42 at the BBB was determined in the normal adult rat by quantifying the permeability coefficient-surface area product (PS) for each protein after correction for the residual plasma volume (Vp) occupied by the protein [labeled with a different isotope of iodine (125I vs 131I)] in blood vessels of different brain regions. After a single i.v. bolus injection, the plasma pharmacokinetics determined by TCA precipitation, paper chromatography, and SDS-PAGE were similar for both 125I-L-A beta 1-40 and 125I-L-A beta 1-42. The PS at the BBB for L-A beta 1-42 was significantly (1.4- to 1.8-fold) higher than for L-A beta 1-40 and ranged from 17.7 to 26.4 x 10(-6) ml/g/s for different brain regions. A comparison of the PS values at the BBB for L-A beta 1-40 showed no significant difference when determined at 15 or 30 min after i.v. bolus injection, times that reflect different levels of degradation in plasma (37.9% at 15 min and 65.5% at 30 min). The PS values obtained, therefore, were representative of the intact protein rather than degradation products. The PS values obtained for the all-D-enantiomer of A beta 1-40 were very low and comparable to that of albumin and IgG, whose mechanism of transport is by passive diffusion. Taken together, these data imply a stereoisomer-specific, ligand-receptor interaction at the BBB for the L-A beta proteins. The high PS values observed for L-A beta 1-40 and 1-42 compare to insulin, whose uptake is decidedly by a receptor-mediated transport process, and suggest a similar mechanism for L-A beta entry into the brain.


Subject(s)
Amyloid beta-Peptides/pharmacokinetics , Blood-Brain Barrier/physiology , Peptide Fragments/pharmacokinetics , Age Factors , Amyloid beta-Peptides/blood , Animals , Cerebrovascular Circulation , Humans , Injections, Intravenous , Iodine Radioisotopes , Peptide Fragments/blood , Radioimmunoassay , Rats , Rats, Sprague-Dawley
12.
J Neurobiol ; 39(3): 371-82, 1999 Jun 05.
Article in English | MEDLINE | ID: mdl-10363910

ABSTRACT

Short synthetic peptides homologous to the central region of Abeta but bearing proline residues as beta-sheet blockers have been shown in vitro to bind to Abeta with high affinity, partially inhibit Abeta fibrillogenesis, and redissolve preformed fibrils. While short peptides have been used extensively as therapeutic drugs in medicine, two important problems associated with their use in central nervous system diseases have to be addressed: (a) rapid proteolytic degradation in plasma, and (b) poor blood-brain barrier (BBB) permeability. Recently, we have demonstrated that the covalent modification of proteins with the naturally occurring polyamines significantly increases their permeability at the BBB. We have extended this technology to iAbeta11, an 11-residue beta-sheet breaker peptide that inhibits Abeta fibrillogenesis, by covalently modifying this peptide with the polyamine, putrescine (PUT), and evaluating its plasma pharmacokinetics and BBB permeability. After a single intravenous bolus injection in rats, both 125I-YiAbeta11 and 125I-PUT-YiAbeta11 showed rapid degradation in plasma as determined by trichloroacetic acid (TCA) precipitation and paper chromatography. By switching to the all D-enantiomers of YiAbeta11 and PUT-YiAbeta11, significant protection from degradation by proteases in rat plasma was obtained with only 1.9% and 5.7% degradation at 15 min after intravenous bolus injection, respectively. The permeability coefficient x surface area product at the BBB was five- sevenfold higher in the cortex and hippocampus for the 125I-PUT-D-YiAbeta11 compared to the 125I-D-YiAbeta11, with no significant difference in the residual plasma volume. In vitro assays showed that PUT-D-YiAbeta11 retains its ability to partially inhibit Abeta fibrillogenesis and dissolve preformed amyloid fibrils. Because of its five- to sevenfold increase in permeability at the BBB and its resistance to proteolysis in the plasma, this polyamine-modified beta-sheet breaker peptide may prove to be an effective inhibitor of amyloidogenesis in vivo and, hence, an important therapy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/antagonists & inhibitors , Blood-Brain Barrier/physiology , Peptides/chemistry , Peptides/pharmacology , Protein Structure, Secondary/drug effects , Amyloid/biosynthesis , Animals , Capillary Permeability/physiology , Drug Resistance , Male , Peptide Hydrolases/blood , Peptide Hydrolases/metabolism , Peptides/blood , Peptides/metabolism , Rats , Rats, Sprague-Dawley
13.
J Neurochem ; 71(4): 1651-60, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751199

ABSTRACT

Previous investigations from our laboratory have demonstrated that the covalent modification of a variety of proteins, including antioxidant enzymes, with the naturally occurring polyamines--putrescine (PUT), spermidine, and spermine--dramatically increases their permeability coefficient-surface area product (PS) at the blood-brain and blood-nerve barriers after parenteral administration. In the present study, we have covalently modified nerve growth factor (NGF) with PUT by targeting carboxylic groups for their graded modification by controlling the ionization of these groups with pH. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis, western, and isoelectric focusing analyses demonstrated conversion of NGF to its polyamine-modified derivatives at different pH values. Although the immunoreactivity of PUT-NGF determined by ELISA and western analysis decreased with decreasing pH, the biological activity of PUT-NGF was not affected at any pH as determined by survival and neurite extension of dorsal root ganglia and PC12 cultures. Plasma pharmacokinetics after a single intravenous bolus administration revealed intact PUT-NGF through 10 min and 73-82% intact protein at 15 min. The PS value for PUT-NGF was maximized and the residual plasma volume (Vp) of the protein in the blood vessels minimized when the pH of the modification reaction was >6.4. The biodistribution of PUT-NGF at 15 min showed 22-33% intact protein in different brain regions, which represented 0.4-5.9 ng of PUT-NGF in different brain regions, a physiological dose that is capable of eliciting a bioresponse. The design of this polyamine-modified NGF derivative that has enhanced permeability at the blood-brain and blood-nerve barriers with retained bioactivity may obviate the necessity to create small-molecule mimics of NGF and may be applicable to neurotrophins, engineered multifunctional chimeric neurotrophins, antioxidant enzymes, and other therapeutic proteins with specific clinical application to neurological diseases.


Subject(s)
Blood-Brain Barrier , Cell Membrane Permeability , Central Nervous System/metabolism , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacokinetics , Putrescine/pharmacology , Animals , Blood-Brain Barrier/drug effects , Cell Membrane Permeability/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Hydrogen-Ion Concentration , Injections, Intravenous , Male , Nerve Growth Factors/blood , Neurites/drug effects , Neurites/physiology , Organ Specificity , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 273(15): 9070-7, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9535895

ABSTRACT

Recent studies demonstrate that cAMP levels are tightly controlled during demyelination and remyelination in Schwann cells as cAMP decreases to 8-10% of normal following both sciatic nerve crush or permanent transection injury and only begins to increase in the crushed nerve after remyelination (Poduslo, J. F., Walikonis, R. S., Domec, M., Berg, C. T., and Holtz-Heppelmann, C. J. (1995) J. Neurochem. 65, 149-159). To investigate the mechanisms responsible for this change in cAMP levels, cAMP phosphodiesterase (PDE) and adenylyl cyclase activities were determined before and after sciatic nerve injury. Basal cAMP PDE activity in soluble endoneurial homogenates of normal nerve was 34.9 +/- 1.9 pmol/mg of protein/min (chi +/- S.E.; n = 10). This activity increased about 3-fold within 6 days following both injuries. Basal PDE activity remained elevated in the transected nerve, but declined to 70 pmol/mg of protein/min in the crushed nerve at 21 and 35 days following injury. Isozyme-specific inhibitors and stimulators were used to identify the PDE families in the sciatic nerve. The low Km cAMP-specific (PDE4) and the Ca2+/calmodulin-stimulated (PDE1) families were found to predominate in assays using endoneurial homogenates. The PDE4 inhibitor rolipram also increased cAMP levels significantly after incubation of endoneurial tissue with various isozyme-specific inhibitors, indicating that PDE4 plays a major role in determining cAMP levels. PDE4 mRNA was localized by in situ hybridization to cells identified as Schwann cells by colabeling of S100, a Schwann cell specific protein. Adenylyl cyclase activity declined following injury, from 3.7 pmol/mg of protein/min in normal nerve to 0.70 pmol/mg/min by 7 days following injury. Both decreased synthesis and increased degradation contribute, therefore, to the reduced levels of cAMP following peripheral nerve injury and are likely critical to the process of Wallerian degeneration.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , 3',5'-Cyclic-GMP Phosphodiesterases/biosynthesis , Adenylyl Cyclases/biosynthesis , Myelin Sheath/physiology , Nerve Crush , Nerve Regeneration , Phosphoric Diester Hydrolases , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Transcription, Genetic/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Gene Expression Regulation, Enzymologic/drug effects , Male , Pyrrolidinones/pharmacology , Quinolones/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reference Values , Rolipram , S100 Proteins/biosynthesis , Time Factors , Xanthines/pharmacology
15.
Brain Res ; 767(1): 128-35, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9365024

ABSTRACT

Much evidence exists in support of the hypothesis that free radicals contribute to the pathogenesis of several neurodegenerative disorders and that mechanisms of free radical generation occur both intracellularly and extracellularly. Previous studies in this laboratory have shown that covalent modification of growth factors and antioxidant enzymes with the naturally occurring polyamine, putrescine, increases their permeability at the blood-nerve and blood-brain barriers (BNB and BBB), but does not significantly inhibit bioactivity. Furthermore, putrescine-modified superoxide dismutase (SOD) was shown to reduce neurodegeneration in a rat model of global cerebral ischemia. The purpose of the present study was to modify the antioxidant enzyme, catalase (CAT), with putrescine (PUT) at carboxylic acid groups whose ionization, and hence reactivity, was controlled with pH and investigate the effects on permeability and enzymatic activity. Modification of CAT with PUT increased its permeability 2-3-fold and preserved 67% of its enzymatic activity compared to native CAT and 137% compared to lyophilized CAT. The results of this study indicate that modification of CAT with putrescine increases its permeability while preserving enzymatic activity. PUT-SOD administered in combination with PUT-CAT may eliminate both the superoxide radical and the H2O2 produced from the dismutation of superoxide, respectively, and thus prevent the formation of hydroxyl radicals. This combination may exhibit increased neuroprotective effects, compared to native enzymes, following systemic administration for the treatment of free radical associated neurodegenerative disorders.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Catalase/therapeutic use , Neuroprotective Agents/therapeutic use , Putrescine/therapeutic use , Superoxide Dismutase/therapeutic use , Animals , Hydrogen-Ion Concentration , Male , Rats , Rats, Sprague-Dawley
16.
Brain Res ; 754(1-2): 46-54, 1997 Apr 18.
Article in English | MEDLINE | ID: mdl-9134958

ABSTRACT

Antioxidant enzymes such as superoxide dismutase (SOD) have shown neuroprotective effects in animal models of cerebral ischemia, but only at very high doses. Modifications to increase the plasma half-life or blood-brain barrier (BBB) permeability of SOD have resulted in limited neuroprotective effects. No one has demonstrated neuroprotection with postischemic administration. The specific aim of the present study was to administer systemically a polyamine-modified SOD, having increased BBB permeability and preserved enzymatic activity, following global cerebral ischemia in rats and analyze the effects on the selective vulnerability of CA1 hippocampal neurons. Following 12 min of four-vessel occlusion, global cerebral ischemia, male Wistar rats were dosed (i.v.) with either saline, native SOD (5000 U/kg), polyamine-modified SOD (5000 U/kg), or enzymatically inactive, polyamine-modified SOD (2.1 mg/kg) twice daily for 3 days. Neuroprotective effects on hippocampal CA1 neurons were assessed using standard histological methods. Saline-treated animals had very few remaining CA1 neurons (1.44 +/- 0.60 neurons/reticle; x +/- S.E.M.) compared to sham rats (58.57 +/- 0.69). Native (10.38 +/- 2.96) or inactive, polyamine-modified SOD (7.32 +/- 2.68) did not show significant neuroprotective effects. Polyamine-modified SOD, however, resulted in the survival of significantly more CA1 neurons (24.61 +/- 5.90; P < 0.01). Postischemic, systemic administration of polyamine-modified SOD, having increased BBB permeability and preserved enzymatic activity, significantly reduced hippocampal CA1 neuron loss following global cerebral ischemia. Similar modification of other antioxidant enzymes and neurotrophic factors with polyamines may provide a useful technique for the systemic delivery of therapeutic proteins across the BBB for the treatment of stroke and other neurodegenerative disorders.


Subject(s)
Hippocampus/physiopathology , Ischemic Attack, Transient/physiopathology , Nerve Degeneration/drug effects , Neuroprotective Agents , Polyamines , Pyramidal Cells/physiology , Superoxide Dismutase/therapeutic use , Animals , Blood-Brain Barrier , Half-Life , Hippocampus/drug effects , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Male , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Superoxide Dismutase/pharmacokinetics
17.
Neurobiol Dis ; 4(1): 27-34, 1997.
Article in English | MEDLINE | ID: mdl-9258909

ABSTRACT

The permeability of normal human, the human Dutch variant, and the rat A beta 1-40 proteins at the blood-brain barrier (BBB) was determined in the normal adult rat by quantifying the permeability coefficient-surface area (PS) product for each protein after correction for the residual plasma volume (Vp) occupied by the protein in the blood vessels of different brain regions. The PS for normal and Dutch A beta ranged from 13 x 10(-6) to 22 x 10(-6) ml/g/s in different brain regions, which is 130 to 220 times greater than albumin. These high PS values compare to that of insulin, whose uptake is decidedly by a receptor-mediated transport process, and suggest a similar mechanism for A beta. Remarkably, the PS for rat A beta was 4 times higher and ranged from 54 x 10(-6) to 82 x 10(-6) ml/g/s for different brain regions, suggesting a distinctive species specificity. While the Vp values of human and rat A beta were comparable, the Dutch variant was 2 to 3 times higher, indicating adherence to the vessel walls in different brain regions, consistent with the heavy A beta deposition that has been described in intracerebral vessel walls with this variant. The high PS values observed for A beta at the BBB suggest that sources outside the nervous system could contribute, at least in part, to the cerebral A beta deposits seen in Alzheimer's disease. SDS-PAGE of 125I-labeled human A beta after 60 min of uptake revealed intact protein in plasma and in different brain regions. In addition, 125I-labeled human A beta binding to a protein of 67,000 in both plasma and brain tissue regions was observed with SDS-PAGE. This protein was tentatively identified as albumin, and it was not detectable in the brain regions of animals that had undergone intracardiac perfusion; hence, a portion of A beta binds tightly to and is likely transported by albumin in plasma. The absence of this A beta-albumin complex in brain regions after perfusion and the low permeability of albumin at the BBB imply that A beta itself is efficiently transported at the BBB to account for the high PS values, although presentation of A beta to the capillary endothelial cell by albumin or other plasma proteins cannot be excluded.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier , Brain/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/genetics , Animals , Capillary Permeability , Genetic Variation , Half-Life , Humans , Insulin/blood , Insulin/metabolism , Iodine Radioisotopes , Molecular Sequence Data , Peptide Fragments/blood , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Plasma Volume , Rats , Rats, Sprague-Dawley , Tissue Distribution
18.
J Neurochem ; 67(2): 734-41, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8764602

ABSTRACT

Our previous studies have demonstrated that modification of superoxide dismutase (SOD) with the naturally occurring polyamines--putrescine (PUT), spermidine, and spermine--dramatically increases the permeability-coefficient surface area (PS) product at the blood-brain barrier and blood-nerve barrier after parenteral administration. Because of this increased permeability, the efficient delivery of polyamine-modified SOD (pSOD) across these barriers may enhance its therapeutic usefulness in treating ischemic neuronal degeneration, neurodegenerative disease, or even aging as an important antioxidant therapeutic strategy. Because PUT-SOD had the highest PS values, SOD was modified in the present experiments by activating carboxylic acid groups to the reactive ester with water-soluble carbodiimide and then reacted with PUT as the nucleophilic reagent. Preservation of SOD enzyme activity while maximizing the permeability was accomplished by adjusting the ionization of the protein carboxylic acid with pH. Both sodium dodecyl sulfate-polyacrylamide gel electrophoresis and isoelectric focusing analyses demonstrated graded conversion of SOD to its polyamine-modified derivative when performed at different pH. Although modification at pH 4.7 resulted in only 6.6% retained SOD activity and the highest PS value (43.35 +/- 3.81 x 10(-6) ml/g/s for the hippocampus), modification at pH 5.7 resulted in 50.1 % retained activity with a PS value of 24.48 1.30 x 10(-6) ml/g/s for nerve endoneurium and 21.95 +/- 1.62 x 10(-6) ml/g/s for hippocampus. This contrasts with a PS of 1.8-3.2 x 10(-6) ml/g/s for native SOD in nerve and various brain regions. Reaction conditions are therefore defined that titrate enzyme activity of PUT-SOD with PS changes in the intact animal after intravenous administration. These studies will allow an evaluation of the therapeutic usefulness of pSOD in animal models of neuronal degeneration.


Subject(s)
Blood-Brain Barrier , Putrescine/metabolism , Superoxide Dismutase/metabolism , Animals , Hydrogen-Ion Concentration , Isoelectric Point , Male , Metabolic Clearance Rate , Nerve Fibers/metabolism , Putrescine/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Superoxide Dismutase/chemistry
19.
J Neurochem ; 66(4): 1599-609, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8627316

ABSTRACT

The permeability of the blood-nerve barrier (BNB) and the blood-brain barrier (BBB) to superoxide dismutase (SOD), insulin, albumin, and IgG in normal adult rats was quantified by measuring the permeability coefficient-surface area product (PS) with the intravenous bolus injection technique before and after covalent protein modification with naturally occurring polyamines-putrescine (PUT), spermidine (SPD), and spermine (SPM). The PS value of the BNB for PUT-SOD was 21.1-fold greater than the native SOD, and the PS values of the BBB for PUT-SOD ranged from 17.6-fold greater for the thalamus to 23.6-fold greater for the caudate-putamen compared with native SOD. In a similar manner, polyamine-modified insulin showed a 1.7-2.0-fold increase in PS of the BNB and BBB compared with the high values of native insulin. Polyamine-modified albumin showed a remarkable 54-165-fold increase in PS of the BNB and BBB compared with native albumin, whereas PUT-IgG resulted in an even higher increase in the PS that ranged from 111- to 349-fold for nerve and different brain regions compared with native IgG. Polyamine modification of proteins, therefore, can dramatically increase the permeability at the BNB and BBB of a variety of proteins with widely differing M(r) and function. It is surprising that the PS values of the BNB and BBB decreased with the increasing number of positive charges of the protonated amino groups on the polyamines (PUT>SPD>SPM). Although cationic proteins are known to interact with fixed anionic charges on the lumen of the microvascular endothelium, this observation of decreased permeability with increased positive charge distribution along the aliphatic carbon chain of the polyamines implies mechanisms other than simple electrostatic interaction involving charge density. It is suggested that the polyamine transporter may be responsible for the transport of these polyamine-modified proteins. Systemic administration of polyamine-modified peptides and proteins might prove to be an efficient approach to deliver therapeutic agents into the CNS and PNS for the treatment of a variety of neurological diseases.


Subject(s)
Blood-Brain Barrier/physiology , Neurons/metabolism , Polyamines/metabolism , Animals , Cations/analysis , Cations/metabolism , Cell Membrane Permeability/physiology , Electric Conductivity , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/metabolism , Immunoglobulin G/analysis , Immunoglobulin G/blood , Injections, Intravenous , Insulin/analysis , Insulin/blood , Iodine Radioisotopes , Isoelectric Focusing , Male , Peptides/metabolism , Polyamines/analysis , Polyamines/pharmacokinetics , Proteins/metabolism , Putrescine/analysis , Putrescine/blood , Rats , Rats, Sprague-Dawley , Serum Albumin/analysis , Spermidine/analysis , Spermidine/blood , Superoxide Dismutase/analysis , Superoxide Dismutase/blood
20.
Brain Res Mol Brain Res ; 36(2): 280-6, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8965648

ABSTRACT

A comparison was made of the permeabilities of different neurotrophic factors at the blood-brain barrier (BBB) and blood-nerve barrier (BNB) in normal adult rats by quantifying the permeability coefficient-surface area (PS) product after correction for the residual plasma volume (Vp) occupied by the protein in the capillary bed of the nerve endoneurium or different brain regions. The i.v. bolus injection technique was used in the cannulated brachial vein and artery using the same protein radioiodinated with a second isotope of iodine (125I vs. 131I) to separately determine the PS and Vp values. The plasma washout showed a decreasing plasma half-life in the order of brain-derived neurotrophic factor (BDNF) < neurotrophin-3 (NT-3) < ciliary neurotrophic factor (CNTF) < nerve growth factor (NGF). The PS at the BNB for NGF was 1.40 +/- 0.15 x 10(-6) ml/g/s (mean +/- SEM). The other neurotrophic proteins were all significantly higher than NGF (CNTF: 9.5 x ; NT-3: 20.8 x ; BDNF: 18.9 x ). The Vp for NGF at the BNB was 1.92 +/- 0.12 microliters/g and was not significantly different from the other proteins except for NGF vs. BDNF (P < 0.05). The PS for NGF at the BBB ranged from 1.5 to 2.7 x 10(-6) ml/g/s for six different brain regions. The PS for CNTF ranged from 6.0 to 8.0-fold higher than NGF; NT-3: 10.6 to 15.2-fold higher; and BDNF: 11.3 to 16.4-fold higher. The Vp values were not significantly different except for CNTF in the hippocampus and cortex (P < 0.05). SDS-PAGE analyses of all the radioiodinated neurotrophic proteins after 60 min of uptake revealed intact protein in the endoneurium and in the six different brain regions with exposure times of 2-42 days. The quantification of the permeability of these neurotrophic proteins provides baseline values for comparison of different protein modifications that enhance the PS while still preserving the neurotrophic activity (e.g., protein glycation; Poduslo and Curran, Mol. Brain Res., 23 (1994) 157). Enhanced permeability following modification might allow the use of systematic delivery of these proteins for practical therapeutic treatment of various neurodegenerative disorders.


Subject(s)
Blood-Brain Barrier/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Permeability/drug effects , Animals , Autoradiography , Ciliary Neurotrophic Factor , Male , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...