Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Exp Neurol ; 269: 56-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25819102

ABSTRACT

Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Programmed death of neuronal cells plays a crucial role in acute and chronic neurodegeneration following TBI. The tumor suppressor protein p53, a transcription factor, has been recognized as an important regulator of apoptotic neuronal death. The p53 inactivator pifithrin-α (PFT-α) has been shown to be neuroprotective against stroke. A previous cellular study indicated that PFT-α oxygen analog (PFT-α (O)) is more stable and active than PFT-α. We aimed to investigate whether inhibition of p53 using PFT-α or PFT-α (O) would be a potential neuroprotective strategy for TBI. To evaluate whether these drugs protect against excitotoxicity in vitro, primary rat cortical cultures were challenged with glutamate (50mM) in the presence or absence of various concentrations of the p53 inhibitors PFT-α or PFT-α (O). Cell viability was estimated by LDH assay. In vivo, adult Sprague Dawley rats were subjected to controlled cortical impact (CCI, with 4m/s velocity, 2mm deformation). Five hours after injury, PFT-α or PFT-α (O) (2mg/kg, i.v.) was administered to animals. Sensory and motor functions were evaluated by behavioral tests at 24h after TBI. The p53-positive neurons were identified by double staining with cell-specific markers. Levels of mRNA encoding for p53-regulated genes (BAX, PUMA, Bcl-2 and p21) were measured by reverse transcription followed by real time-PCR from TBI animals without or with PFT-α/PFT-α (O) treatment. We found that PFT-α(O) (10 µM) enhanced neuronal survival against glutamate-induced cytotoxicity in vitro more effectively than PFT-α (10 µM). In vivo PFT-α (O) treatment enhanced functional recovery and decreased contusion volume at 24h post-injury. Neuroprotection by PFT-α (O) treatment also reduced p53-positive neurons in the cortical contusion region. In addition, p53-regulated PUMA mRNA levels at 8h were significantly reduced by PFT-α (O) administration after TBI. PFT-α (O) treatment also decreased phospho-p53 positive neurons in the cortical contusion region. Our data suggest that PFT-α (O) provided a significant reduction of cortical cell death and protected neurons from glutamate-induced excitotoxicity in vitro, as well as improved neurological functional outcome and reduced brain injury in vivo via anti-apoptotic mechanisms. The inhibition of p53-induced apoptosis by PFT-α (O) provides a useful tool to evaluate reversible apoptotic mechanisms and may develop into a novel therapeutic strategy for TBI.


Subject(s)
Benzothiazoles/pharmacology , Brain Injuries/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Toluene/analogs & derivatives , Animals , Apoptosis/physiology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Survival/drug effects , Disease Models, Animal , Male , Neurons/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Toluene/pharmacology , Treatment Outcome , Tumor Suppressor Protein p53/metabolism
2.
Curr Alzheimer Res ; 9(4): 447-57, 2012 May.
Article in English | MEDLINE | ID: mdl-22272623

ABSTRACT

Activation of inflammatory processes is observed within the brain as well as periphery of subjects with Alzheimer's disease (AD). Whether or not inflammation represents a possible cause of AD or occurs as a consequence of the disease process, or, alternatively, whether the inflammatory response might be beneficial to slow the disease progression remains to be elucidated. The cytokine IL-18 shares with IL-1 the same pro-inflammatory features. Consequent to these similarities, IL-18 and its endogenous inhibitor, IL-18BP, were investigated in the plasma of AD patients versus healthy controls (HC). An imbalance of IL-18 and IL-18BP was observed in AD, with an elevated IL-18/IL-18BP ratio that might be involved in disease pathogenesis. As part of the inflammatory response, altered levels of RANTES, MCP-1 and ICAM- 1, molecules involved in cell recruitment to inflammatory sites, were observed in AD. Hence, correlations between IL-18 and other inflammatory plasma markers were analyzed. A negative correlation was observed between IL-18 and IL-18BP in both AD and HC groups. A positive correlation was observed between IL-18 and ICAM-1 in AD patients, whereas a negative correlation was evident in the HC group. IL-18 positively correlated with Aß in both groups, and no significant correlations were observed between IL-18, RANTES and MCP-1. An important piece of evidence supporting a pathophysiologic role for inflammation in AD is the number of inflammatory mediators that have been found to be differentially regulated in AD patients, and specific ones may provide utility as part of a biomarker panel to not only aid early AD diagnosis, but follow its progression.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Apolipoproteins E/genetics , Cytokines/blood , Inflammation Mediators/blood , Aged , Aged, 80 and over , Chemokine CCL2/blood , Chemokine CCL5/blood , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/genetics , Genotype , Humans , Intercellular Adhesion Molecule-1/blood , Male , RNA, Messenger/metabolism , Statistics, Nonparametric
3.
Neurobiol Aging ; 33(2): 423.e27-36, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21093964

ABSTRACT

Parkinson's disease (PD), an age-related movement disorder, is characterized by severe catecholaminergic neuron loss in the substantia nigra pars compacta (SN(PC))-ventral tegmental area (VTA) and locus coeruleus (LC). To assess the stability of these central catecholaminergic neurons following an acute episode of severe inflammation, 6 to 22 month old C57/Bl6 mice received a maximally tolerated dose of lipopolysaccharide (LPS) followed by euthanasia 2 hours later to assay peak levels of peripheral and central cytokines; and, 14 weeks later for computerized stereology of tyrosine hydroxylase-immunopositive (tyrosine hydroxylase-positive [TH+]) neurons in the SN(PC)-VTA and LC. Two hours after LPS, cytokine levels varied in an age-related manner, with the greatest peripheral and central elevations in old and young mice, respectively. Severe inflammation failed to cause loss of TH+ neurons in SN(PC)-VTA or LC; however, there was an age-related decline in these TH+ neurons in LPS-treated and control groups. Thus, unknown mechanisms in the B6 mouse brain appear to protect against catecholaminergic neuron loss following an acute episode of severe inflammation, while catecholaminergic neuron loss occurs during normal aging.


Subject(s)
Aging/metabolism , Aging/pathology , Encephalitis/metabolism , Lipopolysaccharides , Neuritis/metabolism , Neurons/metabolism , Receptors, Catecholamine/metabolism , Animals , Cell Count , Cell Proliferation , Cell Survival , Cytokines/metabolism , Encephalitis/chemically induced , Encephalitis/pathology , Mice , Mice, Inbred C57BL , Neuritis/chemically induced , Neuritis/pathology
4.
Cell Transplant ; 20(9): 1351-9, 2011.
Article in English | MEDLINE | ID: mdl-21294958

ABSTRACT

A key limiting factor impacting the success of cell transplantation for Parkinson's disease is the survival of the grafted cells, which are often short lived. The focus of this study was to examine a novel strategy to optimize the survival of exogenous fetal ventromesencephalic (VM) grafts by treatment with the p53 inhibitor, pifithrin-α (PFT-α), to improve the biological outcome of parkinsonian animals. Adult male Sprague-Dawley rats were given 6-hydroxydopamine into the left medial forebrain bundle to induce a hemiparkinsonian state. At 7 weeks after lesioning, animals were grafted with fetal VM or cortical tissue into the lesioned striatum and, thereafter, received daily PFT-α or vehicle injections for 5 days. Apomorphine-induced rotational behavior was examined at 2, 6, 9, and 12 weeks after grafting. Analysis of TUNEL or tyrosine hydroxylase (TH) immunostaining was undertaken at 5 days or 4 months after grafting. The transplantation of fetal VM tissue into the lesioned striatum reduced rotational behavior. A further reduction in rotation was apparent in animals receiving PFT-α and VM transplants. By contrast, no significant reduction in rotation was evident in animals receiving cortical grafts or cortical grafts + PFT-α. PFT-α treatment reduced TUNEL labeling and increased TH(+) cell and fiber density in the VM transplants. In conclusion, our data indicate that early postgrafting treatment with PFT-α enhances the survival of dopamine cell transplants and augments behavioral recovery in parkinsonian animals.


Subject(s)
Benzothiazoles/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/transplantation , Parkinson Disease/therapy , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Animals , Behavior, Animal/drug effects , Brain Tissue Transplantation , Cell Count , Cell Survival/drug effects , Dopaminergic Neurons/cytology , In Situ Nick-End Labeling , Male , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Substantia Nigra/pathology , Toluene/pharmacology , Tyrosine 3-Monooxygenase/metabolism
6.
Curr Alzheimer Res ; 7(7): 642-51, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20704560

ABSTRACT

Recent studies of Alzheimer's disease (AD) and other neuropsychiatric drug developments raise questions whether failures of some drugs occur due to flaws in methods. In three case studies of recent AD drug development failures with phenserine, metrifonate, and tarenflurbil we identified methodological lapses able to account for the failures. Errors in complex systems such as drug developments are both almost inescapable due to human mistakes and most frequently hidden at the time of occurrence and thereafter. We propose preemptive error management as a preventive strategy to exclude or control error intrusions into neuropsychiatric drug developments. We illustrate the functions we anticipate for a preemptive error management preventive strategy with a checklist and identify the limitations of this aspect of the proposal with three drug examples. This strategy applies core scientific practices to insure the quality of data within the current context of AD drug development practices.


Subject(s)
Alzheimer Disease/drug therapy , Drug Evaluation/methods , Drug Evaluation/standards , Neuroprotective Agents/pharmacology , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Clinical Trials as Topic/trends , Drug Evaluation/trends , Humans , Neuroprotective Agents/therapeutic use
7.
Mini Rev Med Chem ; 9(10): 1229-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19817713

ABSTRACT

The identification of peripheral biomarkers for neurodegenerative diseases is required to improve the accuracy of clinical diagnosis and monitor both disease progression and response to treatments. The data reviewed in this paper suggest that, in neurodegenerative disease, cytokines are links between peripheral immune system and nervous system dysfunction.


Subject(s)
Alzheimer Disease/immunology , Cytokines/immunology , Parkinson Disease/immunology , Alzheimer Disease/diagnosis , Cytokines/blood , Humans , Parkinson Disease/diagnosis
8.
Curr Alzheimer Res ; 4(4): 370-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17908039

ABSTRACT

We have demonstrated that aged animals show significant improvements in cognitive function and neurogenesis after brain transplantation of human neural stem cells or of human adult mesenchymal stem cells that have been dedifferentiated by transfection of the embryonic stem cell gene. We have also demonstrated that peripheral administration of a pyrimidine derivative increased cognition, endogenous brain stem cell proliferation and neurogenesis. These results indicate a bright future for stem cell therapies in Alzheimer's disease (AD). Before this is realized, however, we need to consider the affect of AD pathology on stem cell biology to establish an effective stem cell therapy for this disease. Although amyloid-beta (Abeta) deposition is a hallmark of AD, an absence of a phenotype in the beta-amyloid precursor protein (APP) knockout mouse, might lead one to underestimate the potential physiological functions of APP and suggest that it is unessential or can be compensated for. We have found, however, that APP is needed for differentiation of neural stem cells (NSCs) in vitro, and that NSCs transplanted into a APP-knockout mouse did not migrate or differentiate -- indicating that APP plays an important role in differentiation or migration process of NSCs in the brain. Then again, treatment with high a concentration of APP or its over-expression increased glial differentiation of NSCs. Human NSCs transplanted into APP-transgenic mouse brain exhibited less neurogenesis and active gliosis around the plaque like formations. Treatment of such animals with the compound, (+)-phenserine, that is known to reduce APP protein levels, increased neurogenesis and suppressed gliosis. These results suggest APP levels can regulate NSC biology in the adult brain, that altered APP metabolism in Down syndrome or AD may have implications for the pathophysiology of these diseases, and that a combination of stem cell therapy and regulation of APP levels could provide a treatment strategy for these disorders.


Subject(s)
Alzheimer Disease/surgery , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Genetic Therapy , Humans
9.
J Neural Transm Suppl ; (71): 237-47, 2006.
Article in English | MEDLINE | ID: mdl-17447434

ABSTRACT

Iron closely regulates the expression of the Alzheimer's Amyloid Precursor Protein (APP) gene at the level of message translation by a pathway similar to iron control of the translation of the ferritin L- and H mRNAs by Iron-responsive Elements in their 5' untranslated regions (5'UTRs). Using transfection based assays in SH-SY5Y neuroblastoma cells we tested the relative efficiency by which iron, copper and zinc up-regulate IRE activity in the APP 5'UTR. Desferrioxamine (high affinity Fe3+ chelator), (ii) clioquinol (low affinity Fe/Cu/Zn chelator), (iii) piperazine-1 (oral Fe chelator), (iv) VK-28 (oral Fe chelator), were tested for their relative modulation of APP 5' UTR directed translation of a luciferase reporter gene. Iron chelation based therapeutic strategies for slowing the progression of Alzheimer's disease (and other neurological disorders that manifest iron imbalance) are discussed with regard to the relative neural toxic action of each chelator in SH-SY5Y cells and in H4 glioblastoma cells.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Iron Chelating Agents/pharmacology , Iron-Regulatory Proteins/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Untranslated Regions/metabolism , Amyloid beta-Protein Precursor/genetics , Cell Line, Tumor , Clioquinol/pharmacology , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Glioblastoma , Humans , Models, Molecular , Neuroblastoma , Transfection/methods , Untranslated Regions/genetics
10.
Chem Biol Interact ; 157-158: 363-5, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16429486

ABSTRACT

Previous studies in rodents and nonhuman primates have demonstrated that pretreatment with cholinesterases can provide significant protection against behavioral and lethal effects of nerve agent intoxication. Human butyrylcholinesterase (HuBuChE) purified from plasma has been shown to protect against up to 5 x LD50s of nerve agents in guinea pigs and non-human primates, and is currently being explored as a bioscavenger pretreatment for human use. A recombinant form of HuBuChE has been expressed in the milk of transgenic goats as a product called Protexia. Protexia was supplied by Nexia Biotechnologies (Que., Canada) as a purified solution with a specific activity of 600 U/mg. Initial in vitro studies using radiolabeled 3H-soman or 3H-DFP (diisopropyl fluorophosphate) demonstrated that these inhibitors specifically bind to Protexia. When Protexia was mixed with soman, sarin, tabun or VX using varying molar ratios of enzyme to nerve agent (8:1, 4:1, 1:1 and 1:4, respectively), the data indicated that 50% inhibition of enzyme activity occurs around the 1:1 molar ratio for each of the nerve agents. Protexia was further characterized for its interaction with pyridostigmine bromide and six unique carbamate inhibitors of cholinesterase. IC50 and Ki values for Protexia were determined to be very similar to those of HuBuChE purified from human plasma. These data suggest that Protexia has biochemical properties very similar to those HuBuChE when compared in vitro. Together these data the continued development of the goat milk-derived recombinant HuBuChE Protexia as a potential bioscavenger of organophosphorus nerve agents.


Subject(s)
Butyrylcholinesterase/pharmacology , Neurons/drug effects , Neurotoxins/antagonists & inhibitors , Animals , Butyrylcholinesterase/chemistry , Carbamates/antagonists & inhibitors , Goats , Humans , Neurons/enzymology , Neurons/pathology , Neurotoxins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
11.
Curr Pharm Des ; 10(25): 3111-9, 2004.
Article in English | MEDLINE | ID: mdl-15544501

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive dementia caused by the loss of the presynaptic markers of the cholinergic system in the brain areas related to memory and learning and brain deposits of amyloid beta peptide (A beta) and neurofibrillary tangles (NFT). A small fraction of early onset familial AD (FAD) is caused by mutations in genes, such as the beta-amyloid precursor protein (APP) and presenilins that increase the load of A beta in the brain. These studies together with findings that A beta is neurotoxic in vitro, provide evidence that some aggregates of this peptide are the key to the pathogenesis of AD. The yield of A beta and the processing and turnover of APP are regulated by a number of pathways including apolipoprotein E, cholesterol and cholinergic agonists. Early studies showed that muscarinic agonists increased APP processing within the A beta sequence (sAPP alpha). More recently, we have presented evidence showing that some, but not all, anticholinesterases reduce secretion of sAPP alpha as well as A beta into the media suggesting that cholinergic agonists modulate A beta levels by multiple mechanisms. Herein we review the recent advances in understanding the function of cholinesterase (ChE) in the brain and the use of ChE-inhibitors in AD. We propose and support the position that the influence of cholinergic stimulation on amyloid formation is critical in light of the early targeting of the cholinergic basal forebrain in AD and the possibility that maintenance of this cholinergic tone might slow amyloid deposition. In this context, the dual action of certain cholinesterase inhibitors on their ability to increase acetylcholine levels and decrease amyloid burden assumes significance as it may identify a single drug to both arrest the progression of the disease as well as treat its symptoms. A new generation of acetyl- and butyryl cholinesterase inhibitors is being studied and tested in human clinical trials for AD. We critically discuss recent trends in AD research, from molecular and genetic to clinical areas, as it relates to the effects of cholinergic agents and their secondary effects on A beta. Finally, we examine different neurobiological mechanisms that provide the basis of new targets for AD drug development.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Technology, Pharmaceutical/methods , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Drug Design , Humans , Technology, Pharmaceutical/trends
12.
J Alzheimers Dis ; 5(2): 81-90, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12719626

ABSTRACT

One of the major neuropathological characteristics of Alzheimer's disease (AD) is the brain depositions of senile plaques that are mainly composed of toxic amyloid beta-peptide (Abeta), which is generated from a family of Abeta containing precursor proteins (AbetaPP; 695-770 amino acids). The role of cytokines and growth factors has been implicated in the pathogenesis of AD. Our goal is to determine the mode of action of cytokines on the regulation of betaPP gene expression. Here we studied the effect of different cytokines on the activity of 5'-untranslated region (5'-UTR) of betaPP mRNA in human astrocytic cells (U-373). We compared betaPP-5'-UTR activity in the presence of interleukin-1 (IL-1alpha and IL-1beta), transforming growth factor (TGF-beta1) and tumor necrosis factor TNF-alpha1. The astrocytic cells, which were treated separately with these agents, were transfected with either the vector (pSV2CAT) or pSV2UTR-CAT construct containing 90 bp of AbetaPP 5'-UTR +54 to 144 bp). This region was cloned upstream of a reporter chloramphenicol acetyl transferase gene (CAT). Our results indicate that the treatment of pSV2UTR-CAT-transfected cells with either IL-1alpha, IL-1beta, TGF-beta1 or TNF-alpha1 stimulated reporter gene activity in a factor-specific manner. This was consistent with their effects on elevating AbetaPP protein levels. Transfection of the same cells with the pSV2CAT vector lacking 5'-UTR resulted in a reduced reporter gene activity with all treatments studied. DNA-gel shift experiments indicate that the 54/144 region binds to a nuclear protein(s) in a cell type specific manner. These results suggest that 5'-UTR of the AbetaPP gene can respond to the stimulation of different cytokines, which likely regulate AbetaPP transcription and translation via regulatory elements present in the AbetaPP promoter and in 5'-UTR, respectively. The characterization of AbetaPP regulatory elements, including the 5'-UTR, will accelerate the development of novel agents against new targets for AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/metabolism , Cytokines/pharmacology , Gene Expression Regulation , 5' Untranslated Regions , Adrenal Gland Neoplasms/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytoma/pathology , Brain Neoplasms/pathology , Chloramphenicol O-Acetyltransferase/genetics , Cloning, Molecular , Genes, Reporter , Humans , Pheochromocytoma/pathology , Rats , Transcriptional Activation , Transfection , Tumor Cells, Cultured
13.
Ann N Y Acad Sci ; 1010: 639-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033804

ABSTRACT

Oxidative stress is a risk factor for Alzheimer's disease (AD) whose major hallmark includes brain depositions of the amyloid beta peptide (Abeta) derived from the beta-amyloid precursor protein (APP). Our aim was to determine whether or not excessive Abeta deposition would alter nitric oxide synthase (NOS) activity, and thereby affect NOS-mediated superoxide formation. We compared NOS activity in brain extracts between Tg mice (expressing APP Swedish double mutation plus presenilin [PS-1] and nontransgenic [nTg] mice. Five brain regions, including cerebral cortex, hippocampus, cerebellum, and striatum from both nTg and Tg mice showed a detectable level of neuronal (n) NOS activity. Cerebellar extracts from both nTg and Tg mice displayed the highest level of nNOS activity, which was fourfold higher than cortical extracts. Although there was an increase in nNOS activity in Tg brain extracts, this did not attain statistical significance. A similar result was obtained for inducible NOS levels. Our results suggest that excess levels of Abeta failed to both trigger NOS activity and change NOS levels.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Nitric Oxide Synthase/genetics , Animals , Brain/enzymology , Humans , Mice , Mice, Transgenic , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Oxidative Stress , Reference Values
14.
Ann N Y Acad Sci ; 965: 364-72, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12105112

ABSTRACT

Alzheimer's disease (AD) is characterized by cerebrovascular deposition of the amyloid beta-peptide (A beta), which is derived from a larger beta-amyloid precursor protein (beta APP). Altered metabolism of beta APP, resulting in increased A beta production, appears central in the neuropathology of AD. The processing of the holoprotein beta APP by different "secretase" enzymes results in three major carboxyl-truncated species. One species, which results from the cleavage of beta APP by gamma-secretase, is secreted into the cerebrospinal fluid (CSF) and is called sAPP gamma as it contains an intact A beta domain. Moreover, AD is characterized by cholinergic dysfunction and the loss of synaptic proteins. Reports of an inverse relation between nicotine intake, due to cigarette smoking, and the incidence of AD prompted us to investigate the effects of nicotine on beta APP processing and synaptic proteins in rats and in cell culture. Nicotine, 1 and 8 mg/kg/day, doses commensurate with cigarette smoking, and a higher but well tolerated dose, respectively, was administered over 14 days to rats. Levels of sAPP in the CSF sample were evaluated by Western blot analysis. The higher dose significantly increased levels of total sAPP; however, both doses significantly reduced sAPP gamma, which contains the amyloidogenic portion of A beta. These actions were blocked by nicotinic receptor antagonism. Nicotinic antagonists alone had no effect on either total sAPP or sAPP gamma levels in CSF. Nicotine did not significantly change the intracellular levels of total beta APP in rat brain extracts, which is consistent with neuronal cell culture data. Similarly, levels of vesicular protein, such as synaptophysin, and presynaptic terminal protein SNAP-25 were unaffected by nicotine treatment both in vivo and in cell culture experiments. Taken together, these results suggest that nicotine modifies beta APP processing away from the formation of potentially amyloidogenic products, without altering the levels of synaptic proteins, and that this can potentially offer therapeutic potential for Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/metabolism , Nicotine/pharmacology , Synapses/physiology , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/drug effects , Animals , Disease Models, Animal , Humans , Male , Nerve Tissue Proteins/drug effects , Rats , Rats, Inbred F344 , Synapses/drug effects
15.
J Med Chem ; 44(24): 4062-71, 2001 Nov 22.
Article in English | MEDLINE | ID: mdl-11708910

ABSTRACT

With the goal of developing potential Alzheimer's pharmacotherapeutics, we have synthesized a series of novel analogues of the potent anticholinesterases phenserine (2) and physostigmine (1). These derivatives contain methyl (3, 4, 6), dimethyl (5, 7, 8, 10, 11) and trimethyl (14) substituents in each position of the phenyl group of the phenylcarbamoyl moieties, and with N-methyl and 6-methyl substituents (12, 13, 31, 33). We also quantified the inhibitory action of these compounds against human acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). An analysis of the structure/anticholinesterase activity relationship of the described compounds, together with molecular modeling, confirmed the catalytic triad mechanism of the binding of this class of carabamate analogues within AChE and BChE and defined structural requirements for their differential inhibition.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Carbamates/chemical synthesis , Cholinesterase Inhibitors/chemical synthesis , Physostigmine/analogs & derivatives , Physostigmine/chemical synthesis , Carbamates/chemistry , Carbamates/pharmacology , Cholinesterase Inhibitors/chemistry , Crystallography, X-Ray , Erythrocytes/chemistry , Humans , Models, Molecular , Molecular Conformation , Physostigmine/chemistry , Structure-Activity Relationship
16.
Endocrinology ; 142(10): 4462-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564711

ABSTRACT

The use of glucagon-like peptide-1 (GLP-1) as a routine treatment for type 2 diabetes mellitus is undermined by its short biological half-life. A cause of degradation is its cleavage at the N-terminal HAE sequence by the enzyme dipeptidyl peptidase IV (DPP IV). To protect from DPP IV, we have studied the biological activity of a GLP-1 analog in which 6-aminohexanoic acid (Aha) is inserted between histidine and alanine at positions 7 and 8. We have compared the biological activity of this new compound, GLP-1 Aha(8), with the previously described GLP-1 8-glycine (GLP-1 Gly(8)) analog. GLP-1 Aha(8) (10 nM) was equipotent with GLP-1 (10 nM) in stimulating insulin secretion in RIN 1046-38 cells. As with GLP-1 Gly(8), the binding affinity of GLP-1 Aha(8) for the GLP-1 receptor in intact Chinese hamster ovary (CHO) cells expressing the human GLP-1 receptor (CHO/GLP-1R cells) was reduced (IC(50): GLP-1, 3.7 +/- 0.2 nM; GLP-1 Gly(8), 41 +/- 9 nM; GLP-1 Aha(8), 22 +/- 7 nM). GLP-1 Aha(8) was also shown to stimulate intracellular cAMP production 4-fold above basal at concentrations as low as 0.5 nM. However, it exhibited a higher ED(50) when compared to GLP-1 and GLP-1 Gly(8) (ED(50): GLP-1, 0.036 +/- 0.002 nM, GLP-1 Gly(8), 0.13 +/- 0.02 nM, GLP-1 Aha(8), 0.58 +/- 0.03 nM). A series of D-amino acid-substituted GLP-1 compounds were also examined to assess the importance of putative peptidase-sensitive cleavage sites present in the GLP-1 molecule. They had poor binding affinity for the GLP-1 receptor, and none of these compounds stimulated the production of intracellular cAMP in CHO/GLP-1R cells or insulin secretion in RIN 1046-38 cells. GLP-1 Aha(8) (24 nmol/kg) administered sc to fasted Zucker (fa/fa) rats (mean blood glucose, 195 +/- 32 mg/dl) lowered blood glucose levels to a nadir of 109 +/- 3 mg/dl, and it remained significantly lower for 8 h. Matrix-assisted linear desorption ionization-time of flight mass spectrometry of GLP-1 Aha(8) incubated with DPP IV (37 C, 2 h) did not exhibit an N-terminal degradation product. Taken together, these results show that insertion of Aha after the 7 position in GLP-1 produces an effective, long-acting GLP-1 analog, which may be useful in the treatment of type 2 diabetes mellitus.


Subject(s)
Glucagon/pharmacology , Hypoglycemic Agents/pharmacology , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Amino Acid Sequence , Aminocaproic Acid , Animals , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucagon/genetics , Glucagon/therapeutic use , Glucagon-Like Peptide 1 , Humans , Hypoglycemic Agents/therapeutic use , Molecular Sequence Data , Mutagenesis, Insertional , Peptide Fragments/genetics , Peptide Fragments/therapeutic use , Protein Precursors/genetics , Protein Precursors/therapeutic use , Rats , Structure-Activity Relationship
17.
Proc Natl Acad Sci U S A ; 98(13): 7605-10, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11404470

ABSTRACT

The reduction in levels of the potentially toxic amyloid-beta peptide (Abeta) has emerged as one of the most important therapeutic goals in Alzheimer's disease. Key targets for this goal are factors that affect the expression and processing of the Abeta precursor protein (betaAPP). Earlier reports from our laboratory have shown that a novel cholinesterase inhibitor, phenserine, reduces betaAPP levels in vivo. Herein, we studied the mechanism of phenserine's actions to define the regulatory elements in betaAPP processing. Phenserine treatment resulted in decreased secretion of soluble betaAPP and Abeta into the conditioned media of human neuroblastoma cells without cellular toxicity. The regulation of betaAPP protein expression by phenserine was posttranscriptional as it suppressed betaAPP protein expression without altering betaAPP mRNA levels. However, phenserine's action was neither mediated through classical receptor signaling pathways, involving extracellular signal-regulated kinase or phosphatidylinositol 3-kinase activation, nor was it associated with the anticholinesterase activity of the drug. Furthermore, phenserine reduced expression of a chloramphenicol acetyltransferase reporter fused to the 5'-mRNA leader sequence of betaAPP without altering expression of a control chloramphenicol acetyltransferase reporter. These studies suggest that phenserine reduces Abeta levels by regulating betaAPP translation via the recently described iron regulatory element in the 5'-untranslated region of betaAPP mRNA, which has been shown previously to be up-regulated in the presence of interleukin-1. This study identifies an approach for the regulation of betaAPP expression that can result in a substantial reduction in the level of Abeta.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cholinesterase Inhibitors/pharmacology , Interleukin-1/pharmacology , Physostigmine/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Astrocytoma , Cell Survival/drug effects , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/genetics , Chromones/pharmacology , Culture Media, Conditioned , Drug Design , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Humans , Interleukin-1/physiology , L-Lactate Dehydrogenase/analysis , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Neuroblastoma , Phosphatidylinositol 3-Kinases/metabolism , Physostigmine/analogs & derivatives , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
18.
J Neurochem ; 77(1): 220-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11279278

ABSTRACT

The tumor suppressor protein p53 is essential for neuronal death in several experimental settings and may participate in human neurodegenerative disorders. Based upon recent studies characterizing chemical inhibitors of p53 in preclinical studies in the cancer therapy field, we synthesized the compound pifithrin-alpha and evaluated its potential neuroprotective properties in experimental models relevant to the pathogenesis of stroke and neurodegenerative disorders. Pifithrin-alpha protected neurons against apoptosis induced by DNA-damaging agents, amyloid beta-peptide and glutamate. Protection by pifithrin-alpha was correlated with decreased p53 DNA-binding activity, decreased expression of the p53 target gene BAX and suppression of mitochondrial dysfunction and caspase activation. Mice given pifithrin-alpha exhibited increased resistance of cortical and striatal neurons to focal ischemic injury and of hippocampal neurons to excitotoxic damage. These preclinical studies demonstrate the efficacy of a p53 inhibitor in models of stroke and neurodegenerative disorders, and suggest that drugs that inhibit p53 may reduce the extent of brain damage in related human neurodegenerative conditions.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain Ischemia/drug therapy , Neurons/drug effects , Seizures/drug therapy , Thiazoles/pharmacology , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzothiazoles , Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Caspase 3 , Caspases/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Glutamic Acid/pharmacology , Kainic Acid , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Curr Med Res Opin ; 17(3): 159-65, 2001.
Article in English | MEDLINE | ID: mdl-11900310

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the elderly, characterised by widespread loss of central cholinergic function. The only symptomatic treatment proven effective to date is the use of cholinesterase (ChE) inhibitors to augment surviving cholinergic activity. ChE inhibitors act on the enzymes that hydrolyse acetylcholine (ACh) following synaptic release. In the healthy brain, acetylcholinesterase (AChE) predominates (80%) and butyrylcholinesterase (BuChE) is considered to play a minor role in regulating brain ACh levels. In the AD brain, BuChE activity rises while AChE activity remains unchanged or declines. Therefore both enzymes are likely to have involvement in regulating ACh levels and represent legitimate therapeutic targets to ameliorate the cholinergic deficit. The two enzymes differ in location, substrate specificity and kinetics. Recent evidence suggests that BuChE may also have a role in the aetiology and progression of AD beyond regulation of synaptic ACh levels. Experimental evidence from the use of agents with enhanced selectivity for BuChE (cymserine, MF-8622) and ChE inhibitors such as rivastigmine, which have a dual inhibitory action on both AChE and BuChE, indicate potential therapeutic benefits of inhibiting both AChE and BuChE in AD and related dementias. The development of specific BuChE inhibitors and the continued use of ChE inhibitors with the ability to inhibit BuChE in addition to AChE should lead to improved clinical outcomes.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/therapeutic use , Humans
20.
Pharmacol Biochem Behav ; 66(3): 475-81, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10899358

ABSTRACT

In imaging studies of brain functions using pharmacological probes, identification of the time point at which central effects of intravenously infused drugs become stable is crucial to separate the effects of experimental variables from the concomitant changes in drug effects over time. We evaluated the time courses of the pharmacokinetics and pharmacodynamics, including butyrylcholinesterase inhibition and central neural responses, of physostigmine in healthy young subjects. Ten positron emission tomography (PET) scans that alternated between a rest condition (eyes open, ears unplugged) and a working memory for faces (WM) task were acquired in healthy subjects. Subjects in the drug group received a saline infusion for the first two scans, providing a baseline measure, then received an infusion of physostigmine for all subsequent scans. Subjects in the control group received a placebo infusion of saline for all scans. Physostigmine plasma levels and percent butyrylcholinesterase inhibition increased over time (p < 0. 0001), and both became stable by 40 min. Physostigmine decreased reaction time (RT) (p = 0.0005), and this effect was detected after 20 min of infusion and stable thereafter. Physostigmine also decreased regional cerebral blood flow (rCBF) in right prefrontal cortex during task (p = 0.0002), and this effect was detected after 40 min of infusion and stable thereafter. No change in RT or rCBF was observed in the control group. These results indicate that a 40-min infusion of physostigmine was necessary to obtain stable central effects. More generally, we have demonstrated that experimental effects can vary with time, especially during the initial phases of a drug infusion, indicating that it is critical that these changes are controlled.


Subject(s)
Brain/metabolism , Physostigmine/pharmacokinetics , Adult , Analysis of Variance , Brain/diagnostic imaging , Brain/drug effects , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Cognition/drug effects , Female , Humans , Male , Middle Aged , Physostigmine/pharmacology , Psychomotor Performance/drug effects , Time Factors , Tomography, Emission-Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...