Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Discov Today Technol ; 23: 45-52, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28647085

ABSTRACT

In vivo phenotypic screening and drug repositioning are strategies developed as alternatives to underperforming hypothesis-driven molecular target based drug discovery efforts. This article reviews examples of drugs identified by phenotypic observations and describes the use of the theraTRACE®in vivo screening platform for finding and developing new indications for discontinued clinical compounds. Clinical proof-of-concept for the platform is exemplified by MLR-1023, a repositioned compound that has recently shown significant clinical efficacy in Type 2 diabetes patients. These findings validate an in vivo screening approach for drug development and underscore the importance of alternatives to target and mechanism based strategies that have failed to produce adequate numbers of new medicines.


Subject(s)
Drug Discovery , Diabetes Mellitus, Type 2/drug therapy , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Pyrimidinones/therapeutic use
2.
Adv Exp Med Biol ; 632: 293-307, 2008.
Article in English | MEDLINE | ID: mdl-19025130

ABSTRACT

To obtain proteins with the complement-depleting activity of Cobra Venom Factor (CVF), but with less immunogenicity, we have prepared human C3/CVF hybrid proteins, in which the C-terminus of the alpha-chain of human C3 is exchanged with homologous regions of the C-terminus of the beta-chain of CVF. We show that these hybrid proteins are able to deplete complement, both in vitro and in vivo. One hybrid protein, HC3-1496, is shown to be effective in reducing complement-mediated damage in two disease models in mice, collagen-induced arthritis and myocardial ischemia/reperfusion injury. Human C3/CVF hybrid proteins represent a novel class ofbiologicals as potential therapeutic agents in many diseases where complement is involved in the pathogenesis.


Subject(s)
Complement C3/chemistry , Complement C3/metabolism , Complement C3/therapeutic use , Complement System Proteins/metabolism , Animals , Arthritis, Experimental/drug therapy , Complement C3/genetics , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Humans , Mice , Myocardial Reperfusion Injury/drug therapy , Protein Engineering , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
3.
Arch Neurol ; 64(4): 541-4, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17420316

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is an environmental risk factor for developing Alzheimer disease. This may be due, in part, to changes associated with beta-amyloid (Abeta) plaque formation, which can occur within hours after injury, regardless of the patient's age. In addition to being precursors of toxic fibrils that deposit into plaques, soluble (nonfibrillar) Abeta peptides are posited to disrupt synaptic function and are associated with cognitive decline in Alzheimer disease. Changes in soluble Abeta levels and their relationship to Abeta plaque formation following TBI are unknown. OBJECTIVE: To quantify brain tissue levels of soluble Abeta peptides and their precursor protein in relation to Abeta plaque formation after TBI in humans. DESIGN: Surgically resected temporal cortex tissue from patients with severe TBI was processed for biochemical assays of soluble Abeta peptides with COOH-termini ending in amino acid 40 (Abeta(40)) or 42 (Abeta(42)) and Abeta precursor protein to compare patients with cortical Abeta plaques and those without. Patients Nineteen subjects admitted to the University of Pittsburgh Medical Center for treatment of severe closed head injury. RESULTS: Patients with severe TBI and cortical plaques had higher levels of soluble Abeta(1-42) but not Abeta(1-40); half of them were apolipoprotein E (APOE) epsilon4 allele carriers. The lowest Abeta levels were in 1 patient without plaques who was the only subject with an APOE epsilon2 allele. beta-Amyloid precursor protein levels were comparable in the 2 TBI groups. CONCLUSIONS: Selective increases in soluble Abeta(1-42) after TBI may predispose individuals with a brain injury to Alzheimer disease pathology. This may be influenced by the APOE genotype, and it may confer increased risk for developing Alzheimer disease later in life.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Adult , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/metabolism , Apolipoprotein E4/genetics , Blotting, Western , Brain Injuries/genetics , Brain Injuries/pathology , Cerebral Cortex/pathology , Female , Heterozygote , Humans , Male , Middle Aged , Peptide Fragments/metabolism , Solubility
4.
Neurobiol Aging ; 27(3): 377-86, 2006 Mar.
Article in English | MEDLINE | ID: mdl-15927307

ABSTRACT

Three mouse models of Alzheimer's disease (AD) were used to assess changes in gene expression potentially critical to amyloid beta-peptide (Abeta)-induced neuronal dysfunction. One mouse model harbored homozygous familial AD (FAD) knock-in mutations in both, amyloid precursor protein (APP) and presenilin 1 (PS-1) genes (APP(NLh/NLh)/PS-1(P264L/P264L)), the other two models harbored APP over-expression of FAD mutations (Tg2576) with the PS-1 knock-in mutation at either one or two alleles. These mouse models of AD had varying levels of Abeta40 and Abeta42 and different latencies and rates of Abeta deposition in brain. To assess changes in gene expression associated with Abeta accumulation, the Affymetrix murine genome array U74A was used to survey gene expression in the cortex of these three models both prior to and following Abeta deposition. Altered genes were identified by comparing the AD models with age-matched control littermates. Thirty-four gene changes were identified in common among the three models in mice with Abeta deposition. Among the up-regulated genes, three major classes were identified that encoded for proteins involved in immune responses, carbohydrate metabolism, and proteolysis. Down-regulated genes of note included pituitary adenylate cyclase-activating peptide (PACAP), brain-derived neurotrophic factor (BDNF), and insulin-like growth factor I receptor (IGF-IR). In young mice without detectable Abeta deposition, there were no regulated genes common among the three models, although 40 genes were similarly altered between the two Tg2576 models with the PS-1 FAD knock-in. Finally, changes in gene expression among the three mouse models of AD were compared with those reported in human AD samples. Sixty-nine up-regulated and 147 down-regulated genes were found in common with human AD brain. These comparisons across different genetic mouse models of AD and human AD brain provide greater support for the involvement of identified gene expression changes in the neuronal dysfunction and cognitive deficits accompanying amyloid deposition in mammalian brain.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cerebral Cortex/metabolism , Disease Models, Animal , Transcription Factors/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Female , Gene Expression Profiling , Humans , Mice , Nerve Tissue Proteins/metabolism , Species Specificity
5.
Exp Neurol ; 197(2): 437-50, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16300758

ABSTRACT

The detrimental effects of traumatic brain injury (TBI) on brain tissue integrity involve progressive axonal damage, necrotic cell loss, and both acute and delayed apoptotic neuronal death due to activation of caspases. Post-injury accumulation of amyloid precursor protein (APP) and its toxic metabolite amyloid-beta peptide (Abeta) has been implicated in apoptosis as well as in increasing the risk for developing Alzheimer's disease (AD) after TBI. Activated caspases proteolyze APP and are associated with increased Abeta production after neuronal injury. Conversely, Abeta and related APP/Abeta fragments stimulate caspase activation, creating a potential vicious cycle of secondary injury after TBI. Blockade of caspase activation after brain injury suppresses apoptosis and improves neurological outcome, but it is not known whether such intervention also prevents increases in Abeta levels in vivo. The present study examined the effect of caspase inhibition on post-injury levels of soluble Abeta, APP, activated caspase-3, and caspase-cleaved APP in the hippocampus of nontransgenic mice expressing human Abeta, subjected to controlled cortical injury (CCI). CCI produced brain tissue damage with cell loss and elevated levels of activated caspase-3, Abeta(1-42) and Abeta(1-40), APP, and caspase-cleaved APP fragments in hippocampal neurons and axons. Post-CCI intervention with intracerebroventricular injection of 100 nM Boc-Asp(OMe)-CH(2)F (BAF, a pan-caspase inhibitor) significantly reduced caspase-3 activation and improved histological outcome, suppressed increases in Abeta and caspase-cleaved APP, but showed no significant effect on overall APP levels in the hippocampus after CCI. These data demonstrate that after TBI, caspase inhibition can suppress elevations in Abeta. The extent to which Abeta suppression contributes to improved outcome following inhibition of caspases after TBI is unclear, but such intervention may be a valuable therapeutic strategy for preventing the long-term evolution of Abeta-mediated pathology in TBI patients who are at risk for developing AD later in life.


Subject(s)
Amyloid beta-Peptides/metabolism , Aspartic Acid/analogs & derivatives , Brain Injuries/metabolism , Caspase Inhibitors , Peptide Fragments/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid/pharmacology , Blotting, Western/methods , Brain Injuries/chemically induced , Caspase 3 , Caspases/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Functional Laterality , Hippocampus/metabolism , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/physiology , Stilbamidines/metabolism , Time Factors
6.
Eur J Pharmacol ; 515(1-3): 179-87, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15904918

ABSTRACT

CEP-11004, a mixed lineage kinase (MLK) inhibitor, was examined for its effects on tumor necrosis factor-alpha (TNF-alpha) production in human THP-1 monocytes, mouse BV-2 microglia, and C57Bl/6 mice. CEP-11004 inhibited TNF-alpha secretion up to 90% in THP-1 cells incubated with 3 mug/ml lipopolysaccharide, with an IC50 of 137+/-14 nM. CEP-11004 also inhibited TNF-alpha production in lipopolysaccharide-stimulated microglial cells, but did not inhibit the initial increase in TNF-alpha mRNA expression as measured by real-time polymerase chain reaction (PCR). The mitogen-activated protein kinases (MAPKs) phospho-c-jun N-terminal kinase (JNK), phospho-p38, and phospho-MAPK kinase 4 (MKK4) levels were increased in THP-1 cells following lipopolysaccharide treatment, and were reduced by CEP-11004 treatment. For in vivo studies, CEP-11004 was injected 2 h prior to lipopolysaccharide (20 mg/kg) administration. CEP-11004 significantly inhibited TNF-alpha production at doses of 1-10 mg/kg as measured by enzyme-linked immunosorbent assay (ELISA). These results suggest that MLK blockade may be useful in inhibiting pro-inflammatory cytokine production in a wide range of diseases.


Subject(s)
Carbazoles/pharmacology , Indoles/pharmacology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 8/metabolism , Monocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cell Survival/drug effects , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-6/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Monocytes/cytology , Monocytes/metabolism , Nitric Oxide/biosynthesis , Phagocytosis/drug effects , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Exp Neurol ; 190(1): 192-203, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15473992

ABSTRACT

Traumatic brain injury (TBI) is a risk factor for the development of Alzheimer's disease (AD). This immunohistochemical study determined the extent of AD-related changes in temporal cortex resected from individuals treated surgically for severe TBI. Antisera generated against Abeta species (total Abeta, Abeta(1-42), and Abeta(1-40)), the C-terminal of the Abeta precursor protein (APP), apolipoprotein E (apoE), and markers of neuron structure and degeneration (tau, ubiquitin, alpha-, beta-, and gamma-synuclein) were used to examine the extent of Abeta plaque deposition and neurodegenerative changes in 18 TBI subjects (ages 18-64 years). Diffuse cortical Abeta deposits were observed in one third of subjects (aged 35-62 years) as early as 2 h after injury, with only one (35-year old) individual exhibiting "mature", dense-cored plaques. Plaque-like deposits, neurons, glia, and axonal changes were also immunostained with APP and apoE antibodies. In plaque-positive cases, the only statistically significant change in cellular immunostaining was increased neuronal APP (P = 0.013). There was no significant correlation between the distribution of Abeta plaques and markers of neuronal degeneration. Diffuse tau immunostaining was localized to neuronal cell soma, axons or glial cells in a larger subset of individuals. Tau-positive, neurofibrillary tangle (NFT)-like changes were detected in only two subjects, both of more advanced age and who were without Abeta deposits. Other neurodegenerative changes, evidenced by ubiquitin- and synuclein-immunoreactive neurons, were abundant in the majority of cases. Our results demonstrate a differential distribution and course of intra- and extra-cellular AD-like changes during the acute phase following severe TBI in humans. Abeta plaques and early evidence of neuronal degenerative changes can develop rapidly after TBI, while fully developed NFTs most likely result from more chronic disease- or injury-related processes. These observations lend further support to the hypothesis that head trauma significantly increases the risk of developing pathological and clinical symptoms of AD, and provide insight into the molecular mechanisms that initiate these pathological cascades very early during severe brain injury.


Subject(s)
Alzheimer Disease/pathology , Brain Injuries/pathology , Temporal Lobe/pathology , Adolescent , Adult , Alzheimer Disease/complications , Amyloid beta-Peptides/analysis , Amyloid beta-Protein Precursor/analysis , Apolipoproteins E/analysis , Biomarkers/analysis , Brain Injuries/complications , Brain Injuries/surgery , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neuropil/pathology , Peptide Fragments/analysis , Plaque, Amyloid/pathology , Risk , Temporal Lobe/chemistry
8.
J Neurosci ; 22(9): 3376-85, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11978814

ABSTRACT

The mechanisms by which neurons and synapses are lost in Alzheimer's disease (AD) are not completely understood. To characterize potential signaling events linked to AD pathogenesis, activation-specific antibodies were used to examine mitogen-activated protein kinase (MAPK) kinase pathways at various ages in mice transgenic for human amyloid precursor protein-695 with the Swedish familial AD mutations (Tg2576) and homozygous for a P264L familial AD mutation introduced by targeting of the presenilin-1 gene (PS1(P264L)). Although the c-Jun N-terminal kinase (JNK) and p38 pathways were significantly activated in the cortex at both 7 and 12 months of age, there was no significant activation of the extracellular signal-regulated kinase pathway. MAPK kinase-4, an upstream activator of JNK, was also significantly activated at 7 and 12 months, whereas c-Jun, a downstream effector of JNK-associated apoptotic signaling, was not induced. The lack of c-Jun activation is consistent with the absence of neuronal loss in both cortex and hippocampal CA1 at 12 months. The JNK activation was localized to amyloid deposits, within neurites containing phosphorylated tau. Synaptophysin was quantified biochemically as a measure of synaptic integrity and was significantly reduced in an age-dependent manner in the Tg2576/PS1(P264L) cortex but not in either PS1(P264L) or Tg2576 cortex. Stress-responsive MAP kinase pathways were activated in the brain of the Tg2576/PS1(P264L) AD model, and this activation was coincident with the age-dependent increase in amyloid deposition, tau phosphorylation, and loss of synaptophysin.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amino Acid Substitution , Amyloid beta-Protein Precursor/genetics , Animals , Cell Count , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Enzyme Activation , Gene Targeting , Hippocampus/pathology , Humans , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neurites/enzymology , Neurites/pathology , Neurons/pathology , Phosphorylation , Presenilin-1 , Synaptophysin/deficiency , Synaptophysin/metabolism , p38 Mitogen-Activated Protein Kinases , tau Proteins/metabolism
9.
J Neurotrauma ; 19(12): 1555-67, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12542857

ABSTRACT

There is increasing evidence linking neurodegenerative mechanisms in Alzheimer's disease (AD) and traumatic brain injury (TBI), including increased production of amyloid precursor protein (APP), and amyloid-beta (Abeta) peptide. In vitro data indicate that expression of APP may be regulated in part by the inflammatory cytokine IL-1beta. To further investigate the mechanisms involved, we measured APP and IL-1beta protein levels and examined immunohistochemical localization of APP in brain tissue from rats subjected to controlled cortical impact (CCI) injury. Animals were examined at time intervals ranging from 3 h to 4 weeks after TBI. The 24-h time point revealed a dramatic increase in APP immunoreactivity, detected with both N- and C-terminal antibodies, in the hippocampus and cortex ipsilateral to injury. This finding was sustained up to 3 days post-injury. At these early time points, APP increase was particularly robust in the white matter axonal tracts. By 14 days after injury, APP immunoreactivity was not significantly different from sham controls in cortex, but remained slightly elevated in hippocampus. Western blot data corroborated early increases in hippocampal and cortical APP in injured versus control animals. Despite profound APP changes, no Abeta deposits were observed at any time after injury. Hippocampal and cortical IL-1beta increases were even more robust, with IL-1beta levels peaking by 6 h post-injury and returning to baseline by 24-72 h. Our results demonstrate that both APP and IL-1beta are rapidly elevated after injury. Because of the rapidity in the IL-1beta peak increase, it may serve a role in regulation of APP expression after TBI.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Injuries/metabolism , Interleukin-1/metabolism , Animals , Blotting, Western , Disease Models, Animal , Immunohistochemistry , Peptide Fragments/metabolism , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...