Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Methods Enzymol ; 584: 295-308, 2017.
Article in English | MEDLINE | ID: mdl-28065268

ABSTRACT

Proteolysis within the membrane is catalyzed by a diverse family of proteases immersed within the hydrophobic environment of cellular membranes. These ubiquitous intramembrane-cleaving proteases (I-CLiPs) hydrolyze the transmembrane domains of a large variety of membrane-embedded proteins to facilitate signaling events essential to normal biological functions found in all forms of life. The importance of this unique class of enzyme is highlighted by its central involvement in a variety of human pathologies, including Alzheimer's disease (AD), Parkinson's disease, cancer, and the virulence of a number of viral, bacterial, and fungal pathogens. I-CLiPs therefore represent promising targets for the therapeutic treatment of numerous diseases. The key to understanding the normal biological function of I-CLiPs and capitalizing on their therapeutic potential is through a thorough understanding of the complex catalytic mechanisms that govern this unusual class of enzyme. This is an intrinsically difficult endeavor, given that these enzymes and their substrates reside within lipid membranes, making any in vitro assay technically challenging to design and execute. Here, we describe several in vitro enzymatic assays for the study of the AD-associated γ-secretase protease, which have aided the development of potent γ-secretase-targeting compounds as candidate therapeutics. These assays have also been applied in various forms for the study of other I-CLiPs, providing valuable mechanistic insights into some of the functional similarities and differences between several members of this fascinating family of proteases.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/chemistry , Enzyme Assays/methods , Proteolysis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/biosynthesis , Humans , Membrane Lipids/chemistry , Signal Transduction , Substrate Specificity
2.
Curr Alzheimer Res ; 7(3): 207-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20088802

ABSTRACT

Overwhelming evidence supports a central role for the amyloid beta-peptide (Abeta) in the pathogenesis of Alzheimer's disease (AD), and the proteases that produce Abeta from its precursor protein APP are top targets for therapeutic intervention. Considerable effort has focused on targeting gamma-secretase, which generates the C-terminus of Abeta; however, gamma-secretase inhibitors cause serious toxicities due to interference with the Notch signaling pathway. We have been working toward compounds that directly alter gamma-secretase activity to reduce Abeta production without affecting the proteolysis of Notch. Using purified enzyme and substrate, we have shown that gamma-secretase can be selectively inhibited in this way by naphthyl-substituted gamma-aminoketones and gamma-aminoalcohols. These early hits, however, suffered from chemical instability and/or poor potency. Iterative design, synthesis and evaluation have led to the discovery of Notch-sparing gamma-secretase inhibitors with substantially increased potencies in biochemical and cellular assays. These compounds are of low molecular weight and are under evaluation for drug-like properties. The discovery and development of these compounds will be discussed.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Receptors, Notch/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
3.
Dev Biol ; 278(2): 381-95, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15680358

ABSTRACT

Notch-Delta signaling has been implicated in several alternative modes of function in the vertebrate retina. To further investigate these functions, we examined retinas from zebrafish embryos in which bidirectional Notch-Delta signaling was inactivated either by the mind bomb (mib) mutation, which disrupts E3 ubiquitin ligase activity, or by treatment with gamma-secretase inhibitors, which prevent intramembrane proteolysis of Notch and Delta. We found that inactivating Notch-Delta signaling did not prevent differentiation of retinal neurons, but it did disrupt spatial patterning in both the apical-basal and planar dimensions of the retinal epithelium. Retinal neurons differentiated, but their laminar arrangement was disrupted. Photoreceptor differentiation was initiated normally, but its progression was slowed. Although confined to the apical retinal surface as in normal retinas, the planar organization of cone photoreceptors was disrupted: cones of the same spectral subtype were clumped rather than regularly spaced. In contrast to neurons, Müller glia failed to differentiate suggesting an instructive role for Notch-Delta signaling in gliogenesis.


Subject(s)
Body Patterning/physiology , Membrane Proteins/physiology , Neuroglia/physiology , Retina/embryology , Animals , Cell Death , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Intracellular Signaling Peptides and Proteins , Microscopy, Fluorescence , Mutation , Receptors, Notch , Retina/cytology , Retinal Cone Photoreceptor Cells/embryology , Retinal Rod Photoreceptor Cells/embryology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Zebrafish/embryology , Zebrafish Proteins/physiology
4.
Biol Lett ; 1(4): 431-4, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-17148225

ABSTRACT

Habitat and biodiversity differences between matched pairs of organic and non-organic farms containing cereal crops in lowland England were assessed by a large-scale study of plants, invertebrates, birds and bats. Habitat extent, composition and management on organic farms was likely to favour higher levels of biodiversity and indeed organic farms tended to support higher numbers of species and overall abundance across most taxa. However, the magnitude of the response varied; plants showed larger and more consistent responses than other taxa. Variation in response across taxa may be partly a consequence of the small size and isolated context of many organic farms. Extension of organic farming could contribute to the restoration of biodiversity in agricultural landscapes.


Subject(s)
Agriculture/standards , Biodiversity , Edible Grain/growth & development , Animals , England , Population Density
6.
Science ; 293(5534): 1449-54, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11520976

ABSTRACT

The amyloid beta-peptide (Abeta) is a principal component of the cerebral plaques found in the brains of patients with Alzeheimer's disease (AD). This insoluble 40- to 42-amino acid peptide is formed by the cleavage of the Abeta precursor protein (APP). The three proteases that cleave APP, alpha-, beta-, and gamma-secretases, have been implicated in the etiology of AD. beta-Secretase is a membrane-anchored protein with clear homology to soluble aspartyl proteases, and alpha-secretase displays characteristics of certain membrane-tethered metalloproteases. gamma-Secretase is apparently an oligomeric complex that includes the presenilins, which may be the catalytic component of this protease. Identification of the alpha-, beta-, and gamma-secretases provides potential targets for designing new drugs to treat AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Endopeptidases/metabolism , Membrane Proteins/metabolism , ADAM Proteins , ADAM17 Protein , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/chemistry , Brain/enzymology , Catalytic Domain , Dimerization , Endopeptidases/chemistry , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Presenilin-1 , Presenilin-2 , Receptors, Notch , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 98(13): 7487-91, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11416218

ABSTRACT

A major therapeutic target in the search for a cure to the devastating Alzheimer's disease is gamma-secretase. This activity resides in a multiprotein enzyme complex responsible for the generation of Abeta42 peptides, precipitates of which are thought to cause the disease. Gamma-secretase is also a critical component of the Notch signal transduction pathway; Notch signals regulate development and differentiation of adult self-renewing cells. This has led to the hypothesis that therapeutic inhibition of gamma-secretase may interfere with Notch-related processes in adults, most alarmingly in hematopoiesis. Here, we show that application of gamma-secretase inhibitors to fetal thymus organ cultures interferes with T cell development in a manner consistent with loss or reduction of Notch1 function. Progression from an immature CD4-/CD8- state to an intermediate CD4+/CD8+ double-positive state was repressed. Furthermore, treatment beginning later at the double-positive stage specifically inhibited CD8+ single-positive maturation but did not affect CD4+ single-positive cells. These results demonstrate that pharmacological gamma-secretase inhibition recapitulates Notch1 loss in a vertebrate tissue and present a system in which rapid evaluation of gamma-secretase-targeted pharmaceuticals for their ability to inhibit Notch activity can be performed in a relevant context.


Subject(s)
Endopeptidases/metabolism , Protease Inhibitors/pharmacology , Receptors, Cell Surface , T-Lymphocytes/physiology , Transcription Factors , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Line , Cells, Cultured , Humans , Kidney , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutagenesis , Organ Culture Techniques , Receptor, Notch1 , Recombinant Proteins/metabolism , Sequence Deletion , Thymus Gland/embryology , Thymus Gland/immunology , Transfection
10.
Brain Res Mol Brain Res ; 87(2): 166-74, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11245918

ABSTRACT

Presenilin1 (PS1) has been implicated in normal Notch1 processing and signaling in several experimental systems. In the present study, the relationship between PS1 and Notch1 in mammalian neurons is studied by analyzing Notch1 cleavage and C-terminal nuclear translocation as well as Notch1 signaling via the transactivation of a CBF1-luciferase reporter construct. We show that full-length Notch1 [N1(FL)] transfected into wild type (WT) primary neurons is cleaved in the presence of its biological ligand Delta (Dl) and translocated to the nucleus within 1--3 min of ligand addition. PS1 deficient neurons show normal Notch1 insertion into the cellular membrane, yet lack Notch1 activation resulting in markedly inhibited nuclear translocation of the C-terminal Notch fragment (NICD). PS1 deficient neurons also have impaired Notch1 signaling which can be restored fully or partially to levels seen in WT littermates by transfection with WT or familial Alzheimer's disease-associated M146L mutant PS1, respectively. We also show that pharmacological inhibition of PS1-associated gamma-secretase activity parallels the effects of genetic PS1 deficiency in these assays. These results support the hypothesis that PS1 deficiency blocks neuronal Notch1 processing and signaling.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurons/enzymology , Nuclear Proteins , Receptors, Cell Surface , Transcription Factors , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cerebral Cortex/cytology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , Hippocampus/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Luciferases/genetics , Mammals , Mice , Neurons/cytology , Presenilin-1 , Receptor, Notch1 , Transfection
12.
J Mol Neurosci ; 17(2): 199-204, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11816793

ABSTRACT

Mutations in the presenilins cause Alzheimer's disease (AD) and alter gamma-secretase activity to increase the production of the 42-residue amyloid-beta peptide (Abeta) found disproportionally in the cerebral plaques that characterize the disease. The serpentine presenilins are required for transmembrane cleavage of both the amyloid-beta precursor protein (APP) and the Notch receptor by y-secretase, and presenilins are biochemically associated with the protease. Inhibitors of gamma-secretase have provided critical clues to the function of presenilins. Pharmacological profiling suggested that gamma-secretase is an aspartyl protease, leading to the identification of two conserved aspartates important to presenilin's role in proteolysis. Conversion of transition-state analogue inhibitors of gamma-secretase to affinity reagents resulted in specific tagging of the heterodimeric form of presenilins, strongly suggesting that the active site of gamma-secretase lies at the interface of the presenilin heterodimer. Heterodimeric presenilin appears to be the catalytic portion of a multi-protein gamma-secretase complex.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/metabolism , Humans , Presenilin-1
13.
Neurobiol Dis ; 7(6 Pt B): 673-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11114265

ABSTRACT

To investigate the mechanism of regulation of Ass production by familial Alzheimer's disease (FAD)-linked presenilin 1 (PS1), we used a cell-free system that allows de novo Ass generation to examine whether PS1 participates directly in the gamma-secretase reaction. Optimal Ass generation in vitro was achieved at mildly acidic pH and could be inhibited by the aspartyl protease inhibitor pepstatin A, consistent with the suggestion that gamma-secretase is an aspartyl protease. Dominant negative mutations of the critical transmembrane aspartates in PS1 or full deletion of PS1 did not alter the maturation of APP in the secretory pathway. Instead, PS1 had a direct effect on the inhibition of Ass production by a designed peptidomimetic inhibitor: the inhibition was significantly less effective in cells expressing FAD-causing mutations in either APP or PS1 than in cells expressing the wild-type proteins. Taken together, these findings suggest that PS1 participates physically in a complex with APP during the gamma-secretase cleavage event.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Binding Sites/drug effects , Binding Sites/genetics , CHO Cells , Cell Fractionation , Cell-Free System/metabolism , Cricetinae , Endopeptidases/drug effects , Genes, Dominant , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Macromolecular Substances , Membrane Proteins/genetics , Microsomes/metabolism , Mutation, Missense , Pepstatins/pharmacology , Presenilin-1 , Protein Binding/drug effects , Protein Processing, Post-Translational
14.
J Med Chem ; 43(18): 3434-42, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10978191

ABSTRACT

The final step in the generation of the amyloid-beta protein (Abeta), implicated in the etiology of Alzheimer's disease, is proteolysis within the transmembrane region of the amyloid precursor protein (APP) by gamma-secretase. Although considered an important target for therapeutic design, gamma-secretase has been neither well-characterized nor definitively identified. Previous studies in our laboratory using substrate-based difluoro ketone and difluoro alcohol transition-state analogue inhibitors suggest that gamma-secretase is an aspartyl protease with loose sequence specificity. To further characterize the active site of gamma-secretase, we prepared a series of difluoro ketone peptide analogues with varying steric bulkiness in the P1 position and tested the ability of these compounds to inhibit Abeta production in APP-transfected cells. Incorporation of bulky, aliphatic P1 side chains, such as sec-butyl or cyclohexylmethyl, led to increased gamma-secretase inhibitory potency, suggesting a large S1 pocket to accommodate these substituents and providing further evidence for loose sequence specificity. The cyclohexylmethyl P1 substituent allowed N-terminal truncation to a low-molecular-weight compound (<600 Da) that effectively blocked Abeta production (IC(50) approximately 5 microM). This finding suggests that optimal S1 binding may allow the development of potent inhibitors with ideal pharmaceutical properties. Moreover, a difluoro alcohol analogue with a cyclohexylmethyl P1 substituent was equipotent with its difluoro ketone counterpart, providing strong evidence that gamma-secretase is an aspartyl protease. All new analogues inhibited total Abeta and Abeta(42) production with the same rank order of potency and increased Abeta(42) production at low concentrations, providing further evidence for distinct gamma-secretases that are nevertheless closely similar with respect to active site topology and mechanism.


Subject(s)
Alzheimer Disease/enzymology , Endopeptidases/metabolism , Ketones/chemical synthesis , Peptides/chemistry , Protease Inhibitors/chemical synthesis , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Animals , CHO Cells , Catalytic Domain , Cell Line , Cricetinae , Drug Design , Ketones/chemistry , Ketones/pharmacology , Molecular Mimicry , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/biosynthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship
15.
Nature ; 406(6797): 681-2, 2000 Aug 17.
Article in English | MEDLINE | ID: mdl-10963576
16.
Proc Natl Acad Sci U S A ; 97(16): 9299-304, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10922078

ABSTRACT

An unusual intramembranous cleavage of the beta-amyloid precursor protein (APP) by gamma-secretase is the final step in the generation of amyloid beta-peptide (Abeta). Two conserved aspartates in transmembrane (TM) domains 6 and 7 of presenilin (PS) 1 are required for Abeta production by gamma-secretase. Here we report that the APP C-terminal fragments, C83 and C99, which are the direct substrates of gamma-secretase, can be coimmunoprecipitated with both PS1 and PS2. PS/C83 complexes were detected in cells expressing endogenous levels of PS. The complexes accumulate when gamma-secretase is inactivated either pharmacologically or by mutating the PS aspartates. PS1/C83 and PS1/C99 complexes were detected in Golgi-rich and trans-Golgi network-rich vesicle fractions. In contrast, complexes of PS1 with APP holoprotein, which is not the immediate substrate of gamma-secretase, occurred earlier in endoplasmic reticulum-rich vesicles. The major portion of intracellular Abeta at steady state was found in the same Golgi/trans-Golgi network-rich vesicles, and Abeta levels in these fractions were markedly reduced when either PS1 TM aspartate was mutated to alanine. Furthermore, de novo generation of Abeta in a cell-free microsomal reaction occurred specifically in these same vesicle fractions and was markedly inhibited by mutating either TM aspartate. Thus, PSs are complexed with the gamma-secretase substrates C83 and C99 in the subcellular locations where Abeta is generated, indicating that PSs are directly involved in the pathogenically critical intramembranous proteolysis of APP.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases , Binding Sites , CHO Cells , Cricetinae , Endopeptidases/drug effects , Golgi Apparatus/metabolism , Humans , Hydrolysis , Presenilin-1
17.
J Pharmacol Exp Ther ; 294(3): 1099-105, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10945865

ABSTRACT

We have previously shown racial differences in propranolol kinetics, with the largest differences appearing to be in its 4-hydroxylation. The purpose of this study was to identify and confirm the cytochrome P450 enzymes (CYP) with propranolol 4-hydroxylase activity, describe their enzyme kinetics, and determine whether there were racial differences in their catalytic activity. Eleven human recombinant, expressed CYPs were screened, but only CYP1A2 and CYP2D6 possessed propranolol 4-hydroxylase activity. Subsequent studies were conducted in human liver microsomes, including correlation, inhibition, enzyme kinetics, and racial comparison studies. Significant correlations were noted between propranolol 4-hydroxylation and ethoxyresorufin-O-deethylation (marker of CYP1A2 activity), with marked improvement in the correlations when CYP2D6-mediated propranolol 4-hydroxylation was inhibited with quinidine. Inhibition studies showed that quinidine inhibited approximately 55% of propranolol 4-hydroxylation and furaphylline (CYP1A2-selective inhibitor) inhibited about 45% of propranolol 4-hydroxylation. Median (range) parameter estimates of (S)-4-hydroxypropranolol [(S)-HOP] formation were a V(max) value of 307 (165-2397) and 721 (84-1975) pmol/mg of protein/60 min for CYP1A2 and CYP2D6, respectively, and a K(m) value of 21.2 (8.9-77.5) and 8.5 (5.9-31.9) microM for CYP1A2 and CYP2D6, respectively. CYP1A2- and CYP2D6-mediated propranolol 4-hydroxylation was about 70 and 100% higher (P <.05 for both), respectively, in African-Americans compared with Caucasians. In summary, we found that both CYP1A2 and CYP2D6 catalyze formation of 4-hydroxypropranolol and that both enzymes exhibited racial differences in this reaction. The observed racial differences in drug metabolism may have relevance to drug efficacy, toxicity, or carcinogen activation for CYP1A2 or CYP2D6 substrates.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Black People , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 Enzyme System/metabolism , Mixed Function Oxygenases/metabolism , Propranolol/metabolism , White People , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 Inhibitors , Enzyme Inhibitors/pharmacology , Female , Humans , Hydroxylation , In Vitro Techniques , Kinetics , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Propranolol/chemistry , Quinidine/pharmacology , Recombinant Proteins/metabolism , Stereoisomerism
18.
J Neurochem ; 75(2): 583-93, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10899933

ABSTRACT

It has been hypothesized that a presenilin 1 (PS1)-related enzymatic activity is responsible for proteolytic cleavage of the C-terminal intracellular protein of Notch1, in addition to its role in beta-amyloid protein (Abeta) formation from the amyloid precursor protein (APP). We developed an assay to monitor ligand-induced Notch1 proteolysis and nuclear translocation in individual cells : Treatment of full-length Notch1-enhanced green fluorescent protein-transfected Chinese hamster ovary (CHO) cells with a soluble preclustered form of the physiologic ligand Delta leads to rapid accumulation of the C terminus of Notch1 in the nucleus and to transcriptional activation of a C-promoter binding factor 1 (CBF1) reporter construct. Nuclear translocation was blocked by cotransfection with Notch's physiologic inhibitor Numb. Using this assay, we now confirm and extend the observation that PS1 is involved in Notch1 nuclear translocation and signaling in mammalian cells. We demonstrate that the D257A and the D385A PS1 mutations, which had been shown previously to block APP gamma-secretase activity, also prevent Notch1 cleavage and translocation to the nucleus but do not alter Notch1 trafficking to the cell surface. We also show that two APP gamma-secretase inhibitors block Notch1 nuclear translocation with an IC(50) similar to that reported for APP gamma-secretase. Notch1 signaling, assessed by measuring the activity of CBF1, a downstream transcription factor, was impaired but not abolished by the PS1 aspartate mutations or gamma-secretase inhibitors. Our results support the hypotheses that (a) PS1-dependent APP gamma-secretase-like enzymatic activity is critical for both APP and Notch processing and (b) the Notch1 signaling pathway remains partially activated even when Notch1 proteolytic processing and nuclear translocation are markedly inhibited. The latter is an important finding from the perspective of therapeutic treatment of Alzheimer's disease by targeting gamma-secretase processing of APP to reduce Abeta production.


Subject(s)
Aspartic Acid , Endopeptidases/metabolism , Membrane Proteins/metabolism , Nuclear Proteins , Receptors, Cell Surface , Transcription Factors , Amino Acid Substitution , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , CHO Cells , Cricetinae , DNA-Binding Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Luciferases/genetics , Luminescent Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Point Mutation , Presenilin-1 , Presenilin-2 , Protease Inhibitors/pharmacology , Receptor, Notch1 , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Transfection
19.
Nat Cell Biol ; 2(7): 428-34, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10878808

ABSTRACT

The beta-amyloid precursor protein (beta-APP), which is involved in the pathogenesis of Alzheimer's disease, and the Notch receptor, which is responsible for critical signalling events during development, both undergo unusual proteolysis within their transmembrane domains by unknown gamma-secretases. Here we show that an affinity reagent designed to interact with the active site of gamma-secretase binds directly and specifically to heterodimeric forms of presenilins, polytopic proteins that are mutated in hereditary Alzheimer's and are known mediators of gamma-secretase cleavage of both beta-APP and Notch. These results provide evidence that heterodimeric presenilins contain the active site of gamma-secretase, and validate presenilins as principal targets for the design of drugs to treat and prevent Alzheimer's disease.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Affinity Labels , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , CHO Cells , Cricetinae , Dimerization , Humans , Membrane Proteins/chemistry , Microsomes/chemistry , Microsomes/metabolism , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Presenilin-1 , Presenilin-2 , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Protein Binding , Protein Processing, Post-Translational , Transfection
20.
Pharmacotherapy ; 20(6): 622-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10853616

ABSTRACT

STUDY OBJECTIVE: To evaluate the impact of gender on labetalol kinetics. DESIGN: Part of a randomized, crossover study. SETTING: Academic medical center. PATIENTS: Nineteen hypertensive patients (14 men, 5 women; 6 blacks, 13 whites). INTERVENTIONS: Participants had labetalol dosages titrated to a specific antihypertensive response, then underwent ambulatory blood pressure monitoring (ABPM) and a pharmacokinetic study. Labetalol plasma concentrations were measured by high-performance liquid chromatography (HPLC) and labetalol stereoisomer ratios were determined in a single plasma sample by chiral HPLC, both with fluorescence detection. MEASUREMENTS AND MAIN RESULTS: Labetalol concentrations were 80% higher in women (area under the concentration-time curve [AUC]/dose x 1000: 6.79 +/- 2.11 in women vs 3.82 +/- 1.37 hr/L in men, p<0.05), yet both genders had a similar antihypertensive response by 24-hour ABPM. Dose-corrected AUC (AUC/dose x 1000) for labetalol's stereoisomers in women and men, respectively, were S,R-labetalol 7.55 +/- 1.47 and 4.83 +/- 1.54 hr/L (p<0.05), S,S-labetalol 8.23 +/- 2.93 and 4.65 +/- 1.78 hr/L (p<0.05), R,S-labetalol 6.99 +/- 3.30 and 4.25 +/- 2.35 hr/L (p=0.11), and R,R-labetalol 3.91 +/- 2.57 and 3.55 +/- 3.08 hr/L (NS). CONCLUSION: The higher labetalol concentration in women than in men was explained largely by differences in inactive and alpha1-blocking stereoisomers. However, concentrations were similar between genders for the beta-blocking stereoisomer (R,R-labetalol), possibly explaining the similarity in antihypertensive response to the drug. This study highlights the importance of determining stereoisomer kinetics for agents administered as racemates, particularly when relating concentrations to pharmacologic response.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Labetalol/pharmacokinetics , Adult , Cross-Over Studies , Female , Humans , Male , Middle Aged , Sex Characteristics , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...