Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 30(14): 127240, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32527542

ABSTRACT

The (Z)-fluoro-olefin amide bioisosteric replacement is an effective tool for addressing various shortcomings of the parent amide. In an effort to fine tune ADME properties of BACE1 preclinical candidate AM-6494, a series of structurally distinct (Z)-fluoro-olefin containing analogs was developed that culminated in compound 15. Herein, we detail design considerations, synthetic challenges, structure activity relationship (SAR) studies, and in vivo properties of an advanced compound in this novel series of BACE1 inhibitors.


Subject(s)
Alkenes/pharmacology , Amides/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Development , Enzyme Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Alkenes/chemical synthesis , Alkenes/chemistry , Amides/chemical synthesis , Amides/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship
2.
J Med Chem ; 63(5): 2263-2281, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31589043

ABSTRACT

ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1) is an aspartyl protease that plays a key role in the production of amyloid ß (Aß) in the brain and has been extensively pursued as a target for the treatment of Alzheimer's disease (AD). BACE2, an aspartyl protease that is structurally related to BACE1, has been recently reported to be involved in melanosome maturation and pigmentation. Herein, we describe the development of a series of cyclopropylthiazines as potent and orally efficacious BACE1 inhibitors. Lead optimization led to the identification of 20, a molecule with biochemical IC50 BACE2/BACE1 ratio of 47. Administration of 20 resulted in no skin/fur color change in a 13-day mouse hypopigmentation study and demonstrated robust and sustained reduction of CSF and brain Aß40 levels in rat and monkey pharmacodynamic models. On the basis of a compelling data package, 20 (AM-6494) was advanced to preclinical development.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cyclopropanes/pharmacology , Enzyme Inhibitors/pharmacology , Thiazines/pharmacology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/drug effects , Brain/metabolism , Cyclopropanes/chemistry , Cyclopropanes/pharmacokinetics , Cyclopropanes/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Male , Mice , Models, Molecular , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/metabolism , Rats, Sprague-Dawley , Thiazines/chemistry , Thiazines/pharmacokinetics , Thiazines/therapeutic use
3.
ACS Med Chem Lett ; 6(2): 210-5, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25699151

ABSTRACT

BACE1 inhibition to prevent Aß peptide formation is considered to be a potential route to a disease-modifying treatment for Alzheimer's disease. Previous efforts in our laboratory using a combined structure- and property-based approach have resulted in the identification of aminooxazoline xanthenes as potent BACE1 inhibitors. Herein, we report further optimization leading to the discovery of inhibitor 15 as an orally available and highly efficacious BACE1 inhibitor that robustly reduces CSF and brain Aß levels in both rats and nonhuman primates. In addition, compound 15 exhibited low activity on the hERG ion channel and was well tolerated in an integrated cardiovascular safety model.

4.
Bioorg Med Chem Lett ; 25(4): 767-74, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25613679

ABSTRACT

The ß-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) is one of the most hotly pursued targets for the treatment of Alzheimer's disease. We used a structure- and property-based drug design approach to identify 2-aminooxazoline 3-azaxanthenes as potent BACE1 inhibitors which significantly reduced CSF and brain Aß levels in a rat pharmacodynamic model. Compared to the initial lead 2, compound 28 exhibited reduced potential for QTc prolongation in a non-human primate cardiovascular safety model.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Xanthenes/chemistry , Xanthenes/pharmacology , Alzheimer Disease/drug therapy , Animals , Cell Line , HEK293 Cells , Humans , Protease Inhibitors/chemical synthesis , Rats , Xanthenes/chemical synthesis
5.
J Med Chem ; 57(23): 9796-810, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25389560

ABSTRACT

The optimization of a series of aminooxazoline xanthene inhibitors of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is described. An early lead compound showed robust Aß lowering activity in a rat pharmacodynamic model, but advancement was precluded by a low therapeutic window to QTc prolongation in cardiovascular models consistent with in vitro activity on the hERG ion channel. While the introduction of polar groups was effective in reducing hERG binding affinity, this came at the expense of higher than desired Pgp-mediated efflux. A balance of low Pgp efflux and hERG activity was achieved by lowering the polar surface area of the P3 substituent while retaining polarity in the P2' side chain. The introduction of a fluorine in position 4 of the xanthene ring improved BACE1 potency (5-10-fold). The combination of these optimized fragments resulted in identification of compound 40, which showed robust Aß reduction in a rat pharmacodynamic model (78% Aß reduction in CSF at 10 mg/kg po) and also showed acceptable cardiovascular safety in vivo.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Oxazolone/analogs & derivatives , Protease Inhibitors/chemical synthesis , Xanthenes/chemical synthesis , Animals , Crystallography, X-Ray , HEK293 Cells , Humans , Inhibitory Concentration 50 , Male , Microsomes, Liver/metabolism , Oxazolone/chemical synthesis , Oxazolone/pharmacology , Rats, Sprague-Dawley , Structure-Activity Relationship , Xanthenes/pharmacology
6.
J Med Chem ; 57(23): 9811-31, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25363711

ABSTRACT

We have previously shown that the aminooxazoline xanthene scaffold can generate potent and orally efficacious BACE1 inhibitors although certain of these compounds exhibited potential hERG liabilities. In this article, we describe 4-aza substitution on the xanthene core as a means to increase BACE1 potency while reducing hERG binding affinity. Further optimization of the P3 and P2' side chains resulted in the identification of 42 (AMG-8718), a compound with a balanced profile of BACE1 potency, hERG binding affinity, and Pgp recognition. This compound produced robust and sustained reductions of CSF and brain Aß levels in a rat pharmacodynamic model and exhibited significantly reduced potential for QTc elongation in a cardiovascular safety model.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Benzopyrans/chemical synthesis , Protease Inhibitors/chemical synthesis , Pyridines/chemical synthesis , Spiro Compounds/chemical synthesis , Amyloid beta-Peptides/metabolism , Animals , Benzopyrans/pharmacology , Ether-A-Go-Go Potassium Channels/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Pyridines/pharmacology , Rats, Sprague-Dawley , Spiro Compounds/pharmacology , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 23(23): 6447-54, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24139583

ABSTRACT

γ-Secretase modulators (GSMs) are potentially disease-modifying treatments for Alzheimer's disease. They selectively lower pathogenic Aß42 levels by shifting the enzyme cleavage sites without inhibiting γ-secretase activity, possibly avoiding known adverse effects observed with complete inhibition of the enzyme complex. A cell-based HTS effort identified the sulfonamide 1 as a GSM lead. Lead optimization studies identified compound 25 with improved cell potency, PKDM properties, and it lowered Aß42 levels in the cerebrospinal fluid (CSF) of Sprague-Dawley rats following oral administration. Further optimization of 25 to improve cellular potency is described.


Subject(s)
Alzheimer Disease/drug therapy , Amides/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Picolines/pharmacology , Alzheimer Disease/enzymology , Amides/chemistry , Animals , HEK293 Cells , Humans , Picolines/chemistry , Rats , Rats, Sprague-Dawley
8.
J Med Chem ; 55(21): 9156-69, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22928914

ABSTRACT

A structure- and property-based drug design approach was employed to identify aminooxazoline xanthenes as potent and selective human ß-secretase inhibitors. These compounds exhibited good isolated enzyme, cell potency, and selectivity against the structurally related aspartyl protease cathepsin D. Our efforts resulted in the identification of a potent, orally bioavailable CNS penetrant compound that exhibited in vivo efficacy. A single oral dose of compound 11a resulted in a significant reduction of CNS Aß40 in naive rats.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/metabolism , Oxazoles/chemical synthesis , Xanthenes/chemical synthesis , Administration, Oral , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Molecular Structure , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Xanthenes/pharmacokinetics , Xanthenes/pharmacology
9.
J Pharmacol Exp Ther ; 343(2): 460-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22911925

ABSTRACT

Sequential proteolytic cleavage of the amyloid precursor protein (APP) by ß-site APP-cleaving enzyme 1 (BACE1) and the γ-secretase complex produces the amyloid-ß peptide (Aß), which is believed to play a critical role in the pathology of Alzheimer's disease (AD). The aspartyl protease BACE1 catalyzes the rate-limiting step in the production of Aß, and as such it is considered to be an important target for drug development in AD. The development of a BACE1 inhibitor therapeutic has proven to be difficult. The active site of BACE1 is relatively large. Consequently, to achieve sufficient potency, many BACE1 inhibitors have required unfavorable physicochemical properties such as high molecular weight and polar surface area that are detrimental to efficient passage across the blood-brain barrier. Using a rational drug design approach we have designed and developed a new series of hydroxyethylamine-based inhibitors of BACE1 capable of lowering Aß levels in the brains of rats after oral administration. Herein we describe the in vitro and in vivo characterization of two of these molecules and the overall relationship of compound properties [e.g., in vitro permeability, P-glycoprotein (P-gp) efflux, metabolic stability, and pharmacological potency] to the in vivo pharmacodynamic effect with more than 100 compounds across the chemical series. We demonstrate that high in vitro potency for BACE1 was not sufficient to provide central efficacy. A combination of potency, high permeability, low P-gp-mediated efflux, and low clearance was required for compounds to produce robust central Aß reduction after oral dosing.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ethylamines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Blood Proteins/metabolism , Cell Membrane Permeability/drug effects , Enzyme Inhibitors/pharmacokinetics , Ethylamines/pharmacokinetics , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , In Vitro Techniques , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Structure-Activity Relationship
10.
J Med Chem ; 55(21): 9009-24, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22468639

ABSTRACT

A series of potent hydroxyethyl amine (HEA) derived inhibitors of ß-site APP cleaving enzyme (BACE1) was optimized to address suboptimal pharmacokinetics and poor CNS partitioning. This work identified a series of benzodioxolane analogues that possessed improved metabolic stability and increased oral bioavailability. Subsequent efforts focused on improving CNS exposure by limiting susceptibility to Pgp-mediated efflux and identified an inhibitor which demonstrated robust and sustained reduction of CNS ß-amyloid (Aß) in Sprague-Dawley rats following oral administration.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/drug effects , Dioxolanes/chemical synthesis , Ethylamines/chemical synthesis , Peptide Fragments/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Animals , Biological Availability , Brain/metabolism , Crystallography, X-Ray , Dioxolanes/pharmacokinetics , Dioxolanes/pharmacology , Dogs , Drug Design , Ethylamines/pharmacokinetics , Ethylamines/pharmacology , Humans , Macaca mulatta , Male , Microsomes, Liver/metabolism , Models, Molecular , Protein Conformation , Protein Transport , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 55(21): 9025-44, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22468684

ABSTRACT

We have previously shown that hydroxyethylamines can be potent inhibitors of the BACE1 enzyme and that the generation of BACE1 inhibitors with CYP 3A4 inhibitory activities in this scaffold affords compounds (e.g., 1) with sufficient bioavailability and pharmacokinetic profiles to reduce central amyloid-ß peptide (Aß) levels in wild-type rats following oral dosing. In this article, we describe further modifications of the P1-phenyl ring of the hydroxyethylamine series to afford potent, dual BACE1/CYP 3A4 inhibitors which demonstrate improved penetration into the CNS. Several of these compounds caused robust reduction of Aß levels in rat CSF and brain following oral dosing, and compound 37 exhibited an improved cardiovascular safety profile relative to 1.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Peptide Fragments/metabolism , Spiro Compounds/chemical synthesis , Thiazoles/chemical synthesis , Administration, Oral , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Blood Proteins/metabolism , Brain/drug effects , Brain/metabolism , Cell Line , Crystallography, X-Ray , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Design , Humans , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Models, Molecular , Peptide Fragments/cerebrospinal fluid , Protein Binding , Protein Conformation , Rats , Rats, Sprague-Dawley , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Swine , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
12.
ACS Med Chem Lett ; 3(11): 886-91, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-24900403

ABSTRACT

ß-Secretase inhibitors are potentially disease-modifying treatments for Alzheimer's disease. Previous efforts in our laboratory have resulted in hydroxyethylamine-derived inhibitors such as 1 with low nanomolar potency against ß-site amyloid precursor protein cleaving enzyme (BACE). When dosed intravenously, compound 1 was also shown to significantly reduce Aß40 levels in plasma, brain, and cerebral spinal fluid. Herein, we report further optimizations that led to the discovery of inhibitor 16 as a novel, potent, and orally efficacious BACE inhibitor.

13.
J Med Chem ; 54(16): 5836-57, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21707077

ABSTRACT

Using fragment-based screening of a focused fragment library, 2-aminoquinoline 1 was identified as an initial hit for BACE1. Further SAR development was supported by X-ray structures of BACE1 cocrystallized with various ligands and molecular modeling studies to expedite the discovery of potent compounds. These strategies enabled us to integrate the C-3 side chain on 2-aminoquinoline 1 extending deep into the P2' binding pocket of BACE1 and enhancing the ligand's potency. We were able to improve the BACE1 potency to subnanomolar range, over 10(6)-fold more potent than the initial hit (900 µM). Further elaboration of the physical properties of the lead compounds to those more consistent with good blood-brain barrier permeability led to inhibitors with greatly improved cellular activity and permeability. Compound 59 showed an IC(50) value of 11 nM on BACE1 and cellular activity of 80 nM. This compound was advanced into rat pharmacokinetic and pharmacodynamic studies and demonstrated significant reduction of Aß levels in cerebrospinal fluid (CSF).


Subject(s)
Aminoquinolines/chemical synthesis , Aminoquinolines/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aminoquinolines/chemistry , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Aspartic Acid Endopeptidases/metabolism , Biocatalysis/drug effects , Brain/drug effects , Brain/metabolism , Catalytic Domain , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Male , Models, Chemical , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
14.
Dev Dyn ; 237(9): 2405-14, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729224

ABSTRACT

Cortical development is disrupted in presenilin-1 null mutant (Psen1-/-) mice. Prior studies have commented on similarities between Psen1-/- and reeler mice. Reelin induces phosphorylation of Dab1 and activates the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Psen1 is known to modulate PI3K/Akt signaling and both known reelin receptors (apoER2 and VLDLR) are substrates for Psen1 associated gamma-secretase activity. The purpose of this study was to determine whether reelin signaling is disrupted in Psen1-/- mice. We show that, while Dab1 is hypophosphorylated late in cortical development in Psen1-/- mice, it is normally phosphorylated at earlier ages and reelin signaling is intact in Psen1-/- primary neuronal cultures. gamma-secretase activity was also not required for reelin-induced phosphorylation of Dab1. Unlike reeler mice the preplate splits in Psen1-/- brain. Thus cortical development in Psen1-/- mice fails only after splitting of the preplate and is not due to an intrinsic failure of reelin signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/embryology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Presenilin-1/physiology , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Cerebral Cortex/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Presenilin-1/genetics , Reelin Protein , Signal Transduction/genetics
15.
Development ; 132(17): 3873-83, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16079160

ABSTRACT

Mice with a null mutation of the presenilin 1 gene (Psen1(-/-)) die during late intrauterine life or shortly after birth and exhibit multiple CNS and non-CNS abnormalities, including cerebral hemorrhages and altered cortical development. The cellular and molecular basis for the developmental effects of Psen1 remain incompletely understood. Psen1 is expressed in neural progenitors in developing brain, as well as in postmitotic neurons. We crossed transgenic mice with either neuron-specific or neural progenitor-specific expression of Psen1 onto the Psen1(-/-) background. We show that neither neuron-specific nor neural progenitor-specific expression of Psen1 can rescue the embryonic lethality of the Psen1(-/-) embryo. Indeed neuron-specific expression rescued none of the abnormalities in Psen1(-/-) mice. However, Psen1 expression in neural progenitors rescued the cortical lamination defects, as well as the cerebral hemorrhages, and restored a normal vascular pattern in Psen1(-/-) embryos. Collectively, these studies demonstrate that Psen1 expression in neural progenitor cells is crucial for cortical development and reveal a novel role for neuroectodermal expression of Psen1 in development of the brain vasculature.


Subject(s)
Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cell Movement , Cerebral Cortex/embryology , Cerebral Hemorrhage/genetics , Embryo Loss , Embryo, Mammalian/blood supply , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Intermediate Filament Proteins/genetics , Introns/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Neurons/pathology , Presenilin-1 , Rats , Stem Cells/pathology , Telencephalon/embryology , Telencephalon/metabolism , Telencephalon/pathology
16.
Neurosci Lett ; 371(2-3): 249-54, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15519767

ABSTRACT

Mice with a null mutation of the presenilin-1 (PS1-/-) gene die during late intrauterine life or shortly after birth and exhibit defects in cortical development. A previous report suggested that neurons differentiate prematurely in PS1-/- brain [M. Handler, X. Yang, J. Shen, Presenilin-1 regulates neuronal differentiation during neurogenesis, Development 127 (2000) 2593-2606]. Here we reexamined the issue of whether premature neuronal differentiation occurs in PS1-/- brain using fresh cell suspensions from embryonic E11.5 and E13.5 telencephalon where individual cell phenotypes can be easily determined with cell type specific markers. Immunostaining with seven neuronal specific markers (MAP2, beta-III tubulin, GABA, reelin, GluR2/3, calbindin, and calretinin) failed to reveal any evidence of premature neuronal differentiation in PS1-/- telencephalon. We also determined the fraction of cells expressing the neural progenitor marker nestin and found no evidence for premature depletion of neural progenitor cells in PS1-/- telencephalon. Moreover, based on MAP2 staining of tissue sections from E12.5 embryos the topography of newly generated neurons also appeared to be undisturbed in the telencephalon of PS1-/- embryos. These studies thus argue that premature neuronal differentiation is unlikely to be a core pathophysiological feature underlying the aberrant cortical development that occurs in PS1-/- brain.


Subject(s)
Cell Differentiation/physiology , Membrane Proteins/deficiency , Neurons/cytology , Stem Cells/cytology , Animals , Cells, Cultured , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Neurons/metabolism , Presenilin-1 , Reelin Protein , Stem Cells/metabolism , Telencephalon/cytology , Telencephalon/embryology , Telencephalon/metabolism
17.
Exp Neurol ; 188(2): 224-37, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15246822

ABSTRACT

The functions of presenilin 1 (PS1) and how PS1 mutations cause familial Alzheimer's disease (FAD) are incompletely understood. PS1 expression is essential for neurogenesis during embryonic development and may also influence neurogenesis in adult brain. We examined how increasing PS1 expression or expressing an FAD mutant would affect neurogenesis in the adult hippocampus. A neuron-specific enolase (NSE) promoter was used to drive neuronal overexpression of either wild-type human PS1 or the FAD mutant P117L in transgenic mice, and the animals were studied under standard-housing conditions or after environmental enrichment. As judged by bromodeoxyuridine (BrdU) labeling, neural progenitor proliferation rate was mostly unaffected by increasing expression of either wild-type or FAD mutant PS1. However, in both housing conditions, the FAD mutant impaired the survival of BrdU-labeled neural progenitor cells leading to fewer new beta-III-tubulin-immunoreactive neurons being generated in FAD mutant animals during the 4-week postlabeling period. The effect was FAD mutant specific in that neural progenitor survival and differentiation in mice overexpressing wild-type human PS1 were similar to nontransgenic controls. Two additional lines of PS1 wild-type and FAD mutant transgenic mice showed similar changes indicating that the effects were not integration site-dependent. These studies demonstrate that a PS1 FAD mutant impairs new neuron production in adult hippocampus by decreasing neural progenitor survival. They also identify a new mechanism whereby PS1 FAD mutants may impair normal neuronal function and may have implications for the physiological functioning of the hippocampus in FAD.


Subject(s)
Alzheimer Disease/genetics , Hippocampus/pathology , Membrane Proteins/genetics , Neurons/pathology , Amino Acid Substitution/genetics , Amyloid beta-Peptides/biosynthesis , Animals , Antigens, Differentiation/biosynthesis , Bromodeoxyuridine , Cell Count , Cell Survival/genetics , Disease Models, Animal , Environment , Gene Expression , Hippocampus/metabolism , Humans , Membrane Proteins/biosynthesis , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Peptide Fragments/biosynthesis , Presenilin-1 , Stem Cells/metabolism , Stem Cells/pathology
18.
Exp Neurol ; 184(1): 408-19, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14637110

ABSTRACT

Aberrant accumulation of neurofilaments is a feature of human motor neuron diseases. Experimentally motor neuron disease can be induced in transgenic mice by overexpressing the mouse neurofilament light subunit (NF-L), the human heavy subunit (NF-H), or mouse peripherin. Here we describe that mice harboring a bacterial artificial chromosome (BAC) transgene containing the human midsized neurofilament subunit (NF-M) gene develop a progressive hind limb paralysis associated with neurofilamentous accumulations in ventral horn motor neurons and axonal loss in ventral motor roots. Biochemical studies revealed that all three mouse neurofilament subunits along with the human NF-M contributed to filament formation, although filaments contained less peripherin. In addition the endogenous mouse NF-M became less phosphorylated in the presence of the human protein and accumulated in the cell bodies of affected neurons even though phosphorylated human NF-M did not. Remaining motor axons contained an increased density of neurofilaments and morphometric studies showed that principally small myelinated axons were lost in the transgenic animals. Removing half of the mouse NF-M by breeding the transgene onto the mouse NF-M heterozygous null background offered no protection against the development of disease, arguing that the effect is not simply due to elevation of total NF-M. Collectively these studies argue that the human and mouse NF-M proteins exhibit distinct biochemical properties and within mouse neurons are not interchangeable and that indeed the human protein may be toxic to some mouse neurons. These studies have implications for the use of human neurofilament transgenic mice as models of amyotrophic lateral sclerosis.


Subject(s)
Membrane Glycoproteins , Motor Neuron Disease/genetics , Neurofilament Proteins/biosynthesis , Neurofilament Proteins/genetics , Animals , Axons/ultrastructure , Blotting, Western , Cell Size , Fluorescent Antibody Technique , Hindlimb/physiopathology , Humans , Inclusion Bodies/pathology , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Electron , Motor Neuron Disease/metabolism , Motor Neurons/pathology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurofilament Proteins/metabolism , Paralysis/chemically induced , Paralysis/genetics , Paralysis/physiopathology , Peripherins , Phosphorylation
19.
Cell ; 114(5): 635-45, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-13678586

ABSTRACT

Presenilin1 (PS1), a protein implicated in Alzheimer's disease (AD), forms complexes with N-cadherin, a transmembrane protein with important neuronal and synaptic functions. Here, we show that a PS1-dependent gamma-secretase protease activity promotes an epsilon-like cleavage of N-cadherin to produce its intracellular domain peptide, N-Cad/CTF2. NMDA receptor agonists stimulate N-Cad/CTF2 production suggesting that this receptor regulates the epsilon-cleavage of N-cadherin. N-Cad/CTF2 binds the transcription factor CBP and promotes its proteasomal degradation, inhibiting CRE-dependent transactivation. Thus, the PS1-dependent epsilon-cleavage product N-Cad/CTF2 functions as a potent repressor of CBP/CREB-mediated transcription. Importantly, PS1 mutations associated with familial AD (FAD) and a gamma-secretase dominant-negative mutation inhibit N-Cad/CTF2 production and upregulate CREB-mediated transcription indicating that FAD mutations cause a gain of transcriptional function by inhibiting production of transcriptional repressor N-Cad/CTF2. These data raise the possibility that FAD mutation-induced transcriptional abnormalities maybe causally related to the dementia associated with FAD.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Transcription, Genetic , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Blotting, Western , Cadherins/chemistry , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cysteine Endopeptidases/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Endopeptidases/metabolism , Genes, Dominant , Genetic Vectors , Humans , Immunoblotting , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Multienzyme Complexes/metabolism , Mutation , Neurons/metabolism , Peptides/chemistry , Precipitin Tests , Presenilin-1 , Proteasome Endopeptidase Complex , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Synapses/metabolism , Time Factors , Transcriptional Activation , Tubulin/metabolism
20.
Neurobiol Dis ; 10(1): 8-19, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12079399

ABSTRACT

Mutations in the presenilin-1 (PS-1) gene are one cause of familial Alzheimer's disease (FAD). However, the functions of the PS-1 protein as well as how PS-1 mutations cause FAD are incompletely understood. Here we investigated if neuronal overexpression of wild-type or FAD mutant PS-1 in transgenic mice affects neurogenesis in the hippocampus of adult animals. We show that either a wild-type or an FAD mutant PS-1 transgene reduces the number of neural progenitors in the dentate gyrus. However, the wild-type, but not the FAD mutant PS-1 promoted the survival and differentiation of progenitors leading to more immature granule cell neurons being generated in PS-1 wild type expressing animals. These studies suggest that PS-1 plays a role in regulating neurogenesis in adult hippocampus and that FAD mutants may have deleterious properties independent of their effects on amyloid deposition.


Subject(s)
Alzheimer Disease/genetics , Hippocampus/cytology , Hippocampus/physiology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mutation/physiology , Neurons/cytology , Neurons/physiology , Aging/genetics , Aging/physiology , Alzheimer Disease/metabolism , Animals , Cell Count , Cell Differentiation/genetics , Cell Survival/genetics , Hippocampus/growth & development , Humans , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Neurons/metabolism , Presenilin-1 , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...