Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mamm Genome ; 34(2): 244-261, 2023 06.
Article in English | MEDLINE | ID: mdl-37160609

ABSTRACT

Rare diseases (RDs) are a challenge for medicine due to their heterogeneous clinical manifestations and low prevalence. There is a lack of specific treatments and only a few hundred of the approximately 7,000 RDs have an approved regime. Rapid technological development in genome sequencing enables the mass identification of potential candidates that in their mutated form could trigger diseases but are often not confirmed to be causal. Knockout (KO) mouse models are essential to understand the causality of genes by allowing highly standardized research into the pathogenesis of diseases. The German Mouse Clinic (GMC) is one of the pioneers in mouse research and successfully uses (preclinical) data obtained from single-gene KO mutants for research into monogenic RDs. As part of the International Mouse Phenotyping Consortium (IMPC) and INFRAFRONTIER, the pan-European consortium for modeling human diseases, the GMC expands these preclinical data toward global collaborative approaches with researchers, clinicians, and patient groups.Here, we highlight proprietary genes that when deleted mimic clinical phenotypes associated with known RD targets (Nacc1, Bach2, Klotho alpha). We focus on recognized RD genes with no pre-existing KO mouse models (Kansl1l, Acsf3, Pcdhgb2, Rabgap1, Cox7a2) which highlight novel phenotypes capable of optimizing clinical diagnosis. In addition, we present genes with intriguing phenotypic data (Zdhhc5, Wsb2) that are not presently associated with known human RDs.This report provides comprehensive evidence for genes that when deleted cause differences in the KO mouse across multiple organs, providing a huge translational potential for further understanding monogenic RDs and their clinical spectrum. Genetic KO studies in mice are valuable to further explore the underlying physiological mechanisms and their overall therapeutic potential.


Subject(s)
Rare Diseases , Mice , Animals , Humans , Mice, Knockout , Rare Diseases/genetics , Gene Knockout Techniques , Phenotype
2.
PLoS Genet ; 16(12): e1009190, 2020 12.
Article in English | MEDLINE | ID: mdl-33370286

ABSTRACT

The genetic landscape of diseases associated with changes in bone mineral density (BMD), such as osteoporosis, is only partially understood. Here, we explored data from 3,823 mutant mouse strains for BMD, a measure that is frequently altered in a range of bone pathologies, including osteoporosis. A total of 200 genes were found to significantly affect BMD. This pool of BMD genes comprised 141 genes with previously unknown functions in bone biology and was complementary to pools derived from recent human studies. Nineteen of the 141 genes also caused skeletal abnormalities. Examination of the BMD genes in osteoclasts and osteoblasts underscored BMD pathways, including vesicle transport, in these cells and together with in silico bone turnover studies resulted in the prioritization of candidate genes for further investigation. Overall, the results add novel pathophysiological and molecular insight into bone health and disease.


Subject(s)
Bone Density/genetics , Gene Expression Regulation/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/genetics , Animals , Female , Gene Ontology , Genetic Pleiotropy , Genome-Wide Association Study , Genotype , Male , Mice , Mice, Transgenic , Mutation , Osteoblasts/pathology , Osteoclasts/pathology , Osteoporosis/metabolism , Phenotype , Promoter Regions, Genetic , Protein Interaction Maps , Sex Characteristics , Transcriptome
3.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166760, 2023 10.
Article in English | MEDLINE | ID: mdl-37230398

ABSTRACT

The alternative oxidase, AOX, provides a by-pass of the cytochrome segment of the mitochondrial respiratory chain when the chain is unavailable. AOX is absent from mammals, but AOX from Ciona intestinalis is benign when expressed in mice. Although non-protonmotive, so does not contribute directly to ATP production, it has been shown to modify and in some cases rescue phenotypes of respiratory-chain disease models. Here we studied the effect of C. intestinalis AOX on mice engineered to express a disease-equivalent mutant of Uqcrh, encoding the hinge subunit of mitochondrial respiratory complex III, which results in a complex metabolic phenotype beginning at 4-5 weeks, rapidly progressing to lethality within a further 6-7 weeks. AOX expression delayed the onset of this phenotype by several weeks, but provided no long-term benefit. We discuss the significance of this finding in light of the known and hypothesized effects of AOX on metabolism, redox homeostasis, oxidative stress and cell signaling. Although not a panacea, the ability of AOX to mitigate disease onset and progression means it could be useful in treatment.


Subject(s)
Electron Transport Complex III , Mitochondria , Animals , Mice , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Mitochondrial Membranes/metabolism , Phenotype , Transcription Factors/metabolism , Mammals/metabolism
4.
Nat Commun ; 13(1): 6830, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36369285

ABSTRACT

Current concepts regarding the biology of aging are primarily based on studies aimed at identifying factors regulating lifespan. However, lifespan as a sole proxy measure for aging can be of limited value because it may be restricted by specific pathologies. Here, we employ large-scale phenotyping to analyze hundreds of markers in aging male C57BL/6J mice. For each phenotype, we establish lifetime profiles to determine when age-dependent change is first detectable relative to the young adult baseline. We examine key lifespan regulators (putative anti-aging interventions; PAAIs) for a possible countering of aging. Importantly, unlike most previous studies, we include in our study design young treated groups of animals, subjected to PAAIs prior to the onset of detectable age-dependent phenotypic change. Many PAAI effects influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these PAAIs have limited effects on aging.


Subject(s)
Aging , Longevity , Mice , Animals , Male , Longevity/genetics , Mice, Inbred C57BL , Aging/physiology , Phenotype
5.
EMBO Mol Med ; 13(12): e14397, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34750991

ABSTRACT

Mitochondrial disorders are clinically and genetically diverse, with isolated complex III (CIII) deficiency being relatively rare. Here, we describe two affected cousins, presenting with recurrent episodes of severe lactic acidosis, hyperammonaemia, hypoglycaemia and encephalopathy. Genetic investigations in both cases identified a homozygous deletion of exons 2 and 3 of UQCRH, which encodes a structural complex III (CIII) subunit. We generated a mouse model with the equivalent homozygous Uqcrh deletion (Uqcrh-/- ), which also presented with lactic acidosis and hyperammonaemia, but had a more severe, non-episodic phenotype, resulting in failure to thrive and early death. The biochemical phenotypes observed in patient and Uqcrh-/- mouse tissues were remarkably similar, displaying impaired CIII activity, decreased molecular weight of fully assembled holoenzyme and an increase of an unexpected large supercomplex (SXL ), comprising mostly of one complex I (CI) dimer and one CIII dimer. This phenotypic similarity along with lentiviral rescue experiments in patient fibroblasts verifies the pathogenicity of the shared genetic defect, demonstrating that the Uqcrh-/- mouse is a valuable model for future studies of human CIII deficiency.


Subject(s)
Mitochondrial Diseases , Animals , Electron Transport Complex III , Exons , Homozygote , Humans , Mice , Mitochondrial Diseases/genetics , Phenotype , Sequence Deletion
6.
Sci Adv ; 6(35): eaaz4551, 2020 08.
Article in English | MEDLINE | ID: mdl-32923617

ABSTRACT

Recently, covalent modifications of RNA, such as methylation, have emerged as key regulators of all aspects of RNA biology and have been implicated in numerous diseases, for instance, cancer. Here, we undertook a combination of in vitro and in vivo screens to test 78 potential methyltransferases for their roles in hepatocellular carcinoma (HCC) cell proliferation. We identified methyltransferase-like protein 6 (METTL6) as a crucial regulator of tumor cell growth. We show that METTL6 is a bona fide transfer RNA (tRNA) methyltransferase, catalyzing the formation of 3-methylcytidine at C32 of specific serine tRNA isoacceptors. Deletion of Mettl6 in mouse stem cells results in changes in ribosome occupancy and RNA levels, as well as impaired pluripotency. In mice, Mettl6 knockout results in reduced energy expenditure. We reveal a previously unknown pathway in the maintenance of translation efficiency with a role in maintaining stem cell self-renewal, as well as impacting tumor cell growth profoundly.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/genetics , Cell Proliferation , Liver Neoplasms/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , RNA , RNA, Transfer/genetics , RNA, Transfer/metabolism , tRNA Methyltransferases
7.
J Neurochem ; 111(6): 1369-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19804379

ABSTRACT

The Swedish mutation within the amyloid precursor protein (APP) causes early-onset Alzheimer's disease due to increased cleavage of APP by BACE1. While beta-secretase shedding of Swedish APP (APPswe) largely results from an activity localized in the late secretory pathway, cleavage of wild-type APP occurs mainly in endocytic compartments. However, we show that liberation of Abeta from APPswe is still dependent on functional internalization from the cell surface. Inspite the unchanged overall beta-secretase cleaved soluble APP released from APP(swe) secretion, mutations of the APPswe internalization motif strongly reduced C99 levels and substantially decreased Abeta secretion. We point out that alpha-secretase activity-mediated conversion of C99 to C83 is the main cause of this Abeta reduction. Furthermore, we demonstrate that alpha-secretase cleavage of C99 even contributes to the reduction of Abeta secretion of internalization deficient wild-type APP. Therefore, inhibition of alpha-secretase cleavage increased Abeta secretion through diminished conversion of C99 to C83 in APP695, APP695swe or C99 expressing cells.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Biotinylation/methods , CHO Cells , Cricetinae , Cricetulus , Gene Expression Regulation/genetics , Humans , Mutation/genetics , Peptide Fragments/genetics , Protein Interaction Domains and Motifs/physiology , Transfection
8.
Neurodegener Dis ; 6(1-2): 1-8, 2009.
Article in English | MEDLINE | ID: mdl-18349521

ABSTRACT

BACKGROUND: Epidemiological studies have suggested that long-term use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with a reduced incidence of Alzheimer's disease (AD). Several mechanisms have been proposed to explain these findings including increased shedding of the soluble ectodomain of the amyloid precursor protein (sAPP), which functions as a neurotrophic and neuroprotective factor in vitroand in vivo. OBJECTIVE: To clarify whether NSAIDs consistently stimulate sAPP secretion. METHODS: 293-EBNA cells with stable overexpression of an APP-alkaline phosphatase fusion protein (APP-AP), SH-SY5Y and PC12 cells or primary telencephalic chicken neurons were treated with ibuprofen or indomethacin. APP shedding was then determined by measuring AP activity in conditioned media, Western blot analysis with antibodies against total sAPP or specific for sAPP-alpha, or in a pulse-chase paradigm. RESULTS: AP activity in conditioned media was not increased after NSAID treatment of 293-EBNA cells whereas it was elevated by phorbol ester. Surprisingly, ibuprofen or indomethacin treatment of SH-SY5Y and PC12 cells expressing endogenous APP did not cause changes in sAPP or sAPP-alpha secretion or downregulation of cellular APP. These findings were further corroborated in primary chicken neuronal cultures. CONCLUSIONS: Using various experimental settings, we were unable to confirm sAPP or sAPP-alpha stimulation with the NSAIDs ibuprofen and indomethacin in transfected and nontransfected cells of neuronal and nonneuronal origin. Importantly, these findings seem to rule out chronic sAPP stimulation as an alternative mechanism of NSAID action in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Alkaline Phosphatase/adverse effects , Amyloid beta-Protein Precursor/drug effects , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Ibuprofen/pharmacology , Indomethacin/pharmacology , Kinetics , Neuroblastoma , PC12 Cells/drug effects , Rats , Tetradecanoylphorbol Acetate/pharmacology
9.
Behav Brain Res ; 352: 187-196, 2018 10 15.
Article in English | MEDLINE | ID: mdl-28966146

ABSTRACT

Since decades, model organisms have provided an important approach for understanding the mechanistic basis of human diseases. The German Mouse Clinic (GMC) was the first phenotyping facility that established a collaboration-based platform for phenotype characterization of mouse lines. In order to address individual projects by a tailor-made phenotyping strategy, the GMC advanced in developing a series of pipelines with tests for the analysis of specific disease areas. For a general broad analysis, there is a screening pipeline that covers the key parameters for the most relevant disease areas. For hypothesis-driven phenotypic analyses, there are thirteen additional pipelines with focus on neurological and behavioral disorders, metabolic dysfunction, respiratory system malfunctions, immune-system disorders and imaging techniques. In this article, we give an overview of the pipelines and describe the scientific rationale behind the different test combinations.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Phenotype , Animals , Humans
10.
Nat Commun ; 9(1): 288, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348434

ABSTRACT

Metabolic diseases are a worldwide problem but the underlying genetic factors and their relevance to metabolic disease remain incompletely understood. Genome-wide research is needed to characterize so-far unannotated mammalian metabolic genes. Here, we generate and analyze metabolic phenotypic data of 2016 knockout mouse strains under the aegis of the International Mouse Phenotyping Consortium (IMPC) and find 974 gene knockouts with strong metabolic phenotypes. 429 of those had no previous link to metabolism and 51 genes remain functionally completely unannotated. We compared human orthologues of these uncharacterized genes in five GWAS consortia and indeed 23 candidate genes are associated with metabolic disease. We further identify common regulatory elements in promoters of candidate genes. As each regulatory element is composed of several transcription factor binding sites, our data reveal an extensive metabolic phenotype-associated network of co-regulated genes. Our systematic mouse phenotype analysis thus paves the way for full functional annotation of the genome.


Subject(s)
Basal Metabolism/genetics , Blood Glucose/metabolism , Body Weight/genetics , Diabetes Mellitus, Type 2/genetics , Obesity/genetics , Oxygen Consumption/genetics , Triglycerides/metabolism , Animals , Area Under Curve , Gene Regulatory Networks , Genome-Wide Association Study , High-Throughput Screening Assays , Humans , Metabolic Diseases/genetics , Mice , Mice, Knockout , Phenotype
11.
G3 (Bethesda) ; 6(12): 4035-4046, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27815347

ABSTRACT

The vertebrate Scube (Signal peptide, CUB, and EGF-like domain-containing protein) family consists of three independent members, Scube1-3, which encode secreted cell surface-associated membrane glycoproteins. Limited information about the general function of this gene family is available, and their roles during adulthood. Here, we present the first Scube3 mutant mouse line (Scube3N294K/N294K), which clearly shows phenotypic alterations by carrying a missense mutation in exon 8, and thus contributes to our understanding of SCUBE3 functions. We performed a detailed phenotypic characterization in the German Mouse Clinic (GMC). Scube3N294K/N294K mutants showed morphological abnormalities of the skeleton, alterations of parameters relevant for bone metabolism, changes in renal function, and hearing impairments. These findings correlate with characteristics of the rare metabolic bone disorder Paget disease of bone (PDB), associated with the chromosomal region of human SCUBE3 In addition, alterations in energy metabolism, behavior, and neurological functions were detected in Scube3N294K/N294K mice. The Scube3N294K/N294K mutant mouse line may serve as a new model for further studying the effect of impaired SCUBE3 gene function.


Subject(s)
Genetic Association Studies , Glycoproteins/genetics , Mutation , Phenotype , Animals , Bone and Bones/metabolism , Calcium-Binding Proteins , Chromosome Mapping , Disease Models, Animal , Energy Metabolism/genetics , Exome , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Kidney/metabolism , Kidney/physiopathology , Kidney Function Tests , Male , Mice , Mice, Knockout , Osteitis Deformans/genetics , Osteitis Deformans/metabolism , Osteitis Deformans/pathology , Skeleton/abnormalities
12.
ChemMedChem ; 3(1): 165-72, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17943713

ABSTRACT

Curcumin binds to the amyloid beta peptide (Abeta) and inhibits or modulates amyloid precursor protein (APP) metabolism. Therefore, curcumin-derived isoxazoles and pyrazoles were synthesized to minimize the metal chelation properties of curcumin. The decreased rotational freedom and absence of stereoisomers was predicted to enhance affinity toward Abeta(42) aggregates. Accordingly, replacement of the 1,3-dicarbonyl moiety with isosteric heterocycles turned curcumin analogue isoxazoles and pyrazoles into potent ligands of fibrillar Abeta(42) aggregates. Additionally, several compounds are potent inhibitors of tau protein aggregation and depolymerized tau protein aggregates at low micromolar concentrations.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Proliferation/drug effects , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Isoxazoles/pharmacology , Pyrazoles/pharmacology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Chickens , Curcumin/analogs & derivatives , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Humans , Inhibitory Concentration 50 , Isoxazoles/chemistry , Magnetic Resonance Spectroscopy , Pyrazoles/chemistry , Radioligand Assay , tau Proteins/metabolism
13.
J Biol Chem ; 283(25): 17049-54, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18426795

ABSTRACT

Proteolytic processing of the amyloid precursor protein by beta- and gamma-secretase generates the amyloid-beta (Abeta) peptides, which are principal drug targets in Alzheimer disease therapeutics. gamma-Secretase has imprecise cleavage specificity and generates the most abundant Abeta40 and Abeta42 species together with longer and shorter peptides such as Abeta38. Several mechanisms could explain the production of multiple Abeta peptides by gamma-secretase, including sequential processing of longer into shorter Abeta peptides. A novel class of gamma-secretase modulators (GSMs) that includes some non-steroidal anti-inflammatory drugs has been shown to selectively lower Abeta42 levels without a change in Abeta40 levels. A signature of GSMs is the concomitant increase in shorter Abeta peptides, such as Abeta38, leading to the suggestion that generation of Abeta42 and Abeta38 peptide species by gamma-secretase is coordinately regulated. However, no evidence for or against such a precursor-product relationship has been provided. We have previously shown that stable overexpression of aggressive presenilin-1 (PS1) mutations associated with early-onset familial Alzheimer disease attenuated the cellular response to GSMs, resulting in greatly diminished Abeta42 reductions as compared with wild type PS1. We have now used this model system to investigate whether Abeta38 production would be similarly affected indicating coupled generation of Abeta42 and Abeta38 peptides. Surprisingly, treatment with the GSM sulindac sulfide increased Abeta38 production to similar levels in four different PS1 mutant cell lines as compared with wild type PS1 cells. This was confirmed with the structurally divergent GSMs ibuprofen and indomethacin. Mass spectrometry analysis and high resolution urea gel electrophoresis further demonstrated that sulindac sulfide did not induce detectable compensatory changes in levels of other Abeta peptide species. These data provide evidence that Abeta42 and Abeta38 species can be independently generated by gamma-secretase and argue against a precursor-product relationship between these peptides.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Ibuprofen/pharmacology , Molecular Sequence Data , Mutation , Presenilin-1/genetics , Sulindac/analogs & derivatives , Sulindac/pharmacology
14.
J Biol Chem ; 282(34): 24504-13, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17573346

ABSTRACT

Abeta42-lowering nonsteroidal anti-inflammatory drugs (NSAIDs) constitute the founding members of a new class of gamma-secretase modulators that avoid side effects of pan-gamma-secretase inhibitors on NOTCH processing and function, holding promise as potential disease-modifying agents for Alzheimer disease (AD). These modulators are active in cell-free gamma-secretase assays indicating that they directly target the gamma-secretase complex. Additional support for this hypothesis was provided by the observation that certain mutations in presenilin-1 (PS1) associated with early-onset familial AD (FAD) change the cellular drug response to Abeta42-lowering NSAIDs. Of particular interest is the PS1-DeltaExon9 mutation, which provokes a pathogenic increase in the Abeta42/Abeta40 ratio and dramatically reduces the cellular response to the Abeta42-lowering NSAID sulindac sulfide. This FAD PS1 mutant is unusual as a splice-site mutation results in deletion of amino acids Thr(291)-Ser(319) including the endoproteolytic cleavage site of PS1, and an additional amino acid exchange (S290C) at the exon 8/10 splice junction. By genetic dissection of the PS1-DeltaExon9 mutation, we now demonstrate that a synergistic effect of the S290C mutation and the lack of endoproteolytic cleavage is sufficient to elevate the Abeta42/Abeta40 ratio and that the attenuated response to sulindac sulfide results partially from the deficiency in endoproteolysis. Importantly, a wider screen revealed that a diminished response to Abeta42-lowering NSAIDs is common among aggressive FAD PS1 mutations. Surprisingly, these mutations were also partially unresponsive to gamma-secretase inhibitors of different structural classes. This was confirmed in a mouse model with transgenic expression of the PS1-L166P mutation, in which the potent gamma-secretase inhibitor LY-411575 failed to reduce brain levels of soluble Abeta42. In summary, these findings highlight the importance of genetic background in drug discovery efforts aimed at gamma-secretase, suggesting that certain AD mouse models harboring aggressive PS mutations may not be informative in assessing in vivo effects of gamma-secretase modulators and inhibitors.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Mutation , Peptide Fragments/metabolism , Peptide Fragments/physiology , Presenilin-1/genetics , Presenilin-1/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Amino Acid
15.
Curr Pharm Des ; 12(33): 4337-55, 2006.
Article in English | MEDLINE | ID: mdl-17105431

ABSTRACT

The amyloid-beta (Abeta) peptides and in particular the longer, highly amyloidogenic isoform Abeta42 are believed by many to be the central disease-causing agents in Alzheimer's disease (AD). Consequently, academic and pharmaceutical laboratories have focused on elucidating the mechanisms of Abeta production and developing strategies to diminish Abeta formation for treatment or prevention of AD. The most substantial advances have been made with respect to inhibitors of the gamma-secretase enzyme, which catalyzes the final step in the generation of Abeta from the amyloid precursor protein (APP). Highly potent gamma-secretase inhibitors which suppress production of all Abeta peptides are available today. However, due to the promiscuous substrate specificity of gamma-secretase and its essential role in the NOTCH signaling pathway overt mechanism-based toxicity has been observed in preclinical studies of gamma-secretase inhibitors. For that reason, specific blockage of Abeta42 production might be preferable over non-discriminatory gamma-secretase inhibition but small molecule inhibitors of Abeta42 production have remained elusive until recently. This has changed with the discovery that certain non-steroidal anti-inflammatory drugs (NSAIDs) including ibuprofen possess preferential Abeta42-lowering activity. These compounds seem to offer a window of modulation where Abeta42 production is potently inhibited whereas processing of the NOTCH receptor and other gamma-secretase substrates remains unaffected. The Abeta42-lowering activity of NSAIDs is not related to inhibition of cyclooxygenases and can be dissociated from the anti-inflammatory properties of this class of drugs. Ongoing efforts concentrate on uncovering the mechanism of action and improving potency and brain permeability of Abeta42-lowering compounds. Hopes are high that in the near future this will lead to the development of clinically viable compounds which selectively target Abeta42 as a key molecule in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/biosynthesis , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Peptide Fragments/biosynthesis , Alzheimer Disease/metabolism , Humans
16.
J Neurochem ; 96(2): 355-65, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16300630

ABSTRACT

Certain non-steroidal anti-inflammatory drugs (NSAIDs) preferentially inhibit production of the amyloidogenic Abeta42 peptide, presumably by direct modulation of gamma-secretase activity. A recent report indicated that NSAIDs could reduce Abeta42 by inhibition of the small GTPase Rho, and a single inhibitor of Rho kinase (ROCK) mimicked the effects of Abeta42-lowering NSAIDs. To investigate whether Abeta42 reduction is a common property of ROCK inhibitors, we tested commercially available compounds in cell lines that were previously used to demonstrate the Abeta42-lowering activity of NSAIDs. Surprisingly, we found that two ROCK inhibitors reduced total Abeta secretion in a dose-dependent manner but showed no selectivity for Abeta42. In addition, ROCK inhibitors did not increase Abeta38 secretion in cell-based assays or reduce Abeta production in gamma-secretase in vitro assays, which are critical characteristics of Abeta42-lowering NSAIDs. The reduction in total Abeta levels by ROCK inhibitors was not accompanied by overall-changes in amyloid precursor protein processing. Targeting ROCK by expression of dominant-negative or constitutively active ROCK mutants failed to modulate Abeta secretion, indicating that ROCK inhibition may either be redundant or insufficient for Abeta reduction by ROCK inhibitors. Taken together, these results seem to exclude a mechanistic involvement of ROCK in the Abeta42-lowering activity of NSAIDs.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Peptide Fragments/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , Endopeptidases/metabolism , Enzyme Inhibitors/poisoning , Intracellular Signaling Peptides and Proteins , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL