Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Kidney Int ; 105(6): 1263-1278, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38286178

ABSTRACT

Current classification of chronic kidney disease (CKD) into stages using indirect systemic measures (estimated glomerular filtration rate (eGFR) and albuminuria) is agnostic to the heterogeneity of underlying molecular processes in the kidney thereby limiting precision medicine approaches. To generate a novel CKD categorization that directly reflects within kidney disease drivers we analyzed publicly available transcriptomic data from kidney biopsy tissue. A Self-Organizing Maps unsupervised artificial neural network machine-learning algorithm was used to stratify a total of 369 patients with CKD and 46 living kidney donors as healthy controls. Unbiased stratification of the discovery cohort resulted in identification of four novel molecular categories of disease termed CKD-Blue, CKD-Gold, CKD-Olive, CKD-Plum that were replicated in independent CKD and diabetic kidney disease datasets and can be further tested on any external data at kidneyclass.org. Each molecular category spanned across CKD stages and histopathological diagnoses and represented transcriptional activation of distinct biological pathways. Disease progression rates were highly significantly different between the molecular categories. CKD-Gold displayed rapid progression, with significant eGFR-adjusted Cox regression hazard ratio of 5.6 [1.01-31.3] for kidney failure and hazard ratio of 4.7 [1.3-16.5] for composite of kidney failure or a 40% or more eGFR decline. Urine proteomics revealed distinct patterns between the molecular categories, and a 25-protein signature was identified to distinguish CKD-Gold from other molecular categories. Thus, patient stratification based on kidney tissue omics offers a gateway to non-invasive biomarker-driven categorization and the potential for future clinical implementation, as a key step towards precision medicine in CKD.


Subject(s)
Disease Progression , Glomerular Filtration Rate , Kidney , Precision Medicine , Renal Insufficiency, Chronic , Transcriptome , Humans , Precision Medicine/methods , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/urine , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/physiopathology , Middle Aged , Female , Male , Kidney/pathology , Kidney/physiopathology , Aged , Biopsy , Adult , Neural Networks, Computer , Case-Control Studies , Gene Expression Profiling , Unsupervised Machine Learning
2.
Cardiovasc Res ; 118(2): 517-530, 2022 01 29.
Article in English | MEDLINE | ID: mdl-33705529

ABSTRACT

AIMS: Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS: Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION: This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Enzyme Inhibitors/pharmacology , Hypertrophy, Left Ventricular/drug therapy , Induced Pluripotent Stem Cells/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Peroxidase/antagonists & inhibitors , Ventricular Function, Left/drug effects , Animals , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cardiomyopathy, Hypertrophic/enzymology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Disease Models, Animal , Humans , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Male , Mice, Inbred C57BL , Mutation, Missense , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Peroxidase/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
3.
Metabolism ; 95: 27-35, 2019 06.
Article in English | MEDLINE | ID: mdl-30902749

ABSTRACT

BACKGROUND AND PURPOSE: In addition to the central nervous system-mediated action, leptin also directly induces fatty acid oxidation in skeletal muscle. Rapid induction of FAO by leptin is mediated by the AMP-activated protein kinase (AMPK) pathway, but the mechanism of prolonged FAO by leptin was previously unknown. In an earlier study, we showed that free fatty acids increase transcription of small ubiquitin-like modifier (SUMO) specific protease 2 (SENP2) in skeletal muscle, and that SENP2 stimulates expression of FAO-associated enzymes by deSUMOylating peroxisome proliferator-activated receptors, PPARδ and PPARγ. In this study, we examine whether SENP2 is involved in prolonged stimulation of FAO by leptin. METHODS: The Effect of leptin on expression of SENP2 and on SENP2-mediated FAO was investigated by using western blotting and real time qPCR of C2C12 myotubes, and of C2C12 myotubes in which expression of specific genes was knocked down using siRNAs. Additionally, muscle-specific SENP2 knockout mice were generated to test the involvement of SENP2 in leptin-induced FAO in vivo. RESULTS: We show that leptin treatment of C2C12 myotubes causes signal transducer and activator of transcription 3 (STAT3) to bind to the Senp2 promoter, inducing SENP2 expression. We also show that leptin increases the binding of PPARδ and PPARγ to PPRE sites in the promoters of two FAO-associated genes: long-chain acyl-CoA synthetase 1 (Acsl1) or carnitine palmitoyl transferase 1b (Cpt1b). When SENP2 is knocked down in myotubes, leptin-induced expression of FAO-associated enzymes and prolonged increase of FAO are suppressed, but rapid increase of FAO is unaffected. In addition, leptin-induced expression of FAO-associated enzymes was not observed in muscle tissue of SENP2 knockout mice. CONCLUSIONS: We demonstrate that the peripheral actions of leptin on FAO are mediated by two different pathways: AMPK causes a rapid increase in FAO, and SENP2 of the STAT3 pathway causes a slow, prolonged increase in FAO.


Subject(s)
Cysteine Endopeptidases/metabolism , Fatty Acids/metabolism , Leptin/pharmacology , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Gene Knockdown Techniques , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Oxidation-Reduction
4.
Int J Cardiol ; 272: 288-297, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30177232

ABSTRACT

BACKGROUND: Preferential utilization of fatty acids for ATP production represents an advanced metabolic phenotype in developing cardiomyocytes. We investigated whether this phenotype could be attained in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) and assessed its influence on mitochondrial morphology, bioenergetics, respiratory capacity and ultra-structural architecture. METHODS AND RESULTS: Whole-cell proteome analysis of day 14 and day 30-CMs maintained in glucose media revealed a positive influence of extended culture on mitochondria-related processes that primed the day 30-CMs for fatty acid metabolism. Supplementing the day 30-CMs with palmitate/oleate (fatty acids) significantly enhanced mitochondrial remodeling, oxygen consumption rates and ATP production. Metabolomic analysis upon fatty acid supplementation revealed a ß-oxidation fueled ATP elevation that coincided with presence of junctional complexes, intercalated discs, t-tubule-like structures and adult isoform of cardiac troponin T. In contrast, glucose-maintained day 30-CMs continued to harbor underdeveloped ultra-structural architecture and more subdued bioenergetics, constrained by suboptimal mitochondria development. CONCLUSION: The advanced metabolic phenotype of preferential fatty acid utilization was attained in hiPSC-CMs, whereby fatty acid driven ß-oxidation sustained cardiac bioenergetics and respiratory capacity resulting in ultra-structural and functional characteristics similar to those of developmentally advanced cardiomyocytes. Better understanding of mitochondrial bioenergetics and ultra-structural adaptation associated with fatty acid metabolism has important implications in the study of cardiac physiology that are associated with late-onset mitochondrial and metabolic adaptations.


Subject(s)
Energy Metabolism/physiology , Fatty Acids/metabolism , Induced Pluripotent Stem Cells/metabolism , Lipid Metabolism/physiology , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/ultrastructure , Mitochondria/ultrastructure , Myocytes, Cardiac/ultrastructure , Phenotype
5.
J Alzheimers Dis ; 64(s1): S547-S554, 2018.
Article in English | MEDLINE | ID: mdl-29758944

ABSTRACT

Spanning over three decades of extensive drug discovery research, the efforts to develop a potent and selective GSK3 inhibitor as a therapeutic for the treatment of type 2 diabetes, Alzheimer's disease (AD), bipolar disorders and cancer have been futile. Since its initial discovery in 1980 and subsequent decades of research, one cannot underscore the importance of the target and the promise of a game changing disease modifier. Several pharmaceutical companies, biotech companies, and academic institutions raged in a quest to unravel the biology and discover potent and selective GSK3 inhibitors, some of which went through clinical trials. However, the conundrum of what happened to the fate of the AstraZeneca's GSK3 inhibitors and the undertaking to find a therapeutic that could control glycogen metabolism and aberrant tau hyperphosphorylation in the brain, and rescue synaptic dysfunction has largely been untold. AstraZeneca was in the forefront of GSK3 drug discovery research with six GSK3 drug candidates, one of which progressed up to Phase II clinical trials in the quest to untangle the tau hypothesis for AD. Analysis of key toxicity issues, serendipitous findings and efficacy, and biomarker considerations in relation to safety margins have limited the potential of small molecule therapeutics as a way forward. To guide future innovation of this important target, we reveal the roller coaster journey comprising of two decades of preclinical and clinical GSK3 drug discovery at AstraZeneca; the understanding of which could lead to improved GSK3 therapies for disease. These learnings in combination with advances in achieving kinase selectivity, different modes of action as well as the recent discovery of novel conjugated peptide technology targeting specific tissues have potentially provided a venue for scientific innovation and a new beginning for GSK3 drug discovery.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Clinical Trials as Topic , Glycogen Synthase Kinase 3/metabolism , Humans
6.
JAMA Cardiol ; 3(6): 481-488, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29641836

ABSTRACT

Importance: The genetic variant MYBPC3Δ25bp occurs in 4% of South Asian descendants, with an estimated 100 million carriers worldwide. MYBPC3 Δ25bp has been linked to cardiomyopathy and heart failure. However, the high prevalence of MYBPC3Δ25bp suggests that other stressors act in concert with MYBPC3Δ25bp. Objective: To determine whether there are additional genetic factors that contribute to the cardiomyopathic expression of MYBPC3Δ25bp. Design, Setting, andParticipants: South Asian individuals living in the United States were screened for MYBPC3Δ25bp, and a subgroup was clinically evaluated using electrocardiograms and echocardiograms at Loyola University, Chicago, Illinois, between January 2015 and July 2016. Main Outcomes and Measures: Next-generation sequencing of 174 cardiovascular disease genes was applied to identify additional modifying gene mutations and correlate genotype-phenotype parameters. Cardiomyocytes derived from human-induced pluripotent stem cells were established and examined to assess the role of MYBPC3Δ25bp. Results: In this genotype-phenotype study, individuals of South Asian descent living in the United States from both sexes (36.23% female) with a mean population age of 48.92 years (range, 18-84 years) were recruited. Genetic screening of 2401 US South Asian individuals found an MYBPC3Δ25bpcarrier frequency of 6%. A higher frequency of missense TTN variation was found in MYBPC3Δ25bp carriers compared with noncarriers, identifying distinct genetic backgrounds within the MYBPC3Δ25bp carrier group. Strikingly, 9.6% of MYBPC3Δ25bp carriers also had a novel MYBPC3 variant, D389V. Family studies documented D389V was in tandem on the same allele as MYBPC3Δ25bp, and D389V was only seen in the presence of MYBPC3Δ25bp. In contrast to MYBPC3Δ25bp, MYBPC3Δ25bp/D389V was associated with hyperdynamic left ventricular performance (mean [SEM] left ventricular ejection fraction, 66.7 [0.7%]; left ventricular fractional shortening, 36.6 [0.6%]; P < .03) and stem cell-derived cardiomyocytes exhibited cellular hypertrophy with abnormal Ca2+ transients. Conclusions and Relevance: MYBPC3Δ25bp/D389V is associated with hyperdynamic features, which are an early finding in hypertrophic cardiomyopathy and thought to reflect an unfavorable energetic state. These findings support that a subset of MYBPC3Δ25bp carriers, those with D389V, account for the increased risk attributed to MYBPC3Δ25bp.


Subject(s)
Asian/genetics , Cardiomyopathy, Hypertrophic/ethnology , Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/genetics , Mutation/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cardiomyopathy, Hypertrophic/physiopathology , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Stroke Volume , Young Adult
7.
J Am Heart Assoc ; 6(6)2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28611096

ABSTRACT

BACKGROUND: Thymosin beta-4 (TB4) is an X-linked gene product with cardioprotective properties. Little is known about plasma concentration of TB4 in heart failure (HF), and its relationship with other cardiovascular biomarkers. We sought to evaluate circulating TB4 in HF patients with preserved (HFpEF) or reduced (HFrEF) ejection fraction compared to non-HF controls. METHODS AND RESULTS: TB4 was measured using a liquid chromatography and mass spectrometry assay in age- and sex-matched HFpEF (n=219), HFrEF (n=219) patients, and controls (n=219) from a prospective nationwide study. Additionally, a 92-marker multiplex proximity extension assay was measured to identify biomarker covariates. Compared with controls, plasma TB4 was elevated in HFpEF (985 [421-1723] ng/mL versus 1401 [720-2379] ng/mL, P<0.001), but not in HFrEF (1106 [556-1955] ng/mL, P=0.642). Stratifying by sex, only women (1623 [1040-2625] ng/mL versus 942 [386-1891] ng/mL, P<0.001), but not men (1238.5 [586-1967] ng/mL versus 1004 [451-1538] ng/mL, P=1.0), had significantly elevated TB4 in the setting of HFpEF. Adjusted for New York Heart Association class, N-terminal pro B-type natriuretic peptide, age, and myocardial infarction, hazard ratio to all-cause mortality is significantly higher in women with elevated TB4 (1.668, P=0.036), but not in men (0.791, P=0.456) with HF. TB4 is strongly correlated with a cluster of 7 markers from the proximity extension assay panel, which are either X-linked, regulated by sex hormones, or involved with NF-κB signaling. CONCLUSIONS: We show that plasma TB4 is elevated in women with HFpEF and has prognostic information. Because TB4 can preserve EF in animal studies of cardiac injury, the relation of endogenous, circulating TB4 to X chromosome biology and differential outcomes in female heart disease warrants further study.


Subject(s)
Heart Failure/blood , Stroke Volume/physiology , Thymosin/blood , Aged , Biomarkers/blood , Chromatography, Liquid , Disease Progression , Female , Follow-Up Studies , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Male , Mass Spectrometry , Microfilament Proteins , Middle Aged , Prognosis , Prospective Studies , Sex Factors
8.
Alzheimers Dement ; 9(4): 452-458.e1, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23809366

ABSTRACT

For decades, researchers have focused primarily on a pathway initiated by amyloid beta aggregation, amyloid deposition, and accumulation in the brain as the key mechanism underlying the disease and the most important treatment target. However, evidence increasingly suggests that amyloid is deposited early during the course of disease, even prior to the onset of clinical symptoms. Thus, targeting amyloid in patients with mild to moderate Alzheimer's disease (AD), as past failed clinical trials have done, may be insufficient to halt further disease progression. Scientists are investigating other molecular and cellular pathways and processes that contribute to AD pathogenesis. Thus, the Alzheimer's Association's Research Roundtable convened a meeting in April 2012 to move beyond amyloid and explore AD as a complex multifactorial disease, with the goal of using a more inclusive perspective to identify novel treatment strategies.


Subject(s)
Alzheimer Disease/drug therapy , Molecular Targeted Therapy , Nootropic Agents/therapeutic use , Aging , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Autophagy/drug effects , Biomarkers , Brain/metabolism , Cell Cycle/drug effects , Cooperative Behavior , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Inflammation , Insulin Resistance , Lysosomes/drug effects , Lysosomes/physiology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/pathology , Models, Neurological , Neuroimaging , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nootropic Agents/pharmacology , Public-Private Sector Partnerships , Resource Allocation , tau Proteins/drug effects , tau Proteins/physiology
9.
J Neurochem ; 125(3): 446-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23410232

ABSTRACT

Abnormal tau phosphorylation resulting in detachment of tau from microtubules and aggregation are critical events in neuronal dysfunction, degeneration, and neurofibrillary pathology seen in Alzheimer's disease. Glycogen synthase kinase-3ß (GSK3ß) is a key target for drug discovery in the treatment of Alzheimer's disease and related tauopathies because of its potential to abnormally phosphorylate proteins and contribute to synaptic degeneration. We report the discovery of AZD1080, a potent and selective GSK3 inhibitor that demonstrates peripheral target engagement in Phase 1 clinical studies. AZD1080 inhibits tau phosphorylation in cells expressing human tau and in intact rat brain. Interestingly, subchronic but not acute administration with AZD1080 reverses MK-801-induced deficits, measured by long-term potentiation in hippocampal slices and in a cognitive test in mice, suggesting that reversal of synaptic plasticity deficits in dysfunctional systems requires longer term modifications of proteins downstream of GSK3ß signaling. The inhibitory pattern on tau phosphorylation reveals a prolonged pharmacodynamic effect predicting less frequent dosing in humans. Consistent with the preclinical data, in multiple ascending dose studies in healthy volunteers, a prolonged suppression of glycogen synthase activity was observed in blood mononuclear cells providing evidence of peripheral target engagement with a selective GSK3 inhibitor in humans.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Long-Term Potentiation/drug effects , tau Proteins/metabolism , Animals , Cell Line, Transformed , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Crystallography , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Double-Blind Method , Electric Stimulation , Enzyme Inhibitors/chemistry , Excitatory Amino Acid Antagonists/toxicity , Excitatory Postsynaptic Potentials/drug effects , Glycogen Synthase/metabolism , Glycogen Synthase Kinase 3/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Humans , In Vitro Techniques , Indoles/pharmacology , Indoles/therapeutic use , Leukocytes, Mononuclear/drug effects , Long-Term Potentiation/physiology , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinases/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
10.
J Med Chem ; 55(21): 9107-19, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22489897

ABSTRACT

Glycogen synthase kinase-3ß, also called tau phosphorylating kinase, is a proline-directed serine/threonine kinase which was originally identified due to its role in glycogen metabolism. Active forms of GSK3ß localize to pretangle pathology including dystrophic neuritis and neurofibrillary tangles in Alzheimer's disease (AD) brain. By using a high throughput screening (HTS) approach to search for new chemical series and cocrystallization of key analogues to guide the optimization and synthesis of our pyrazine series, we have developed highly potent and selective inhibitors showing cellular efficacy and blood-brain barrier penetrance. The inhibitors are suitable for in vivo efficacy testing and may serve as a new treatment strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Glycogen Synthase Kinase 3/antagonists & inhibitors , Pyrazines/chemical synthesis , 3T3 Cells , Animals , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cattle , Crystallography, X-Ray , Drug Design , Glycogen Synthase Kinase 3 beta , Humans , Mice , Models, Molecular , Molecular Structure , Permeability , Phosphorylation , Pyrazines/chemistry , Pyrazines/pharmacology , Solubility , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology , tau Proteins/metabolism
11.
J Biol Chem ; 286(36): 31153-8, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21768095

ABSTRACT

The γ-secretase complex is responsible for intramembrane processing of over 60 substrates and is involved in Notch signaling as well as in the generation of the amyloid ß-peptide (Aß). Aggregated forms of Aß have a pathogenic role in Alzheimer disease and, thus, reducing the Aß levels by inhibiting γ-secretase is a possible treatment strategy for Alzheimer disease. Regrettably, clinical trials have shown that inhibition of γ-secretase results in Notch-related side effects. Therefore, it is of great importance to find ways to inhibit amyloid precursor protein (APP) processing without disturbing vital signaling pathways such as Notch. Nicastrin (Nct) is part of the γ-secretase complex and has been proposed to be involved in substrate recognition and selection. We have investigated how the four evenly spaced and conserved cysteine residues in the Nct ectodomain affect APP and Notch processing. We mutated these cysteines to serines and analyzed them in cells lacking endogenous Nct. We found that two mutants, C213S (C2) and C230S (C3), differentially affected APP and Notch processing. Both the formation of Aß and the intracellular domain of amyloid precursor protein (AICD) were reduced, whereas the production of Notch intracellular domain (NICD) was maintained on a high level, although C230S (C3) showed impaired complex assembly. Our data demonstrate that single residues in a γ-secretase component besides presenilin are able to differentially affect APP and Notch processing.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/biosynthesis , Membrane Glycoproteins/physiology , Mutation , Receptors, Notch/metabolism , Alzheimer Disease/drug therapy , Amino Acid Substitution , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cells, Cultured , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Signal Transduction/genetics
12.
Mol Brain ; 2: 22, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19583853

ABSTRACT

BACKGROUND: The signalling mechanisms involved in the induction of N-methyl-D-aspartate (NMDA) receptor-dependent long-term depression (LTD) in the hippocampus are poorly understood. Numerous studies have presented evidence both for and against a variety of second messengers systems being involved in LTD induction. Here we provide the first systematic investigation of the involvement of serine/threonine (ser/thr) protein kinases in NMDAR-LTD, using whole-cell recordings from CA1 pyramidal neurons. RESULTS: Using a panel of 23 inhibitors individually loaded into the recorded neurons, we can discount the involvement of at least 57 kinases, including PKA, PKC, CaMKII, p38 MAPK and DYRK1A. However, we have been able to confirm a role for the ser/thr protein kinase, glycogen synthase kinase 3 (GSK-3). CONCLUSION: The present study is the first to investigate the role of 58 ser/thr protein kinases in LTD in the same study. Of these 58 protein kinases, we have found evidence for the involvement of only one, GSK-3, in LTD.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Long-Term Synaptic Depression , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Long-Term Synaptic Depression/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats
13.
J Clin Invest ; 119(5): 1382-95, 2009 May.
Article in English | MEDLINE | ID: mdl-19363289

ABSTRACT

Our aging society is confronted with a dramatic increase of patients suffering from tauopathies, which include Alzheimer disease and certain frontotemporal dementias. These disorders are characterized by typical neuropathological lesions including hyperphosphorylation and subsequent aggregation of TAU protein and neuronal cell death. Currently, no mechanism-based cures are available. We generated fluorescently labeled TAU transgenic zebrafish, which rapidly recapitulated key pathological features of tauopathies, including phosphorylation and conformational changes of human TAU protein, tangle formation, neuronal and behavioral disturbances, and cell death. Due to their optical transparency and small size, zebrafish larvae are well suited for both in vivo imaging and drug development. TAU-induced neuronal cell death was imaged by time-lapse microscopy in vivo. Furthermore, we used this zebrafish model to identify compounds targeting the TAU kinase glycogen synthase kinase 3beta (GSK3beta). We identified a newly developed highly active GSK3beta inhibitor, AR-534, by rational drug design. AR-534 reduced TAU phosphorylation in TAU transgenic zebrafish. This transgenic zebrafish model may become a valuable tool for further studies of the neuropathology of dementia.


Subject(s)
Disease Models, Animal , Drug Evaluation, Preclinical/methods , Neurons/pathology , Tauopathies/drug therapy , Tauopathies/pathology , Zebrafish , Animals , Animals, Genetically Modified , Cell Death , Drug Design , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Escape Reaction , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Larva/anatomy & histology , Larva/drug effects , Larva/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Molecular , Molecular Structure , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Protein Conformation , Sequence Alignment , Spinal Cord/metabolism , Spinal Cord/pathology , Synaptotagmins/metabolism , Tauopathies/metabolism , Zebrafish/genetics , tau Proteins/genetics , tau Proteins/metabolism , Red Fluorescent Protein
14.
Proc Natl Acad Sci U S A ; 102(19): 6990-5, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15867159

ABSTRACT

Neurofibrillary tangles composed of hyperphosphorylated, aggregated tau are a common pathological feature of tauopathies, including Alzheimer's disease. Abnormal phosphorylation of tau by kinases or phosphatases has been proposed as a pathogenic mechanism in tangle formation. To investigate whether kinase inhibition can reduce tauopathy and the degeneration associated with it in vivo, transgenic mice overexpressing mutant human tau were treated with the glycogen synthase kinase-3 (GSK-3) inhibitor lithium chloride. Treatment resulted in significant inhibition of GSK-3 activity. Lithium administration also resulted in significantly lower levels of phosphorylation at several epitopes of tau known to be hyperphosphorylated in Alzheimer's disease and significantly reduced levels of aggregated, insoluble tau. Administration of a second GSK-3 inhibitor also correlated with reduced insoluble tau levels, supporting the idea that lithium exerts its effect through GSK-3 inhibition. Levels of aggregated tau correlated strongly with degree of axonal degeneration, and lithium-chloride-treated mice showed less degeneration if administration was started during early stages of tangle development. These results support the idea that kinases are involved in tauopathy progression and that kinase inhibitors may be effective therapeutically.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lithium Chloride/pharmacology , Animals , Disease Progression , Epitopes , Humans , Image Processing, Computer-Assisted , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Lithium/chemistry , Mice , Mice, Transgenic , Neurodegenerative Diseases/pathology , Neurons/pathology , Phosphorylation , Tauopathies , tau Proteins/chemistry
15.
Int J Neuropsychopharmacol ; 7(4): 387-90, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15315719

ABSTRACT

The mechanism by which lithium exerts either its anti-manic or antidepressant effects remains to be fully elucidated. Although lithium inhibits the enzyme glycogen synthase kinase-3 (GSK-3) at concentrations that are relevant for treatment of bipolar disorder, it is unclear whether GSK-3-related mechanisms are responsible for its therapeutic effects in the treatment of this disease. We report that AR-A014418 (a selective GSK-3 inhibitor) induces behavioural changes that are consistent with the effects of antidepressant medications. Subacute intraperitoneal injections of AR-A014418 reduced immobility time in rats exposed to the forced swim test, a well-established model for antidepressant efficacy. In addition, the specificity of this effect is supported by our finding that AR-A014418 decreased spontaneous as well as amphetamine-induced activity. Taken together, these data support the hypothesis that lithium may exert its antidepressant effects through inhibition of GSK-3, and that novel small-molecule GSK-3 inhibitors may be useful for the treatment of bipolar disorder and depression.


Subject(s)
Antidepressive Agents , Enzyme Inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Swimming/psychology , Thiazoles/pharmacology , Urea/analogs & derivatives , Urea/pharmacology , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
16.
J Neurochem ; 89(6): 1313-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15189333

ABSTRACT

Abstract Glycogen synthase kinase3 (GSK3) is emerging as a prominent drug target in the CNS. The most exciting of the possibilities of GSK3 lies within the treatment of Alzheimer's disease (AD) where abnormal increases in GSK3 levels and activity have been associated with neuronal death, paired helical filament tau formation and neurite retraction as well as a decline in cognitive performance. Abnormal activity of GSK3 is also implicated in stroke. Lithium, a widely used drug for affective disorders, inhibits GSK3 at therapeutically relevant concentrations. Thus while the rationale remains testable, pharmaceutical companies are investing in finding a selective inhibitor of GSK3. In the present review, we summarize the properties of GSK3, and discuss the potential for such a therapy in AD, and other CNS disorders.


Subject(s)
Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/enzymology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Insulin Resistance , Lithium/pharmacology , Neuronal Plasticity , Neurons/drug effects , Neurons/enzymology , Phosphorylation
17.
Brain Res ; 1007(1-2): 57-64, 2004 May 08.
Article in English | MEDLINE | ID: mdl-15064135

ABSTRACT

Tau is the main component of the paired helical filaments (PHFs), aberrant structures that develop in the brain of Alzheimer's disease (AD) patients and other tauopathies like frontotemporal dementia and parkinsonism associated to chromosome 17 (FTDP-17). Previous work has shown that tau overexpression in Sf9 insect cells results in the formation of long cytoplasmatic extensions as a consequence of microtubule stabilization and bundling. Throughout this work, we have taken studies in this system further by overexpression of an altered form of tau characteristic of FTDP-17, which includes three mutations (G272V, P301L and R406W) and biochemically behaves as a hyperphosphorylated form of the protein, with the aim of developing an in vitro model which would favour the formation of tau aggregates. Our results indicate that filaments resembling PHFs assemble when Sf9 cells overexpress FTDP-17 tau. The amount of these polymers is reduced in lithium treated cells which suggests that phosphorylation of FTDP-17 tau by GSK3 induces a conformational change favouring the formation of fibrillar polymers.


Subject(s)
Actin Cytoskeleton/metabolism , Alzheimer Disease/metabolism , tau Proteins/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Animals , Baculoviridae , Blotting, Western/methods , Cell Line/drug effects , Cell Line/ultrastructure , Cell Line/virology , Female , Fluorescent Antibody Technique/methods , Gene Expression Regulation , Humans , Insecta , Lithium/pharmacology , Microscopy, Immunoelectron/methods , Microtubule-Associated Proteins , Microtubules/drug effects , Microtubules/ultrastructure , Mutagenesis, Site-Directed , Mutation , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/ultrastructure , Ovary , Phosphorylation/drug effects , Transfection/methods , tau Proteins/genetics , tau Proteins/physiology , tau Proteins/ultrastructure
18.
J Biol Chem ; 279(5): 3801-6, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14602710

ABSTRACT

Tauopathies, including Alzheimer's disease, are neurodegenerative disorders in which tau protein accumulates as a consequence of alterations in its metabolism. At least three different types of alterations have been described; in some cases, an aberrant mRNA splicing of tau exon 10 occurs; in other cases, the disorder is a consequence of missense mutations and, in most cases, aberrant tau hyperphosphorylation takes place. Glycogen synthase kinase-3 (GSK-3) has emerged as a key kinase that is able to interact with several proteins involved in the etiology of Alzheimer's disease and other tauopathies. Here, we have evaluated whether GSK-3 is also able to modulate tau-mRNA splicing. Our data demonstrate that GSK-3 inhibition in cultured neurons affects tau splicing resulting in an increase in tau mRNA containing exon 10. Pre-mRNA splicing is catalyzed by a multimolecular complex including members of the serine/arginine-rich (SR) family of splicing factors. Immunofluorescence studies showed that after GSK-3 inhibition, SC35, a member of the SR family, is redistributed and enriched in nuclear speckles and colocalizes with the kinase. Furthermore, immunoprecipitated SC35 is phosphorylated by recombinant GSK-3beta. Phosphorylation of a peptide from the SR domain by GSK-3 revealed that the peptide needs to be prephosphorylated, suggesting the involvement of a priming kinase. Our results demonstrate that GSK-3 plays a crucial role in tau exon 10 splicing, raising the possibility that GSK3 could contribute to tauopathies via aberrant tau splicing.


Subject(s)
Alzheimer Disease/metabolism , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3/physiology , Nuclear Proteins/biosynthesis , RNA Splicing , Ribonucleoproteins , tau Proteins/genetics , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Animals , Arginine/chemistry , Cells, Cultured , Exons , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Mutation, Missense , Neurons/metabolism , Peptides/chemistry , Phosphorylation , Precipitin Tests , Protein Isoforms , RNA, Messenger/metabolism , Serine/chemistry , Serine-Arginine Splicing Factors , Time Factors , tau Proteins/biosynthesis
19.
J Biol Chem ; 278(46): 45937-45, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-12928438

ABSTRACT

Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase that has been implicated in pathological conditions such as diabetes and Alzheimer's disease. We report the characterization of a GSK3 inhibitor, AR-A014418, which inhibits GSK3 (IC50 = 104 +/- 27 nM), in an ATP-competitive manner (Ki = 38 nM). AR-A014418 does not significantly inhibit cdk2 or cdk5 (IC50 > 100 microM) or 26 other kinases demonstrating high specificity for GSK3. We report the co-crystallization of AR-A014418 with the GSK3beta protein and provide a description of the interactions within the ATP pocket, as well as an understanding of the structural basis for the selectivity of AR-A014418. AR-A014418 inhibits tau phosphorylation at a GSK3-specific site (Ser-396) in cells stably expressing human four-repeat tau protein. AR-A014418 protects N2A neuroblastoma cells against cell death mediated by inhibition of the phosphatidylinositol 3-kinase/protein kinase B survival pathway. Furthermore, AR-A014418 inhibits neurodegeneration mediated by beta-amyloid peptide in hippocampal slices. AR-A014418 may thus have important applications as a tool to elucidate the role of GSK3 in cellular signaling and possibly in Alzheimer's disease. AR-A014418 is the first compound of a family of specific inhibitors of GSK3 that does not significantly inhibit closely related kinases such as cdk2 or cdk5.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Thiazoles/chemistry , Thiazoles/metabolism , Urea/chemistry , Urea/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , CDC2-CDC28 Kinases/metabolism , Cell Death , Cell Line, Tumor , Cell Survival , Crystallography, X-Ray , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Electrons , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Models, Chemical , Models, Molecular , NIH 3T3 Cells , Neurons/metabolism , Peptides/chemistry , Protein Binding , Signal Transduction , Urea/analogs & derivatives , tau Proteins/chemistry
20.
J Pharmacol Exp Ther ; 304(1): 433-40, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490620

ABSTRACT

Caspase-3 is an intracellular cysteine protease, activated as part of the apoptotic response to cell injury. Its interest as a therapeutic target has led many to pursue the development of inhibitors. To date, only one series of nonpeptidic inhibitors have been described, and these have limited selectivity within the caspase family. Here we report the properties of a series of anilinoquinazolines (AQZs) as potent small molecule inhibitors of caspase-3. The AQZs inhibit human caspase-3 with Ki values in the 90 to 800 nM range. A subset of AQZs are equipotent against caspase-6, although most lack activity against this isoform and caspase-1, -2, -7, and -8. The AQZs inhibit endogenous caspase-3 activity toward a cell permeable, exogenously added substrate in staurosporine-treated SH-SY5Y cells. The AQZs reduce biochemical and cellular features of apoptosis that are thought to be a consequence of caspase-3 activation including DNA fragmentation, TUNEL staining, and the various morphological features that define the terminal stages of apoptotic cell death. Moreover, the AQZs also inhibit apoptosis induced by nerve growth factor withdrawal from differentiated PC12 cells. Thus, the AQZs represent a new and structurally novel class of inhibitors, some of which selectively inhibit caspase-3 and will thereby allow evaluation of the role of caspase-3 activity in various cellular models of apoptosis.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Quinazolines/pharmacology , Aniline Compounds/chemical synthesis , Animals , Apoptosis/physiology , Caspase 3 , Cell Line , Coloring Agents , DNA Fragmentation/drug effects , Enzyme Inhibitors/chemical synthesis , Fluorescent Dyes , Humans , In Situ Nick-End Labeling , Kinetics , Nerve Growth Factor/pharmacology , PC12 Cells , Phenotype , Quinazolines/chemical synthesis , Rats , Recombinant Proteins/metabolism , Staurosporine/pharmacology , Structure-Activity Relationship , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...