Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Alzheimers Dis ; 4(4): 291-301, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12446931

ABSTRACT

Withdrawal of NGF (NGF-W) in PC12 cells leads to caspase and GSK3beta activation which results in cell death. Our recent findings suggest that inhibition of GSK3beta promotes PC12 cell survival after NGF-W. To determine whether these pathways interact from a signalling perspective, we compared the effects of BAF (a general caspase inhibitor), Li+ (a GSK3beta inhibitor) and insulin on NGF-W induced PC12 cell death. Maximal increase in DNA fragmentation was observed 3 h after NGF-W and was inhibited by BAF (7.5 microM), Li+ (IC(50) = 2 mM) and insulin (IC(50) = 100 nM). BAF inhibited caspase-3 activity and delayed cell death up to 6 h after NGF-W indicating that caspase inhibition is sufficient to prevent apoptosis. BAF had no major effect on GSK3betaactive site phosphorylation or activity suggesting the caspase pathway does not regulate GSK3beta activity. Conversely, Li+ inhibited caspase activity by only 20% but promoted cell survival for 24 h after NGF-W. Overexpression of dominant negative mutants of GSK3beta also inhibited apoptosis, but had only a minor effect on caspase activity after NGF-W. Taken together, these results suggest that GSK3beta is upstream of caspase signalling, and exerts a small effect on the caspase pathway.


Subject(s)
Caspases/physiology , Cell Death/physiology , Glycogen Synthase Kinase 3/physiology , Nerve Growth Factor/physiology , Signal Transduction/physiology , Tumor Cells, Cultured/physiology , Animals , Apoptosis/physiology , Glycogen Synthase Kinase 3 beta , PC12 Cells , Rats
11.
Neurosignals ; 11(5): 251-61, 2002.
Article in English | MEDLINE | ID: mdl-12566926

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) is a kinase that plays a pivotal role in numerous cellular functions from modulation of microtubule dynamics and cell death. It also affects higher functions such as cognition and mood. Deregulation of GSK3beta activity in the adult brain is implicated in several CNS disorders, such as affective disorders, schizophrenia, stroke and neurodegenerative diseases, such as Alzheimer's disease (AD). In AD, GSK3beta has a major role in microtubule stability by its ability to phosphorylate the microtubule associated protein tau. The present review focuses on recent developments in the understanding of GSK3beta with an emphasis on events likely to be critical to the pathophysiology of AD.


Subject(s)
Alzheimer Disease/physiopathology , Glycogen Synthase Kinase 3/metabolism , Alzheimer Disease/pathology , Animals , Central Nervous System Diseases/physiopathology , Gene Expression Regulation, Enzymologic , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Microtubules/pathology , Models, Molecular , Protein Structure, Secondary , Psychotic Disorders/physiopathology , Signal Transduction , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...