Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 38(6): e23505, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38507255

ABSTRACT

Aortic stenosis (AS) and hypertrophic cardiomyopathy (HCM) are distinct disorders leading to left ventricular hypertrophy (LVH), but whether cardiac metabolism substantially differs between these in humans remains to be elucidated. We undertook an invasive (aortic root, coronary sinus) metabolic profiling in patients with severe AS and HCM in comparison with non-LVH controls to investigate cardiac fuel selection and metabolic remodeling. These patients were assessed under different physiological states (at rest, during stress induced by pacing). The identified changes in the metabolome were further validated by metabolomic and orthogonal transcriptomic analysis, in separately recruited patient cohorts. We identified a highly discriminant metabolomic signature in severe AS in all samples, regardless of sampling site, characterized by striking accumulation of long-chain acylcarnitines, intermediates of fatty acid transport across the inner mitochondrial membrane, and validated this in a separate cohort. Mechanistically, we identify a downregulation in the PPAR-α transcriptional network, including expression of genes regulating fatty acid oxidation (FAO). In silico modeling of ß-oxidation demonstrated that flux could be inhibited by both the accumulation of fatty acids as a substrate for mitochondria and the accumulation of medium-chain carnitines which induce competitive inhibition of the acyl-CoA dehydrogenases. We present a comprehensive analysis of changes in the metabolic pathways (transcriptome to metabolome) in severe AS, and its comparison to HCM. Our results demonstrate a progressive impairment of ß-oxidation from HCM to AS, particularly for FAO of long-chain fatty acids, and that the PPAR-α signaling network may be a specific metabolic therapeutic target in AS.


Subject(s)
Aortic Valve Stenosis , Cardiomyopathy, Hypertrophic , Humans , Peroxisome Proliferator-Activated Receptors , Cardiomyopathy, Hypertrophic/genetics , Hypertrophy, Left Ventricular/genetics , Aortic Valve Stenosis/genetics , Fatty Acids/metabolism
2.
Mol Biol Evol ; 39(3)2022 03 02.
Article in English | MEDLINE | ID: mdl-35192718

ABSTRACT

The indigenous population of the United Arab Emirates (UAE) has a unique demographic and cultural history. Its tradition of endogamy and consanguinity is expected to produce genetic homogeneity and partitioning of gene pools while population movements and intercontinental trade are likely to have contributed to genetic diversity. Emiratis and neighboring populations of the Middle East have been underrepresented in the population genetics literature with few studies covering the broader genetic history of the Arabian Peninsula. Here, we genotyped 1,198 individuals from the seven Emirates using 1.7 million markers and by employing haplotype-based algorithms and admixture analyses, we reveal the fine-scale genetic structure of the Emirati population. Shared ancestry and gene flow with neighboring populations display their unique geographic position while increased intra- versus inter-Emirati kinship and sharing of uniparental haplogroups, reflect the endogamous and consanguineous cultural traditions of the Emirates and their tribes.


Subject(s)
Genetic Structures , Genetics, Population , Consanguinity , Geography , Humans , United Arab Emirates
3.
Brain ; 144(9): 2879-2891, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34687210

ABSTRACT

Epilepsies of early childhood are frequently resistant to therapy and often associated with cognitive and behavioural comorbidity. Aetiology focused precision medicine, notably gene-based therapies, may prevent seizures and comorbidities. Epidemiological data utilizing modern diagnostic techniques including whole genome sequencing and neuroimaging can inform diagnostic strategies and therapeutic trials. We present a 3-year, multicentre prospective cohort study, involving all children under 3 years of age in Scotland presenting with epilepsies. We used two independent sources for case identification: clinical reporting and EEG record review. Capture-recapture methodology was then used to improve the accuracy of incidence estimates. Socio-demographic and clinical details were obtained at presentation, and 24 months later. Children were extensively investigated for aetiology. Whole genome sequencing was offered for all patients with drug-resistant epilepsy for whom no aetiology could yet be identified. Multivariate logistic regression modelling was used to determine associations between clinical features, aetiology, and outcome. Three hundred and ninety children were recruited over 3 years. The adjusted incidence of epilepsies presenting in the first 3 years of life was 239 per 100 000 live births [95% confidence interval (CI) 216-263]. There was a socio-economic gradient to incidence, with a significantly higher incidence in the most deprived quintile (301 per 100 000 live births, 95% CI 251-357) compared with the least deprived quintile (182 per 100 000 live births, 95% CI 139-233), χ2 odds ratio = 1.7 (95% CI 1.3-2.2). The relationship between deprivation and incidence was only observed in the group without identified aetiology, suggesting that populations living in higher deprivation areas have greater multifactorial risk for epilepsy. Aetiology was determined in 54% of children, and epilepsy syndrome was classified in 54%. Thirty-one per cent had an identified genetic cause for their epilepsy. We present novel data on the aetiological spectrum of the most commonly presenting epilepsies of early childhood. Twenty-four months after presentation, 36% of children had drug-resistant epilepsy (DRE), and 49% had global developmental delay (GDD). Identification of an aetiology was the strongest determinant of both DRE and GDD. Aetiology was determined in 82% of those with DRE, and 75% of those with GDD. In young children with epilepsy, genetic testing should be prioritized as it has the highest yield of any investigation and is most likely to inform precision therapy and prognosis. Epilepsies in early childhood are 30% more common than previously reported. Epilepsies of undetermined aetiology present more frequently in deprived communities. This likely reflects increased multifactorial risk within these populations.


Subject(s)
Epilepsy/classification , Epilepsy/epidemiology , Socioeconomic Factors , Causality , Child, Preschool , Cohort Studies , Drug Resistant Epilepsy/classification , Drug Resistant Epilepsy/diagnosis , Drug Resistant Epilepsy/epidemiology , Drug Resistant Epilepsy/genetics , Epilepsy/diagnosis , Epilepsy/genetics , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Prospective Studies , Retrospective Studies , Scotland/epidemiology
4.
Genes Immun ; 21(1): 63-70, 2020 01.
Article in English | MEDLINE | ID: mdl-31462703

ABSTRACT

Invasive group A streptococcal (GAS) disease is uncommon but carries a high case-fatality rate relative to other infectious diseases. Given the ubiquity of mild GAS infections, it remains unclear why healthy individuals will occasionally develop life-threatening infections, raising the possibility of host genetic predisposition. Here, we present the results of a case-control study including 43 invasive GAS cases and 1540 controls. Using HLA imputation and linear mixed models, we find each copy of the HLA-DQA1*01:03 allele associates with a twofold increased risk of disease (odds ratio 2.3, 95% confidence interval 1.3-4.4, P = 0.009), an association which persists with classical HLA typing of a subset of cases and analysis with an alternative large control dataset with validated HLA data. Moreover, we propose the association is driven by the allele itself rather than the background haplotype. Overall this finding provides impetus for further investigation of the immunogenetic basis of this devastating bacterial disease.


Subject(s)
HLA Antigens/genetics , HLA-DQ alpha-Chains/genetics , Streptococcal Infections/immunology , Adult , Case-Control Studies , Female , Gene Frequency , Genes, MHC Class II , Genetic Predisposition to Disease/genetics , HLA Antigens/immunology , HLA-DQ alpha-Chains/metabolism , Haplotypes , Histocompatibility Antigens Class I/genetics , Humans , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide , Risk Factors , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity
5.
FASEB J ; 32(5): 2587-2600, 2018 05.
Article in English | MEDLINE | ID: mdl-29269398

ABSTRACT

AMPK is a critical energy sensor and target for widely used antidiabetic drugs. In ß cells, elevated glucose concentrations lower AMPK activity, and the ablation of both catalytic subunits [ß-cell-specific AMPK double-knockout (ßAMPKdKO) mice] impairs insulin secretion in vivo and ß-cell identity. MicroRNAs (miRNAs) are small RNAs that silence gene expression that are essential for pancreatic ß-cell function and identity and altered in diabetes. Here, we have explored the miRNAs acting downstream of AMPK in mouse and human ß cells. We identified 14 down-regulated and 9 up-regulated miRNAs in ßAMPKdKO vs. control islets. Gene ontology analysis of targeted transcripts revealed enrichment in pathways important for ß-cell function and identity. The most down-regulated miRNA was miR-184 (miR-184-3p), an important regulator of ß-cell function and compensatory expansion that is controlled by glucose and reduced in diabetes. We demonstrate that AMPK is a potent regulator and an important mediator of the negative effects of glucose on miR-184 expression. Additionally, we reveal sexual dimorphism in miR-184 expression in mouse and human islets. Collectively, these data demonstrate that glucose-mediated changes in AMPK activity are central for the regulation of miR-184 and other miRNAs in islets and provide a link between energy status and gene expression in ß cells.-Martinez-Sanchez, A., Nguyen-Tu, M.-S., Cebola, I., Yavari, A., Marchetti, P., Piemonti, L., de Koning, E., Shapiro, A. M. J., Johnson, P., Sakamoto, K., Smith, D. M., Leclerc, I., Ashrafian, H., Ferrer, J., Rutter, G. A. MiR-184 expression is regulated by AMPK in pancreatic islets.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/metabolism , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , MicroRNAs/biosynthesis , AMP-Activated Protein Kinases/genetics , Animals , Cell Line , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Energy Metabolism/genetics , Female , Glucose/genetics , Glucose/metabolism , Humans , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Sex Characteristics
6.
Proc Natl Acad Sci U S A ; 113(26): E3706-15, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27303042

ABSTRACT

Folliculin (FLCN) is a tumor-suppressor protein mutated in the Birt-Hogg-Dubé (BHD) syndrome, which associates with two paralogous proteins, folliculin-interacting protein (FNIP)1 and FNIP2, forming a complex that interacts with the AMP-activated protein kinase (AMPK). Although it is clear that this complex influences AMPK and other metabolic regulators, reports of its effects have been inconsistent. To address this issue, we created a recessive loss-of-function variant of Fnip1 Homozygous FNIP1 deficiency resulted in profound B-cell deficiency, partially restored by overexpression of the antiapoptotic protein BCL2, whereas heterozygous deficiency caused a loss of marginal zone B cells. FNIP1-deficient mice developed cardiomyopathy characterized by left ventricular hypertrophy and glycogen accumulation, with close parallels to mice and humans bearing gain-of-function mutations in the γ2 subunit of AMPK. Concordantly, γ2-specific AMPK activity was elevated in neonatal FNIP1-deficient myocardium, whereas AMPK-dependent unc-51-like autophagy activating kinase 1 (ULK1) phosphorylation and autophagy were increased in FNIP1-deficient B-cell progenitors. These data support a role for FNIP1 as a negative regulator of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , B-Lymphocytes/cytology , Cardiomyopathies/metabolism , Carrier Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , AMP-Activated Protein Kinases/genetics , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , Cardiomyopathies/genetics , Carrier Proteins/metabolism , Cell Count , Humans , Mice , Mice, Inbred C57BL , Mutation , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
7.
J Mol Cell Cardiol ; 121: 287-296, 2018 08.
Article in English | MEDLINE | ID: mdl-30048712

ABSTRACT

Cysteine and glycine rich protein 3 (CSRP3) encodes Muscle LIM Protein (MLP), a well-established disease gene for Hypertrophic Cardiomyopathy (HCM). MLP, in contrast to the proteins encoded by the other recognised HCM disease genes, is non-sarcomeric, and has important signalling functions in cardiomyocytes. To gain insight into the disease mechanisms involved, we generated a knock-in mouse (KI) model, carrying the well documented HCM-causing CSRP3 mutation C58G. In vivo phenotyping of homozygous KI/KI mice revealed a robust cardiomyopathy phenotype with diastolic and systolic left ventricular dysfunction, which was supported by increased heart weight measurements. Transcriptome analysis by RNA-seq identified activation of pro-fibrotic signalling, induction of the fetal gene programme and activation of markers of hypertrophic signalling in these hearts. Further ex vivo analyses validated the activation of these pathways at transcript and protein level. Intriguingly, the abundance of MLP decreased in KI/KI mice by 80% and in KI/+ mice by 50%. Protein depletion was also observed in cellular studies for two further HCM-causing CSRP3 mutations (L44P and S54R/E55G). We show that MLP depletion is caused by proteasome action. Moreover, MLP C58G interacts with Bag3 and results in a proteotoxic response in the homozygous knock-in mice, as shown by induction of Bag3 and associated heat shock proteins. In conclusion, the newly generated mouse model provides insights into the underlying disease mechanisms of cardiomyopathy caused by mutations in the non-sarcomeric protein MLP. Furthermore, our cellular experiments suggest that protein depletion and proteasomal overload also play a role in other HCM-causing CSPR3 mutations that we investigated, indicating that reduced levels of functional MLP may be a common mechanism for HCM-causing CSPR3 mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Cardiomyopathy, Hypertrophic/genetics , Heart/physiopathology , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Animals , Cardiomyopathy, Hypertrophic/physiopathology , Disease Models, Animal , Gene Knock-In Techniques , Humans , Mice , Mutation , Sarcomeres/genetics
8.
J Cardiovasc Magn Reson ; 20(1): 88, 2018 12 24.
Article in English | MEDLINE | ID: mdl-30580760

ABSTRACT

BACKGROUND: Heart failure (HF) is characterized by altered myocardial substrate metabolism which can lead to myocardial triglyceride accumulation (steatosis) and lipotoxicity. However its role in mild HF with preserved ejection fraction (HFpEF) is uncertain. We measured myocardial triglyceride content (MTG) in HFpEF and assessed its relationships with diastolic function and exercise capacity. METHODS: Twenty seven HFpEF (clinical features of HF, left ventricular EF >50%, evidence of mild diastolic dysfunction and evidence of exercise limitation as assessed by cardiopulmonary exercise test) and 14 controls underwent 1H-cardiovascular magnetic resonance spectroscopy (1H-CMRS) to measure MTG (lipid/water, %), 31P-CMRS to measure myocardial energetics (phosphocreatine-to-adenosine triphosphate - PCr/ATP) and feature-tracking cardiovascular magnetic resonance (CMR) imaging for diastolic strain rate. RESULTS: When compared to controls, HFpEF had 2.3 fold higher in MTG (1.45 ± 0.25% vs. 0.64 ± 0.16%, p = 0.009) and reduced PCr/ATP (1.60 ± 0.09 vs. 2.00 ± 0.10, p = 0.005). HFpEF had significantly reduced diastolic strain rate and maximal oxygen consumption (VO2 max), which both correlated significantly with elevated MTG and reduced PCr/ATP. On multivariate analyses, MTG was independently associated with diastolic strain rate while diastolic strain rate was independently associated with VO2 max. CONCLUSIONS: Myocardial steatosis is pronounced in mild HFpEF, and is independently associated with impaired diastolic strain rate which is itself related to exercise capacity. Steatosis may adversely affect exercise capacity by indirect effect occurring via impairment in diastolic function. As such, myocardial triglyceride may become a potential therapeutic target to treat the increasing number of patients with HFpEF.


Subject(s)
Energy Metabolism , Exercise Tolerance , Heart Failure/diagnostic imaging , Magnetic Resonance Imaging, Cine , Myocardial Contraction , Myocardium/metabolism , Triglycerides/metabolism , Ventricular Function, Left , Adenosine Triphosphate/metabolism , Aged , Biomarkers/metabolism , Biomechanical Phenomena , Case-Control Studies , Exercise Test , Female , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Middle Aged , Myocardium/pathology , Oxygen Consumption , Phosphocreatine/metabolism , Predictive Value of Tests , Prospective Studies , Proton Magnetic Resonance Spectroscopy , Severity of Illness Index
9.
J Biol Chem ; 290(43): 25907-19, 2015 10 23.
Article in English | MEDLINE | ID: mdl-26370078

ABSTRACT

We have reported previously that a missense mutation in the mitochondrial fission gene Dynamin-related protein 1 (Drp1) underlies the Python mouse model of monogenic dilated cardiomyopathy. The aim of this study was to investigate the consequences of the C452F mutation on Drp1 protein function and to define the cellular sequelae leading to heart failure in the Python monogenic dilated cardiomyopathy model. We found that the C452F mutation increased Drp1 GTPase activity. The mutation also conferred resistance to oligomer disassembly by guanine nucleotides and high ionic strength solutions. In a mouse embryonic fibroblast model, Drp1 C452F cells exhibited abnormal mitochondrial morphology and defective mitophagy. Mitochondria in C452F mouse embryonic fibroblasts were depolarized and had reduced calcium uptake with impaired ATP production by oxidative phosphorylation. In the Python heart, we found a corresponding progressive decline in oxidative phosphorylation with age and activation of sterile inflammation. As a corollary, enhancing autophagy by exposure to a prolonged low-protein diet improved cardiac function in Python mice. In conclusion, failure of Drp1 disassembly impairs mitophagy, leading to a downstream cascade of mitochondrial depolarization, aberrant calcium handling, impaired ATP synthesis, and activation of sterile myocardial inflammation, resulting in heart failure.


Subject(s)
Biopolymers/physiology , Dynamins/physiology , Heart Failure/etiology , Mitophagy , Myocarditis/etiology , Animals , Biopolymers/genetics , Biopolymers/metabolism , Cells, Cultured , Dynamins/genetics , Dynamins/metabolism , Heart Failure/physiopathology , Mice , Mutation , Myocarditis/physiopathology , Oxidative Phosphorylation
10.
Circulation ; 132(18): 1719-25, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26338956

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is associated with significant morbidity and mortality but is currently refractory to therapy. Despite limited evidence, heart rate reduction has been advocated, on the basis of physiological considerations, as a therapeutic strategy in HFpEF. We tested the hypothesis that heart rate reduction improves exercise capacity in HFpEF. METHODS AND RESULTS: We conducted a randomized, crossover study comparing selective heart rate reduction with the If blocker ivabradine at 7.5 mg twice daily versus placebo for 2 weeks each in 22 symptomatic patients with HFpEF who had objective evidence of exercise limitation (peak oxygen consumption at maximal exercise [o2 peak] <80% predicted for age and sex). The result was compared with 22 similarly treated matched asymptomatic hypertensive volunteers. The primary end point was the change in o2 peak. Secondary outcomes included tissue Doppler-derived E/e' at echocardiography, plasma brain natriuretic peptide, and quality-of-life scores. Ivabradine significantly reduced peak heart rate compared with placebo in the HFpEF (107 versus 129 bpm; P<0.0001) and hypertensive (127 versus 145 bpm; P=0.003) cohorts. Ivabradine compared with placebo significantly worsened the change in o2 peak in the HFpEF cohort (-2.1 versus 0.9 mL·kg(-1)·min(-1); P=0.003) and significantly reduced submaximal exercise capacity, as determined by the oxygen uptake efficiency slope. No significant effects on the secondary end points were discernable. CONCLUSION: Our observations bring into question the value of heart rate reduction with ivabradine for improving symptoms in a HFpEF population characterized by exercise limitation. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02354573.


Subject(s)
Benzazepines/therapeutic use , Heart Failure/drug therapy , Heart Rate/drug effects , Stroke Volume , Aged , Aged, 80 and over , Asymptomatic Diseases , Biomarkers , Cross-Over Studies , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Double-Blind Method , Endpoint Determination , Exercise Test , Exercise Tolerance , Female , Heart Failure/blood , Heart Failure/physiopathology , Humans , Hypertension/blood , Hypertension/drug therapy , Hypertension/physiopathology , Ivabradine , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Oxygen Consumption/drug effects , Sinoatrial Node/drug effects , Sinoatrial Node/physiopathology , Treatment Failure
11.
Am J Physiol Endocrinol Metab ; 311(4): E706-E719, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27577855

ABSTRACT

AMP-activated protein kinase (AMPK) plays diverse roles and coordinates complex metabolic pathways for maintenance of energy homeostasis. This could be explained by the fact that AMPK exists as multiple heterotrimer complexes comprising a catalytic α-subunit (α1 and α2) and regulatory ß (ß1 and ß2)- and γ (γ1, γ2, γ3)-subunits, which are uniquely distributed across different cell types. There has been keen interest in developing specific and isoform-selective AMPK-activating drugs for therapeutic use and also as research tools. Moreover, establishing ways of enhancing cellular AMPK activity would be beneficial for both purposes. Here, we investigated if a recently described potent AMPK activator called 991, in combination with the commonly used activator 5-aminoimidazole-4-carboxamide riboside or contraction, further enhances AMPK activity and glucose transport in mouse skeletal muscle ex vivo. Given that the γ3-subunit is exclusively expressed in skeletal muscle and has been implicated in contraction-induced glucose transport, we measured the activity of AMPKγ3 as well as ubiquitously expressed γ1-containing complexes. We initially validated the specificity of the antibodies for the assessment of isoform-specific AMPK activity using AMPK-deficient mouse models. We observed that a low dose of 991 (5 µM) stimulated a modest or negligible activity of both γ1- and γ3-containing AMPK complexes. Strikingly, dual treatment with 991 and 5-aminoimidazole-4-carboxamide riboside or 991 and contraction profoundly enhanced AMPKγ1/γ3 complex activation and glucose transport compared with any of the single treatments. The study demonstrates the utility of a dual activator approach to achieve a greater activation of AMPK and downstream physiological responses in various cell types, including skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Benzimidazoles/pharmacology , Benzoates/pharmacology , Enzyme Activators/pharmacology , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Muscle, Skeletal/drug effects , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases/drug effects , Aminoimidazole Carboxamide/pharmacology , Animals , Antibodies, Blocking/pharmacology , Humans , In Vitro Techniques , Isoenzymes , Mice , Mice, Knockout , Muscle Contraction/drug effects
12.
Circ Res ; 113(6): 660-75, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23989711

ABSTRACT

Despite the striking advances in medical and surgical therapy, the morbidity, mortality, and economic burden of heart failure (HF) remain unacceptably high. There is increasing evidence that the risk and course of HF depend on genetic predisposition; however, the genetic contribution to HF is heterogeneous and complex. At one end of the spectrum are the familial monogenic HF syndromes in which causative mutations are rare but highly penetrant. At the other, HF susceptibility and course may be influenced by more common, less penetrant genetic variants. As detailed in this review, efforts to unravel the basis of the familial cardiomyopathies at the mendelian end of the spectrum already have begun to deliver on the promise of informative mechanisms, novel gene-based diagnostics, and therapies for distinct subtypes of HF. However, continued progress requires the differentiation of pathogenic mutations, disease modifiers, and rare, benign variants in the deluge of data emerging from increasingly accessible novel sequencing technologies. This represents a significant challenge and demands a sustained effort in analysis of extended family pedigrees, diligent clinical phenotyping, and systematic annotation of human genetic variation.


Subject(s)
Cardiomyopathies/genetics , Heart Failure/etiology , Cardiomyopathies/complications , Cardiomyopathies/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Mutation , Myocardial Contraction/genetics
13.
Circ Res ; 110(9): 1192-201, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22456184

ABSTRACT

RATIONALE: AMP-activated protein kinase (AMPK) is an important regulator of energy balance and signaling in the heart. Mutations affecting the regulatory γ2 subunit have been shown to cause an essentially cardiac-restricted phenotype of hypertrophy and conduction disease, suggesting a specific role for this subunit in the heart. OBJECTIVE: The γ isoforms are highly conserved at their C-termini but have unique N-terminal sequences, and we hypothesized that the N-terminus of γ2 may be involved in conferring substrate specificity or in determining intracellular localization. METHODS AND RESULTS: A yeast 2-hybrid screen of a human heart cDNA library using the N-terminal 273 residues of γ2 as bait identified cardiac troponin I (cTnI) as a putative interactor. In vitro studies showed that cTnI is a good AMPK substrate and that Ser150 is the principal residue phosphorylated. Furthermore, on AMPK activation during ischemia, Ser150 is phosphorylated in whole hearts. Using phosphomimics, measurements of actomyosin ATPase in vitro and force generation in demembraneated trabeculae showed that modification at Ser150 resulted in increased Ca(2+) sensitivity of contractile regulation. Treatment of cardiomyocytes with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) resulted in increased myocyte contractility without changing the amplitude of Ca(2+) transient and prolonged relaxation despite shortening the time constant of Ca(2+) transient decay (tau). Compound C prevented the effect of AICAR on myocyte function. These results suggest that AMPK activation increases myocyte contraction and prolongs relaxation by increasing myofilament Ca(2+) sensitivity. CONCLUSIONS: We conclude that cTnI phosphorylation by AMPK may represent a novel mechanism of regulation of cardiac function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Myocardial Contraction , Myocytes, Cardiac/enzymology , Troponin I/metabolism , Ventricular Function, Left , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Calcium Signaling , Enzyme Activation , Enzyme Activators/pharmacology , Heart Ventricles/enzymology , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myosins/drug effects , Myosins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ribonucleotides/pharmacology , Serine , Time Factors , Troponin I/genetics , Two-Hybrid System Techniques , Ventricular Function, Left/drug effects
14.
J Cardiovasc Magn Reson ; 16: 92, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25410203

ABSTRACT

BACKGROUND: Myocardial T1 relaxation times have been reported to be markedly abnormal in diverse myocardial pathologies, ascribed to interstitial changes, evaluated by T1 mapping and calculation of extracellular volume (ECV). T1 mapping is sensitive to myocardial water content of both intra- and extracellular in origin, but the effect of intravascular compartment changes on T1 has been largely neglected. We aimed to assess the role of intravascular compartment on native (pre-contrast) T1 values by studying the effect of adenosine-induced vasodilatation in patients with severe aortic stenosis (AS) before and after aortic valve replacement (AVR). METHODS: 42 subjects (26 patients with severe AS without obstructive coronary artery disease and 16 controls) underwent cardiovascular magnetic resonance at 3 T for native T1-mapping (ShMOLLI), first-pass perfusion (myocardial perfusion reserve index-MPRI) at rest and during adenosine stress, and late gadolinium enhancement (LGE). RESULTS: AS patients had increased resting myocardial T1 (1196±47 ms vs. 1168±27 ms, p=0.037), reduced MPRI (0.92±0.31 vs. 1.74±0.32, p<0.001), and increased left ventricular mass index (LVMI) and LGE volume compared to controls. During adenosine stress, T1 in AS was similar to controls (1240±51 ms vs. 1238±54 ms, p=0.88), possibly reflecting a similar level of maximal coronary vasodilatation in both groups. Conversely, the T1 response to stress was blunted in AS (ΔT1 3.7±2.7% vs. 6.0±4.2% in controls, p=0.013). Seven months after AVR (n=16) myocardial T1 and response to adenosine stress recovered towards normal. Native T1 values correlated with reduced MPRI, aortic valve area, and increased LVMI. CONCLUSIONS: Our study suggests that native myocardial T1 values are not only influenced by interstitial and intracellular water changes, but also by changes in the intravascular compartment. Performing T1 mapping during or soon after vasodilator stress may affect ECV measurements given that hyperemia alone appears to substantially alter T1 values.


Subject(s)
Adenosine , Aortic Valve Stenosis/diagnosis , Coronary Circulation , Magnetic Resonance Imaging, Cine/methods , Myocardial Contraction , Myocardial Perfusion Imaging/methods , Vasodilation , Vasodilator Agents , Ventricular Function, Left , Aged , Aortic Valve/physiopathology , Aortic Valve/surgery , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/physiopathology , Aortic Valve Stenosis/surgery , Case-Control Studies , Contrast Media , Female , Fibrosis , Gadolinium DTPA , Heart Valve Prosthesis Implantation , Humans , Male , Middle Aged , Myocardium/pathology , Predictive Value of Tests , Prospective Studies , Severity of Illness Index , Treatment Outcome
15.
J Cardiovasc Magn Reson ; 16: 29, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24779370

ABSTRACT

BACKGROUND: Left ventricular (LV) hypertrophy in aortic stenosis (AS) is characterized by reduced myocardial perfusion reserve due to coronary microvascular dysfunction. However, whether this hypoperfusion leads to tissue deoxygenation is unknown. We aimed to assess myocardial oxygenation in severe AS without obstructive coronary artery disease, and to investigate its association with myocardial energetics and function. METHODS: Twenty-eight patients with isolated severe AS and 15 controls underwent cardiovascular magnetic resonance (CMR) for assessment of perfusion (myocardial perfusion reserve index-MPRI) and oxygenation (blood-oxygen level dependent-BOLD signal intensity-SI change) during adenosine stress. LV circumferential strain and phosphocreatine/adenosine triphosphate (PCr/ATP) ratios were assessed using tagging CMR and 31P MR spectroscopy, respectively. RESULTS: AS patients had reduced MPRI (1.1 ± 0.3 vs. controls 1.7 ± 0.3, p < 0.001) and BOLD SI change during stress (5.1 ± 8.9% vs. controls 18.2 ± 10.1%, p = 0.001), as well as reduced PCr/ATP (1.45 ± 0.21 vs. 2.00 ± 0.25, p < 0.001) and LV strain (-16.4 ± 2.7% vs. controls -21.3 ± 1.9%, p < 0.001). Both perfusion reserve and oxygenation showed positive correlations with energetics and LV strain. Furthermore, impaired energetics correlated with reduced strain. Eight months post aortic valve replacement (AVR) (n = 14), perfusion (MPRI 1.6 ± 0.5), oxygenation (BOLD SI change 15.6 ± 7.0%), energetics (PCr/ATP 1.86 ± 0.48) and circumferential strain (-19.4 ± 2.5%) improved significantly. CONCLUSIONS: Severe AS is characterized by impaired perfusion reserve and oxygenation which are related to the degree of derangement in energetics and associated LV dysfunction. These changes are reversible on relief of pressure overload and hypertrophy regression. Strategies aimed at improving oxygen demand-supply balance to preserve myocardial energetics and LV function are promising future therapies.


Subject(s)
Aortic Valve Stenosis/diagnosis , Coronary Circulation , Energy Metabolism , Hypertrophy, Left Ventricular/diagnosis , Magnetic Resonance Imaging, Cine , Myocardial Perfusion Imaging/methods , Myocardium/metabolism , Oxygen Consumption , Ventricular Dysfunction, Left/diagnosis , Ventricular Function, Left , Adenosine , Adenosine Triphosphate/metabolism , Aged , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/physiopathology , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Magnetic Resonance Spectroscopy , Male , Middle Aged , Phosphocreatine/metabolism , Predictive Value of Tests , Severity of Illness Index , Vasodilator Agents , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
16.
Basic Res Cardiol ; 108(3): 343, 2013 May.
Article in English | MEDLINE | ID: mdl-23525499

ABSTRACT

Mitochondrial aldehyde dehydrogenase-2 (ALDH-2) is involved in preconditioning pathways, but its role in remote ischaemic preconditioning (rIPC) is unknown. We investigated its role in animal and human models of rIPC. (i) In a rabbit model of myocardial infarction, rIPC alone reduced infarct size [69 ± 5.8 % (n = 11) to 40 ± 6.5 % (n = 12), P = 0.019]. However, rIPC protection was lost after pre-treatment with the ALDH-2 inhibitor cyanamide (62 ± 7.6 % controls, n = 10, versus 61 ± 6.9 % rIPC after cyanamide, n = 10, P > 0.05). (ii) In a forearm plethysmography model of endothelial ischaemia-reperfusion injury, 24 individuals of Asian ethnic origin underwent combined rIPC and ischaemia-reperfusion (IR). 11 had wild-type (WT) enzyme and 13 carried the Glu504Lys (ALDH2*2) polymorphism (rendering ALDH-2 functionally inactive). In WT individuals, rIPC protected against impairment of response to acetylcholine (P = 0.9), but rIPC failed to protect carriers of Glu504Lys polymorphism (P = 0.004). (iii) In a second model of endothelial IR injury, 12 individuals participated in a double-blind placebo-controlled crossover study, receiving the ALDH-2 inhibitor disulfiram 600 mg od or placebo for 48 h prior to assessment of flow-mediated dilation (FMD) before and after combined rIPC and IR. With placebo, rIPC was effective with no difference in FMD before and after IR (6.18 ± 1.03 % and 4.76 ± 0.93 % P = 0.1), but disulfiram inhibited rIPC with a reduction in FMD after IR (7.87 ± 1.27 % and 3.05 ± 0.53 %, P = 0.001). This study demonstrates that ALDH-2 is involved in the rIPC pathway in three distinct rabbit and human models. This has potential implications for future clinical studies of remote conditioning.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Cyanamide/pharmacology , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , Forearm/blood supply , Hindlimb/blood supply , Ischemic Preconditioning/methods , Myocardial Infarction/prevention & control , Myocardium/enzymology , Reperfusion Injury/prevention & control , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Analysis of Variance , Animals , Cross-Over Studies , Disease Models, Animal , Dose-Response Relationship, Drug , Double-Blind Method , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Genotype , Humans , Linear Models , Mutation , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Phenotype , Plethysmography , Rabbits , Regional Blood Flow/drug effects , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
17.
Circ Res ; 109(1): 86-96, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21700950

ABSTRACT

As described in earlier reviews in this series on the molecular basis of hypertrophic cardiomyopathy (HCM), HCM is one of the archetypal monogenic cardiovascular disorders to be understood at the molecular level. Twenty years after the discovery of the first HCM disease gene, genetic studies still confirm that HCM is principally a disease of the sarcomere. At the biophysical level, myofilament mutations generally enhance Ca(2+) sensitivity, maximal force production, and ATPase activity. These defects ultimately appear to converge on energy deficiency and altered Ca(2+) handling as major common paths leading to the anatomic (hypertrophy, myofiber disarray, and fibrosis) and functional features (pathological signaling and diastolic dysfunction) characteristic of HCM. In this review, we provide an account of the consequences of HCM mutations and describe how specifically targeting these molecular features has already yielded early promise for novel therapies for HCM. Although substantial efforts are still required to understand the molecular link between HCM mutations and their clinical consequences, HCM endures as an exemplar of how novel insights derived from molecular characterization of Mendelian disorders can inform the understanding of biological processes and translate into rational therapies.


Subject(s)
Cardiomyopathy, Hypertrophic/etiology , Actin Cytoskeleton/physiology , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/genetics , Energy Metabolism , Humans , Mutation , Myocardium/metabolism , Signal Transduction
18.
J Cardiovasc Magn Reson ; 15: 19, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23414451

ABSTRACT

BACKGROUND: Alterations in cardiac metabolism accompany many diseases of the heart. The advent of cardiac hyperpolarized magnetic resonance spectroscopy (MRS), via dynamic nuclear polarization (DNP), has enabled a greater understanding of the in vivo metabolic changes that occur as a consequence of myocardial infarction, hypertrophy and diabetes. However, all cardiac studies performed to date have focused on rats and larger animals, whereas more information could be gained through the study of transgenic mouse models of heart disease. Translation from the rat to the mouse is challenging, due in part to the reduced heart size (1/10(th)) and the increased heart rate (50%) in the mouse compared to the rat. METHODS AND RESULTS: In this study, we have investigated the in vivo metabolism of [1-(13)C]pyruvate in the mouse heart. To demonstrate the sensitivity of the method to detect alterations in pyruvate dehydrogenase (PDH) flux, two well characterised methods of PDH modulation were performed; overnight fasting and infusion of sodium dichloroacetate (DCA). Fasting resulted in an 85% reduction in PDH flux, whilst DCA infusion increased PDH flux by 123%. A comparison of three commonly used control mouse strains was performed revealing significant metabolic differences between strains. CONCLUSIONS: We have successfully demonstrated a hyperpolarized DNP protocol to investigate in vivo alterations within the diseased mouse heart. This technique offers a significant advantage over existing in vitro techniques as it reduces animal numbers and decreases biological variability. Thus [1-(13)C]pyruvate can be used to provide an in vivo cardiac metabolic profile of transgenic mice.


Subject(s)
Energy Metabolism , Magnetic Resonance Spectroscopy , Myocardium/metabolism , Animals , Carbon Isotopes , Dichloroacetic Acid/administration & dosage , Energy Metabolism/drug effects , Fasting/metabolism , Infusions, Intravenous , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Pyruvate Dehydrogenase Complex , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Species Specificity , Time Factors
19.
PLoS Genet ; 6(6): e1001000, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585624

ABSTRACT

Mutations in a number of genes have been linked to inherited dilated cardiomyopathy (DCM). However, such mutations account for only a small proportion of the clinical cases emphasising the need for alternative discovery approaches to uncovering novel pathogenic mutations in hitherto unidentified pathways. Accordingly, as part of a large-scale N-ethyl-N-nitrosourea mutagenesis screen, we identified a mouse mutant, Python, which develops DCM. We demonstrate that the Python phenotype is attributable to a dominant fully penetrant mutation in the dynamin-1-like (Dnm1l) gene, which has been shown to be critical for mitochondrial fission. The C452F mutation is in a highly conserved region of the M domain of Dnm1l that alters protein interactions in a yeast two-hybrid system, suggesting that the mutation might alter intramolecular interactions within the Dnm1l monomer. Heterozygous Python fibroblasts exhibit abnormal mitochondria and peroxisomes. Homozygosity for the mutation results in the death of embryos midway though gestation. Heterozygous Python hearts show reduced levels of mitochondria enzyme complexes and suffer from cardiac ATP depletion. The resulting energy deficiency may contribute to cardiomyopathy. This is the first demonstration that a defect in a gene involved in mitochondrial remodelling can result in cardiomyopathy, showing that the function of this gene is needed for the maintenance of normal cellular function in a relatively tissue-specific manner. This disease model attests to the importance of mitochondrial remodelling in the heart; similar defects might underlie human heart muscle disease.


Subject(s)
Cardiomyopathy, Dilated/genetics , GTP Phosphohydrolases/genetics , Genes, Mitochondrial , Genetic Predisposition to Disease , Microtubule-Associated Proteins/genetics , Mutation , Amino Acid Sequence , Animals , Base Sequence , Cardiomyopathy, Dilated/congenital , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Dynamins , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL