Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Metabolomics ; 17(9): 76, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34424431

ABSTRACT

INTRODUCTION: Acute myocardial ischaemia and the transition from reversible to irreversible myocardial injury are associated with abnormal metabolic patterns. Advances in metabolomics have extended our capabilities to define these metabolic perturbations on a metabolome-wide scale. OBJECTIVES: This study was designed to identify cardiac metabolic changes in serum during the first 5 min following early myocardial ischaemia in humans, applying an untargeted metabolomics approach. METHODS: Peripheral venous samples were collected from 46 patients in a discovery study (DS) and a validation study (VS) (25 for DS, 21 for VS). Coronary sinus venous samples were collected from 7 patients (4 for DS, 3 for VS). Acute myocardial ischaemia was induced by transient coronary occlusion during percutaneous coronary intervention (PCI). Plasma samples were collected at baseline (prior to PCI) and at 1 and 5 min post-coronary occlusion. Samples were analyzed by Ultra Performance Liquid Chromatography-Mass Spectrometry in an untargeted metabolomics approach. RESULTS: The study observed changes in the circulating levels of metabolites at 1 and 5 min following transient coronary ischaemia. Both DS and VS identified 54 and 55 metabolites as significant (P < 0.05) when compared to baseline levels, respectively. Fatty acid beta-oxidation and anaerobic respiration, lysoglycerophospholipids, arachidonic acid, docosahexaenoic acid, tryptophan metabolism and sphingosine-1-phosphate were identified as mechanistically important. CONCLUSION: Using an untargeted metabolomics approach, the study identified important cardiac metabolic changes in peripheral and coronary sinus plasma, in a human model of controlled acute myocardial ischaemia. Distinct classes of metabolites were shown to be involved in the rapid cardiac response to ischemia and provide insights into diagnostic and interventional targets.


Subject(s)
Coronary Artery Disease , Coronary Occlusion , Percutaneous Coronary Intervention , Humans , Metabolome , Metabolomics
2.
Sci Rep ; 11(1): 1518, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452399

ABSTRACT

Ischaemic heart disease is the world's leading cause of mortality. Survival rates from acute myocardial infarction (MI) have improved in recent years; however, this has led to an increase in the prevalence of heart failure (HF) due to chronic remodelling of the infarcted myocardium, for which treatment options remain poor. We have previously shown that inhibition of isoform 4 of the plasma membrane calcium ATPase (PMCA4) prevents chronic remodelling and HF development during pressure overload, through fibroblast mediated Wnt signalling modulation. Given that Wnt signalling also plays a prominent role during remodelling of the infarcted heart, this study investigated the effect of genetic and functional loss of PMCA4 on cardiac outcomes following MI. Neither genetic deletion nor pharmacological inhibition of PMCA4 affected chronic remodelling of the post-MI myocardium. This was the case when PMCA4 was deleted globally, or specifically from cardiomyocytes or fibroblasts. PMCA4-ablated hearts were however less prone to acute arrhythmic events, which may offer a slight survival benefit. Overall, this study demonstrates that PMCA4 inhibition does not affect chronic outcomes following MI.


Subject(s)
Arrhythmias, Cardiac/genetics , Calcium-Transporting ATPases/metabolism , Myocardial Infarction/genetics , Animals , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Calcium-Transporting ATPases/genetics , Disease Models, Animal , Female , Fibroblasts/metabolism , Heart Failure/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Vascular Remodeling/genetics , Vascular Remodeling/physiology , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
3.
Curr Res Physiol ; 1: 1-10, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32699840

ABSTRACT

BACKGROUND: The mouse model of transverse aortic constriction (TAC) has been widely used as a cardiac stress in the investigation of the molecular mechanisms of cardiac hypertrophy. Recently, the International Knockout Mouse Consortium has selected the C57BL/6NTac (BL/6N) mouse strain to generate null alleles for all mouse genes; however, a range of genetic and cardiac phenotypic differences have been reported between this substrain and the commonly used C57BL/6J (BL/6J) substrain. It has been reported by Garcia-Menendez and colleagues that 12-week C57BL/6NTac mice are susceptible to heart failure but little is known about the cardiac remodeling in this substrain as cardiac function progresses from compensation to decompensation. METHODS: BL/6J and BL/6N mice were subjected to pressure overload via TAC. The impact of both age and duration of cardiac pressure overload induced by TAC on cardiac remodelling were systematically assessed. RESULTS: Our data showed that BL/6N mice developed eccentric hypertrophy with age- and time-dependent deterioration in cardiac function, accompanied by considerable interstitial fibrosis. In contrast, BL/6J mice were more resilient to TAC-induced cardiac stress and developed variable cardiac phenotypes independent of age and the duration of pressure overload. This was likely due to the greater variability in pre-TAC aortic arch dimension as measured by echocardiography. In addition to increased expression of brain natriuretic peptide and collagen gene type 1 and 3, BL/6N mice also had greater angiotensin II type 2 receptor (AT2R) gene expression than BL/6J counterparts at baseline and after 2-weeks TAC, which may contribute to the exacerbated interstitial fibrosis. CONCLUSIONS: BL/6N and BL/6J mice have very different responses to TAC stimulation and these differences should be taken into consideration when using the substrains to investigate the mechanisms of hypertrophy and heart failure.

4.
J Cell Mol Med ; 22(2): 861-872, 2018 02.
Article in English | MEDLINE | ID: mdl-29193716

ABSTRACT

Cardiovascular disease is the world's leading cause of morbidity and mortality, with high blood pressure (BP) contributing to increased severity and number of adverse outcomes. Plasma membrane calcium ATPase 4 (PMCA4) has been previously shown to modulate systemic BP. However, published data are conflicting, with both overexpression and inhibition of PMCA4 in vivo shown to increase arterial contractility. Hence, our objective was to determine the role of PMCA4 in the regulation of BP and to further understand how PMCA4 functionally regulates BP using a novel specific inhibitor to PMCA4, aurintricarboxylic acid (ATA). Our approach assessed conscious BP and contractility of resistance arteries from PMCA4 global knockout (PMCA4KO) mice compared to wild-type animals. Global ablation of PMCA4 had no significant effect on BP, arterial structure or isolated arterial contractility. ATA treatment significantly reduced BP and arterial contractility in wild-type mice but had no significant effect in PMCA4KO mice. The effect of ATAin vivo and ex vivo was abolished by the neuronal nitric oxide synthase (nNOS) inhibitor Vinyl-l-NIO. Thus, this highlights differences in the effects of PMCA4 ablation and acute inhibition on the vasculature. Importantly, for doses here used, we show the vascular effects of ATA to be specific for PMCA4 and that ATA may be a further experimental tool for elucidating the role of PMCA4.


Subject(s)
Blood Pressure , Mesenteric Arteries/physiopathology , Nitric Oxide Synthase Type I/metabolism , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Animals , Aurintricarboxylic Acid/pharmacology , Blood Pressure/drug effects , Calcium/metabolism , Consciousness , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mice, Knockout , Models, Biological , Peptides/pharmacology , Plasma Membrane Calcium-Transporting ATPases/metabolism
5.
Aging Cell ; 16(5): 1104-1113, 2017 10.
Article in English | MEDLINE | ID: mdl-28795531

ABSTRACT

Hypertension is a well-established risk factor for adverse cardiovascular events, and older age is a risk factor for the development of hypertension. Genomewide association studies have linked ATP2B1, the gene for the plasma membrane calcium ATPase 1 (PMCA1), to blood pressure (BP) and hypertension. Here, we present the effects of reduction in the expression of PMCA1 on BP and small artery structure and function when combined with advancing age. Heterozygous PMCA1 null mice (PMCA1Ht ) were generated and conscious BP was measured at 6 to 18 months of age. Passive and active properties of isolated small mesenteric arteries were examined by pressure myography. PMCA1Ht mice exhibited normal BP at 6 and 9 months of age but developed significantly elevated BP when compared to age-matched wild-type controls at ≥12 months of age. Decreased lumen diameter, increased wall thickness and increased wall:lumen ratio were observed in small mesenteric arteries from animals 9 months of age and older, indicative of eutrophic remodelling. Increases in mesenteric artery intrinsic tone and global intracellular calcium were evident in animals at both 6 and 18 months of age. Thus, decreased expression of PMCA1 is associated with increased BP when combined with advancing age. Changes in arterial structure precede the elevation of BP. Pathways involving PMCA1 may be a novel target for BP regulation in the elderly.


Subject(s)
Aging/genetics , Hypertension/genetics , Mesenteric Arteries/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Vascular Remodeling/genetics , Vascular Resistance/genetics , Aging/metabolism , Animals , Blood Pressure/physiology , Calcium/metabolism , Gene Expression , Heterozygote , Hypertension/metabolism , Hypertension/physiopathology , Male , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myography , Plasma Membrane Calcium-Transporting ATPases/deficiency
6.
J Mol Cell Cardiol ; 109: 38-47, 2017 08.
Article in English | MEDLINE | ID: mdl-28684310

ABSTRACT

AIMS: Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS: We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS: Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.


Subject(s)
Calcium-Transporting ATPases/metabolism , Cell Membrane/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Aurintricarboxylic Acid/pharmacology , Calcium-Transporting ATPases/genetics , Cell Membrane/genetics , Cell Movement/drug effects , Cell Movement/physiology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mice , Mice, Knockout , Neovascularization, Physiologic/drug effects , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Physiol Rev ; 97(3): 1089-1125, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28566538

ABSTRACT

The Ca2+ extrusion function of the four mammalian isoforms of the plasma membrane calcium ATPases (PMCAs) is well established. There is also ever-increasing detail known of their roles in global and local Ca2+ homeostasis and intracellular Ca2+ signaling in a wide variety of cell types and tissues. It is becoming clear that the spatiotemporal patterns of expression of the PMCAs and the fact that their abundances and relative expression levels vary from cell type to cell type both reflect and impact on their specific functions in these cells. Over recent years it has become increasingly apparent that these genes have potentially significant roles in human health and disease, with PMCAs1-4 being associated with cardiovascular diseases, deafness, autism, ataxia, adenoma, and malarial resistance. This review will bring together evidence of the variety of tissue-specific functions of PMCAs and will highlight the roles these genes play in regulating normal physiological functions and the considerable impact the genes have on human disease.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Membrane/enzymology , Disease/etiology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Disease/genetics , Genetic Predisposition to Disease , Genetic Variation , Homeostasis , Humans , Organ Specificity , Phenotype , Plasma Membrane Calcium-Transporting ATPases/chemistry , Plasma Membrane Calcium-Transporting ATPases/genetics , Protein Conformation , Structure-Activity Relationship
8.
Nephrol Dial Transplant ; 32(6): 1006-1013, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27257278

ABSTRACT

BACKGROUND: Cardiac abnormalities are frequent in patients with atherosclerotic renovascular disease (ARVD). The Angioplasty and Stenting for Renal Artery Lesions (ASTRAL) trial studied the effect of percutaneous renal revascularization combined with medical therapy compared with medical therapy alone in 806 patients with ARVD. METHODS: This was a pre-specified sub-study of ASTRAL (clinical trials registration, current controlled trials number: ISRCTN59586944), designed to consider the effect of percutaneous renal artery angioplasty and stenting on change in cardiac structure and function, measured using cardiac magnetic resonance (CMR) imaging. Fifty-one patients were recruited from six selected ASTRAL centres. Forty-four completed the study (medical therapy n = 21; revascularization n = 23). Full analysis of CMR was possible in 40 patients (18 medical therapy and 22 revascularization). CMR measurements of left and right ventricular end systolic (LV and RVESV) and diastolic volume (LV and RVEDV), ejection fraction (LVEF) and mass (LVM) were made shortly after recruitment and before revascularization in the interventional group, and again after 12 months. Reporting was performed by CMR analysts blinded to randomization arm. RESULTS: Groups were well matched for mean age (70 versus 72 years), blood pressure (148/71 versus 143/74 mmHg), degree of renal artery stenosis (75 versus 75%) and comorbid conditions. In both randomized groups, improvements in cardiac structural parameters were seen at 12 months, but there were no significant differences between treatment groups. Median left ventricular changes between baseline and 12 months (medical versus revascularization) were LVEDV -1.9 versus -5.8 mL, P = 0.4; LVESV -2.1 versus 0.3 mL, P = 0.7; LVM -5.4 versus -6.3 g, P = 0.8; and LVEF -1.5 versus -0.8%, P = 0.7. Multivariate regression also found that randomized treatment assignment was not associated with degree of change in any of the CMR measurements. CONCLUSIONS: In this sub-study of the ASTRAL trial, renal revascularization did not offer additional benefit to cardiac structure or function in unselected patients with ARVD.


Subject(s)
Atherosclerosis/surgery , Heart Ventricles/pathology , Renal Artery Obstruction/surgery , Aged , Aged, 80 and over , Angioplasty , Atherosclerosis/physiopathology , Blood Pressure , Female , Glomerular Filtration Rate , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myocardium/pathology , Renal Artery/surgery , Renal Artery Obstruction/physiopathology , Treatment Outcome
9.
Nat Commun ; 7: 11074, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-27020607

ABSTRACT

The heart responds to pathological overload through myocyte hypertrophy. Here we show that this response is regulated by cardiac fibroblasts via a paracrine mechanism involving plasma membrane calcium ATPase 4 (PMCA4). Pmca4 deletion in mice, both systemically and specifically in fibroblasts, reduces the hypertrophic response to pressure overload; however, knocking out Pmca4 specifically in cardiomyocytes does not produce this effect. Mechanistically, cardiac fibroblasts lacking PMCA4 produce higher levels of secreted frizzled related protein 2 (sFRP2), which inhibits the hypertrophic response in neighbouring cardiomyocytes. Furthermore, we show that treatment with the PMCA4 inhibitor aurintricarboxylic acid (ATA) inhibits and reverses cardiac hypertrophy induced by pressure overload in mice. Our results reveal that PMCA4 regulates the development of cardiac hypertrophy and provide proof of principle for a therapeutic approach to treat this condition.


Subject(s)
Calcium-Transporting ATPases/metabolism , Cardiomegaly/pathology , Cell Membrane/enzymology , Fibroblasts/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Signal Transduction , Animals , Animals, Newborn , Aorta/pathology , Aurintricarboxylic Acid/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/deficiency , Cardiomegaly/complications , Cell Membrane/drug effects , Constriction, Pathologic , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Fibroblasts/drug effects , Gene Deletion , Membrane Proteins/metabolism , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pressure , Signal Transduction/drug effects
10.
Pharmacol Ther ; 159: 23-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26820758

ABSTRACT

The incidence of hypertension, the major modifiable risk factor for cardiovascular disease, is increasing. Thus, there is a pressing need for the development of new and more effective strategies to prevent and treat hypertension. Development of these relies on a continued evolution of our understanding of the mechanisms which control blood pressure (BP). Resistance arteries are important in the regulation of total peripheral resistance and BP; changes in their structure and function are strongly associated with hypertension. Anti-hypertensives which both reduce BP and reverse changes in resistance arterial structure reduce cardiovascular risk more than therapies which reduce BP alone. Hence, identification of novel potential vascular targets which modify BP is important. Hypertension is a multifactorial disorder which may include a genetic component. Genome wide association studies have identified ATP2B1, encoding the calcium pump plasma membrane calcium ATPase 1 (PMCA1), as having a strong association with BP and hypertension. Knockdown or reduced PMCA1 expression in mice has confirmed a physiological role for PMCA1 in BP and resistance arterial regulation. Altered expression or inhibition of PMCA4 has also been shown to modulate these parameters. The mechanisms whereby PMCA1 and 4 can modulate vascular function remain to be fully elucidated but may involve regulation of intracellular calcium homeostasis and/or comprise a structural role. However, clear physiological links between PMCA and BP, coupled with experimental studies directly linking PMCA1 and 4 to changes in BP and arterial function, suggest that they may be important targets for the development of new pharmacological modulators of BP.


Subject(s)
Hypertension/drug therapy , Plasma Membrane Calcium-Transporting ATPases/physiology , Animals , Arteries/physiology , Blood Pressure/physiology , Essential Hypertension , Humans , Hypertension/metabolism , Hypertension/physiopathology , Plasma Membrane Calcium-Transporting ATPases/metabolism
11.
Biochem Biophys Res Commun ; 467(1): 152-6, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26392310

ABSTRACT

The physiological importance of the intestinal plasma membrane calcium pump, isoform 1, (Pmca1, Atp2b1), in calcium absorption and homeostasis has not been previously demonstrated in vivo. Since global germ-line deletion of the Pmca1 in mice is associated with embryonic lethality, we selectively deleted the Pmca1 in intestinal absorptive cells. Mice with loxP sites flanking exon 2 of the Pmca1 gene (Pmca1(fl/fl)) were crossed with mice expressing Cre recombinase in the intestine under control of the villin promoter to give mice in which the Pmca1 had been deleted in the intestine (Pmca1(EKO) mice). Pmca1(EKO) mice were born at a reduced frequency and were small at the time of birth when compared to wild-type (Wt) littermates. At two months of age, Pmca1(EKO) mice fed a 0.81% calcium, 0.34% phosphorus, normal vitamin D diet had reduced whole body bone mineral density (P < 0.037), and reduced femoral bone mineral density (P < 0.015). There was a trend towards lower serum calcium and higher serum parathyroid hormone (PTH) and 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) concentrations in Pmca1(EKO) mice compared to Wt mice but the changes were not statistically significant. The urinary phosphorus/creatinine ratio was increased in Pmca1(EKO) mice (P < 0.004). Following the administration of 200 ng of 1α,25(OH)2D3 intraperitoneally to Wt mice, active intestinal calcium transport increased ∼2-fold, whereas Pmca1(EKO) mice administered an equal amount of 1α,25(OH)2D3 failed to show an increase in active calcium transport. Deletion of the Pmca1 in the intestine is associated with reduced growth and bone mineralization, and a failure to up-regulate calcium absorption in response to 1α,25(OH)2D3.


Subject(s)
Bone Density/physiology , Calcitriol/pharmacology , Intestinal Mucosa/metabolism , Plasma Membrane Calcium-Transporting ATPases/deficiency , Animals , Bone Density/drug effects , Bone Density/genetics , Bone Density Conservation Agents/pharmacology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Calcification, Physiologic/physiology , Female , Gene Knockout Techniques/methods , Intestinal Absorption/drug effects , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Male , Mice , Mice, Knockout , Plasma Membrane Calcium-Transporting ATPases/genetics
12.
Arterioscler Thromb Vasc Biol ; 34(10): 2310-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147342

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) has been identified as a crucial regulator of physiological and pathological angiogenesis. Among the intracellular signaling pathways triggered by VEGF, activation of the calcineurin/nuclear factor of activated T cells (NFAT) signaling axis has emerged as a critical mediator of angiogenic processes. We and others previously reported a novel role for the plasma membrane calcium ATPase (PMCA) as an endogenous inhibitor of the calcineurin/NFAT pathway, via interaction with calcineurin, in cardiomyocytes and breast cancer cells. However, the functional significance of the PMCA/calcineurin interaction in endothelial pathophysiology has not been addressed thus far. APPROACH AND RESULTS: Using in vitro and in vivo assays, we here demonstrate that the interaction between PMCA4 and calcineurin in VEGF-stimulated endothelial cells leads to downregulation of the calcineurin/NFAT pathway and to a significant reduction in the subsequent expression of the NFAT-dependent, VEGF-activated, proangiogenic genes RCAN1.4 and Cox-2. PMCA4-dependent inhibition of calcineurin signaling translates into a reduction in endothelial cell motility and blood vessel formation that ultimately impairs in vivo angiogenesis by VEGF. CONCLUSIONS: Given the importance of the calcineurin/NFAT pathway in the regulation of pathological angiogenesis, targeted modulation of PMCA4 functionality might open novel therapeutic avenues to promote or attenuate new vessel formation in diseases that occur with angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Calcineurin/metabolism , Calcium-Transporting ATPases/metabolism , Endothelial Cells/drug effects , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Plasma Membrane Calcium-Transporting ATPases/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Calcium-Binding Proteins , Calcium-Transporting ATPases/deficiency , Calcium-Transporting ATPases/genetics , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , DNA-Binding Proteins , Disease Models, Animal , Endothelial Cells/enzymology , HEK293 Cells , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/enzymology , Ischemia/physiopathology , Mice , Mice, Knockout , Muscle Proteins/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection
13.
J Biol Chem ; 289(35): 24275-88, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25035424

ABSTRACT

The Hippo signaling pathway has recently moved to center stage in cardiac research because of its key role in cardiomyocyte proliferation and regeneration of the embryonic and newborn heart. However, its role in the adult heart is incompletely understood. We investigate here the role of mammalian Ste20-like kinase 2 (Mst2), one of the central regulators of this pathway. Mst2(-/-) mice showed no alteration in cardiomyocyte proliferation. However, Mst2(-/-) mice exhibited a significant reduction of hypertrophy and fibrosis in response to pressure overload. Consistently, overexpression of MST2 in neonatal rat cardiomyocytes significantly enhanced phenylephrine-induced cellular hypertrophy. Mechanistically, Mst2 positively modulated the prohypertrophic Raf1-ERK1/2 pathway. However, activation of the downstream effectors of the Hippo pathway (Yes-associated protein) was not affected by Mst2 ablation. An initial genetic study in mitral valve prolapse patients revealed an association between a polymorphism in the human MST2 gene and adverse cardiac remodeling. These results reveal a novel role of Mst2 in stress-dependent cardiac hypertrophy and remodeling in the adult mouse and likely human heart.


Subject(s)
Cardiomegaly/physiopathology , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , Animals , Apoptosis , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cell Proliferation , Humans , In Situ Nick-End Labeling , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Phenylephrine/adverse effects , Proto-Oncogene Proteins c-raf/metabolism , Serine-Threonine Kinase 3
14.
Am Heart J ; 167(6): 900-8.e1, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24890541

ABSTRACT

BACKGROUND: Cardiogenic shock (CS) remains the leading cause of mortality in patients hospitalized with acute myocardial infarction (AMI). The transradial access site (TRA) has become increasingly adopted as a default access site for percutaneous coronary intervention (PCI); however, even in experienced centers that favor the radial artery as the primary access site during PCI, patients presenting in CS are often treated via the transfemoral access site (TFA); and commentators have suggested that CS remains the final frontier that has given even experienced radial operators pause. We studied the use of TRA in patients presenting in CS in a nonselected high-risk cohort from the British Cardiovascular Intervention database over a 7-year period (2006-2012). METHODS: Mortality (30-day) and major adverse cardiac and cerebrovascular events (a composite of in-hospital mortality, in-hospital myocardial reinfarction, target vessel revascularization, and cerebrovascular events) were studied based on TFA and TRA utilization in CS patients. The influence of access site selection was studied in 7,231 CS patients; TFA was used in 5,354 and TRA in 1,877 patients. RESULTS: Transradial access site was independently associated with a lower 30-day mortality (hazard ratio [HR] 0.56, 95% CI 0.46-0.69, P = 0 < .001), in-hospital major adverse cardiac and cerebrovascular events (HR 0.64, 95% CI 0.53-0.76, P < .0001) and major bleeding (HR 0.37, 95% CI 0.18-0.73, P = .004). CONCLUSIONS: Although the majority of PCI cases performed in patients with cardiogenic shock in the United Kingdom are performed through the TFA, the radial artery represents an alternative viable access site in this high-risk cohort of patients in experienced centers.


Subject(s)
Femoral Artery , Myocardial Infarction/therapy , Percutaneous Coronary Intervention/methods , Radial Artery , Shock, Cardiogenic/therapy , Aged , Cohort Studies , Female , Hemorrhage/epidemiology , Hospital Mortality , Humans , Male , Multivariate Analysis , Myocardial Infarction/complications , Myocardial Infarction/mortality , Proportional Hazards Models , Retrospective Studies , Shock, Cardiogenic/etiology , Shock, Cardiogenic/mortality , Stroke/epidemiology , Treatment Outcome , United Kingdom
15.
Cardiovasc Res ; 103(1): 47-59, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24776599

ABSTRACT

AIMS: Tumour necrosis factor-α (TNF-α) plays a key role in the regulation of cardiac contractility. Although cardiomyocytes are known to express the TNF-α receptors (TNFRs), the mechanism of TNF-α signal transmission is incompletely understood. The aim of this study was to investigate whether the tumour suppressor Ras-association domain family protein 1 isoform A (RASSF1A) modulates TNF-α signalling in cardiomyocytes. METHODS AND RESULTS: We used RASSF1A knockout (RASSF1A(-/-)) mice and wild-type (WT) littermates in this study. Acute stimulation with a low dose of TNF-α (10 µg/kg iv) increased cardiac contractility and intracellular calcium transients' amplitude in WT mice. In contrast, RASSF1A(-/-) mice showed a blunted contractile response. Mechanistically, RASSF1A was essential in the formation of the TNFR complex (TNFRC), where it functions as an adaptor molecule to facilitate the recruitment of TNFR type 1-associated death domain protein and TNFR-associated factor 2 to form the TNF-α receptor complex. In the absence of RASSF1A, signal transmission from the TNF-α receptor complex to the downstream effectors, such as cytoplasmic phospholipase A2 and protein kinase A, was attenuated leading to the reduction in the activation of calcium handling molecules, such as L-type Ca(2+) channel and ryanodine receptors. CONCLUSION: Our data indicate an essential role of RASSF1A in regulating TNF-α signalling in cardiomyocytes, with RASSF1A being key in the formation of the TNFRC and in signal transmission to the downstream targets.


Subject(s)
Myocytes, Cardiac/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Calcium Signaling , Mice , Mice, Knockout , Myocardial Contraction/physiology , NF-kappa B/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/metabolism , Sequence Deletion , Signal Transduction , TNF Receptor-Associated Death Domain Protein/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
16.
PLoS One ; 9(2): e88577, 2014.
Article in English | MEDLINE | ID: mdl-24551120

ABSTRACT

BACKGROUND: Contemporary data remains limited regarding mortality and major adverse cardiac events (MACE) outcomes in patients undergoing PCI for different manifestations of coronary artery disease. OBJECTIVES: We evaluated mortality and MACE outcomes in patients treated with PCI for STEMI (ST-elevation myocardial infarction), NSTEMI (non ST-elevation myocardial infarction) and stable angina through analysis of data derived from the Nobori-2 study. METHODS: Clinical endpoints were cardiac mortality and MACE (a composite of cardiac death, myocardial infarction and target vessel revascularization). RESULTS: 1909 patients who underwent PCI were studied; 1332 with stable angina, 248 with STEMI and 329 with NSTEMI. Age-adjusted Charlson co-morbidity index was greatest in the NSTEMI cohort (3.78±1.91) and lowest in the stable angina cohort (3.00±1.69); P<0.0001. Following Cox multivariate analysis cardiac mortality was independently worse in the NSTEMI vs the stable angina cohort (HR 2.31 (1.10-4.87), p = 0.028) but not significantly different for STEMI vs stable angina cohort (HR 0.72 (0.16-3.19), p = 0.67). Similar observations were recorded for MACE (<180 days) (NSTEMI vs stable angina: HR 2.34 (1.21-4.55), p = 0.012; STEMI vs stable angina: HR 2.19 (0.97-4.98), p = 0.061. CONCLUSIONS: The longer-term Cardiac mortality and MACE were significantly worse for patients following PCI for NSTEMI even after adjustment of clinical demographics and Charlson co-morbidity index whilst the longer-term prognosis of patients following PCI STEMI was favorable, with similar outcomes as those patients with stable angina following PCI.


Subject(s)
Acute Coronary Syndrome/therapy , Angina, Stable/therapy , Aged , Death , Demography , Drug-Eluting Stents , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Proportional Hazards Models , Prospective Studies , Treatment Outcome
17.
EuroIntervention ; 10(7): 824-32, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24472679

ABSTRACT

Transradial (TR) coronary intervention is associated with fewer access-site-related bleeding complications and is independently associated with a lower risk of mortality following PCI compared to procedures undertaken through the femoral route. However, recent studies that have undertaken imaging of the radial artery through the use of IVUS and OCT, as well as histological studies, suggest that TR cardiac catheterisation is associated with significant injury to the radial artery wall resulting in significant endothelial cell dysfunction. The vascular endothelium plays a central role in the regulation of vascular tone, angiogenesis and vascular remodelling through the release of vasoactive mediators in response to a variety of stimuli. Hence, trauma to the vascular endothelium and subsequent changes in endothelial cell function may contribute to patterns of injury such as intimal hyperplasia and radial artery occlusion observed following TR cardiac catheterisation. Such injury patterns to the radial artery following TR procedures may limit the success and future utility of the TR approach. Minimisation of radial artery injury should be a key procedural component of procedures undertaken through the transradial approach.


Subject(s)
Percutaneous Coronary Intervention/methods , Radial Artery/pathology , Vascular Diseases/prevention & control , Anticoagulants/therapeutic use , Cardiac Catheterization , Endothelium, Vascular/physiology , Hemostasis , Humans , NF-kappa B/physiology , Percutaneous Coronary Intervention/adverse effects , Spasm/prevention & control , Tomography, Optical Coherence , Ultrasonography, Interventional , Vascular Diseases/etiology
18.
Heart Fail Rev ; 19(5): 669-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23856884

ABSTRACT

Pacing-induced ventricular dysfunction and pacing-induced cardiomyopathy (PiCMP) are recognized complications of chronic right ventricular pacing. Alterations in myocardial perfusion and sympathetic innervation contribute to the development of pacing-induced heart disease. However, it is unlikely that these are the only processes involved. Matrix metalloproteinases (MMPs) are proteolytic enzymes that degrade the collagenous extracellular matrix and play a central role in left ventricular remodelling during the development of heart failure. While the pathophysiological mechanisms and altered MMP expression that occur in chronic pressure overload, ischaemic and non-ischaemic dilated cardiomyopathy have been defined, those that occur in the clinical setting of pacing-induced ventricular dysfunction and PiCMP have not been reported. Here we review the clinical epidemiology of pacing-induced ventricular dysfunction and discuss how data derived from animal models provide insight into how changes in MMP expression and function contribute to the development of PiCMP. The review concludes by exploring pacing strategies that may be used to prevent pacing-induced ventricular dysfunction.


Subject(s)
Cardiac Pacing, Artificial , Heart Failure/enzymology , Heart Ventricles/physiopathology , Matrix Metalloproteinases/metabolism , Ventricular Dysfunction, Left/enzymology , Animals , Heart Failure/physiopathology , Humans , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/physiology
19.
J Card Fail ; 19(8): 592-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23910590

ABSTRACT

The complexity of standard medical treatment for heart failure is growing, and such therapy typically involves 5 or more different medications. Given these pressures, there is increasing interest in harnessing cardiovascular biomarkers for clinical application to more effectively guide diagnosis, risk stratification, and therapy. It may be possible to realize an era of personalized medicine for heart failure treatment in which therapy is optimized and costs are controlled. The direct mechanistic coupling of biologic processes and therapies achieved in cancer treatment remains elusive in heart failure. Recent clinical trials and meta-analyses of biomarkers in heart failure have produced conflicting evidence. In this article, which comprises a summary of discussions from the Global Cardiovascular Clinical Trialists Forum held in Paris, France, we offer a brief overview of the background and rationale for biomarker testing in heart failure, describe opportunities and challenges from a regulatory perspective, and summarize current positions from government agencies in the United States and European Union.


Subject(s)
European Union , Heart Failure/metabolism , Heart Failure/therapy , Biomarkers/metabolism , Europe/epidemiology , Heart Failure/epidemiology , Humans , Paris , Randomized Controlled Trials as Topic/trends , Treatment Outcome , United States/epidemiology
20.
J Pharm Pharm Sci ; 16(2): 217-30, 2013.
Article in English | MEDLINE | ID: mdl-23958191

ABSTRACT

PURPOSE: ATPases, which constitute a major category of ion transporters in the human body, have a variety of significant biological and pathological roles. However, the lack of high throughput assays for ATPases has significantly limited drug discovery in this area. We have recently found that the genetic deletion of the ATP dependent calcium pump PMCA4 (plasma membrane calcium/calmodulin dependent ATPase, isoform 4) results in infertility in male mice due to a selective defect in sperm motility. In addition, recent discoveries in humans have indicated that a single nucleotide polymorphism (SNP) in the PMCA4 gene determines the susceptibility towards malaria plasmodium infection. Therefore, there is an urgent need to develop specific PMCA4 inhibitors. In the current study, we aim to optimise and validate a high throughput screening compatible assay using recombinantly expressed PMCA4 and the HTRF® Transcreener® ADP (TR-FRET) assay to screen a drug library. METHODS AND RESULTS: PMCA4 membrane microsomes were prepared from HEK293 cells overexpressing PMCA4. Western blot quantification revealed nearly nine-fold increased expression of PMCA4 compared to LacZ (control virus)-infected cells. Maximal PMCA4 microsomal activity was achieved in the TR-FRET assay with 15ng/µl microsomal concentration, 30-minute pre-incubation with compounds at 37°C, and calcium buffering with 1mM EGTA providing 1µM free-calcium. Finally a dose-response curve for carboxyeosin (a non-specific PMCA inhibitor) under optimised conditions showed significant PMCA4 inhibition. Upon confirmation that the assay was suitable for high-throughput screening, we have screened the ChemBioNet small molecule library (~21,000 compounds) against the PMCA4 assay to identify those that are its apparent inhibitors. This screening yielded 1,494 primary hits. CONCLUSIONS: We have optimised the HTRF® Transcreener® ADP assay for high-throughput screening to identify PMCA4 inhibitors. The output of the screening campaign has provided preliminary chemical starting points that could be further developed to specific PMCA4 inhibitors for non-hormonal contraception or anti-malaria therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Contraception , HEK293 Cells , Humans , Malaria/drug therapy , Malaria/metabolism , Microsomes/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...