scispace - formally typeset
Search or ask a question

Showing papers by "Edward G. Lakatta published in 2016"


Journal ArticleDOI
Cristian Pattaro, Alexander Teumer1, Mathias Gorski2, Audrey Y. Chu3  +732 moreInstitutions (157)
TL;DR: A meta-analysis of genome-wide association studies for estimated glomerular filtration rate suggests that genetic determinants of eGFR are mediated largely through direct effects within the kidney and highlight important cell types and biological pathways.
Abstract: Reduced glomerular filtration rate defines chronic kidney disease and is associated with cardiovascular and all-cause mortality. We conducted a meta-analysis of genome-wide association studies for estimated glomerular filtration rate (eGFR), combining data across 133,413 individuals with replication in up to 42,166 individuals. We identify 24 new and confirm 29 previously identified loci. Of these 53 loci, 19 associate with eGFR among individuals with diabetes. Using bioinformatics, we show that identified genes at eGFR loci are enriched for expression in kidney tissues and in pathways relevant for kidney development and transmembrane transporter activity, kidney structure, and regulation of glucose metabolism. Chromatin state mapping and DNase I hypersensitivity analyses across adult tissues demonstrate preferential mapping of associated variants to regulatory regions in kidney but not extra-renal tissues. These findings suggest that genetic determinants of eGFR are mediated largely through direct effects within the kidney and highlight important cell types and biological pathways.

409 citations


Journal ArticleDOI
Georg Ehret1, Georg Ehret2, Teresa Ferreira3, Daniel I. Chasman4  +372 moreInstitutions (101)
TL;DR: In this article, the authors identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals, and 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues.
Abstract: To dissect the genetic architecture of blood pressure and assess effects on target organ damage, we analyzed 128,272 SNPs from targeted and genome-wide arrays in 201,529 individuals of European ancestry, and genotypes from an additional 140,886 individuals were used for validation. We identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals. The 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues. The 66 index SNPs combined in a risk score showed comparable effects in 64,421 individuals of non-European descent. The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney. Our findings expand current knowledge of blood pressure-related pathways and highlight tissues beyond the classical renal system in blood pressure regulation.

332 citations


01 Jan 2016
TL;DR: The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney, expanding current knowledge of blood pressure–related pathways and highlighting tissues beyond the classical renal system in blood pressure regulation.

272 citations


Journal ArticleDOI
Pim van der Harst1, Jessica van Setten2, Niek Verweij1, Georg Vogler3  +182 moreInstitutions (54)
TL;DR: A genome-wide association meta-analysis of 4 QRS traits in up to 73,518 individuals of European ancestry provides new insights into genes and biological pathways controlling myocardial mass and may help identify novel therapeutic targets.

109 citations


Journal ArticleDOI
TL;DR: This review will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.
Abstract: Aging is the major risk factor for cardiovascular diseases, which are the leading cause of death in the United States. Traditionally, the effort to prevent cardiovascular disease has been focused on addressing the conventional risk factors, including hypertension, hyperglycemia, hypercholesterolemia, and high circulating levels of triglycerides. However, recent preclinical studies have identified new approaches to combat cardiovascular disease. Calorie restriction has been reproducibly shown to prolong lifespan in various experimental model animals. This has led to the development of calorie restriction mimetics and other pharmacological interventions capable to delay age-related diseases. In this review, we will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.

60 citations



Journal ArticleDOI
TL;DR: Of all PWA-derived biomarkers, SC and DC were consistently identified as valuable parameters for incremental cardiovascular risk prediction in two large prospective cohorts and could independently and consistently predict cardiovascular mortality in both cohorts and the combined cohort.

39 citations


Journal ArticleDOI
TL;DR: A high short-term BPV is significantly predictive of long-term cardiovascular mortality in untreated hypertensive but not normotensive community-based subjects, independently of office or 24-hour SBP.

39 citations


Journal ArticleDOI
TL;DR: Changes in mechanisms intrinsic to pacemaker cells increase the average BIs and BIV in the mice of advanced age, and compensation reduces the BIV due to both the imbalance of autonomic neural input to thepacemaker cells and altered pacemaker cell responses to neural input.
Abstract: Summary We aimed to determine how age-associated changes in mechanisms extrinsic and intrinsic to pacemaker cells relate to basal beating interval variability (BIV) reduction in vivo. Beating intervals (BIs) were measured in aged (23–25 months) and adult (3–4 months) C57BL/6 male mice (i) via ECG in vivo during light anesthesia in the basal state, or in the presence of 0.5 mg mL−1 atropine + 1 mg mL−1 propranolol (in vivo intrinsic conditions), and (ii) via a surface electrogram, in intact isolated pacemaker tissue. BIV was quantified in both time and frequency domains using linear and nonlinear indices. Although the average basal BI did not significantly change with age under intrinsic conditions in vivo and in the intact isolated pacemaker tissue, the average BI was prolonged in advanced age. In vivo basal BIV indices were found to be reduced with age, but this reduction diminished in the intrinsic state. However, in pacemaker tissue BIV indices increased in advanced age vs. adults. In the isolated pacemaker tissue, the sensitivity of the average BI and BIV in response to autonomic receptor stimulation or activation of mechanisms intrinsic to pacemaker cells by broad-spectrum phosphodiesterase inhibition declined in advanced age. Thus, changes in mechanisms intrinsic to pacemaker cells increase the average BIs and BIV in the mice of advanced age. Autonomic neural input to pacemaker tissue compensates for failure of molecular intrinsic mechanisms to preserve average BI. But this compensation reduces the BIV due to both the imbalance of autonomic neural input to the pacemaker cells and altered pacemaker cell responses to neural input.

38 citations


Journal ArticleDOI
TL;DR: High basal CaMKII activation modifies the phosphorylation state of Ca( 2+) cycling proteins PLB, RyR, L-type Ca(2+) channels, adjusting LCR period and characteristics, and ultimately regulates both normal and reserve cardiac pacemaker function.
Abstract: Spontaneous beating of the heart pacemaker, the sinoatrial node, is generated by sinoatrial node cells (SANC) due to gradual change of the membrane potential called diastolic depolarization (DD). Spontaneous, submembrane local Ca(2+) releases (LCR) from ryanodine receptors (RyR) occur during late DD and activate an inward Na(+)/Ca(2+)exchange current to boost the DD rate and fire an action potential (AP). Here we studied the extent of basal Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation and the role of basal CaMKII-dependent protein phosphorylation in generation of LCRs and regulation of normal automaticity of intact rabbit SANC. The basal level of activated (autophosphorylated) CaMKII in rabbit SANC surpassed that in ventricular myocytes (VM) by approximately twofold, and this was accompanied by high basal level of protein phosphorylation. Specifically, phosphorylation of phospholamban (PLB) at the CaMKII-dependent Thr(17) site was approximately threefold greater in SANC compared with VM, and RyR phosphorylation at CaMKII-dependent Ser(2815) site was ∼10-fold greater in the SA node, compared with that in ventricle. CaMKII inhibition reduced phosphorylation of PLB and RyR, decreased LCR size, increased LCR periods (time from AP-induced Ca(2+) transient to subsequent LCR), and suppressed spontaneous SANC firing. Graded changes in CaMKII-dependent phosphorylation (indexed by PLB phosphorylation at the Thr(17)site) produced by CaMKII inhibition, β-AR stimulation or phosphodiesterase inhibition were highly correlated with changes in SR Ca(2+) replenishment times and LCR periods and concomitant changes in spontaneous SANC cycle lengths (R(2) = 0.96). Thus high basal CaMKII activation modifies the phosphorylation state of Ca(2+) cycling proteins PLB, RyR, L-type Ca(2+) channels (and likely others), adjusting LCR period and characteristics, and ultimately regulates both normal and reserve cardiac pacemaker function.

34 citations


Journal ArticleDOI
TL;DR: The increase of collagen content is a hallmark of arterial aging, and this work provides a potential mechanistic link between RAGE signaling, NF-κB activation, and aging-associated arterial alterations in structure and function.
Abstract: Advanced glycation end products (AGEs) are sugar-modified biomolecules that accumulate in the body with advancing age, and are implicated in the development of multiple age-associated structural and functional abnormities and diseases. It has been well documented that AGEs signal via their receptor RAGE to activate several cellular programs including NF-κB, leading to inflammation. A large number of stimuli can activate NF-κB; yet different stimuli, or the same stimulus for NF-κB in different cellular settings, produce a very different transcriptional landscape and physiological outcome. The NF-κB barcode hypothesis posits that cellular network dynamics generate signal-specific post-translational modifications, or a "barcode" to NF-κB, and that a signature "barcode" mediates a specific gene expression pattern. In the current study, we established that AGE-RAGE signaling results in NF-κB activation that directs collagen Ia1 and Ia2 expression. We further demonstrated that AGE-RAGE signal induces phosphorylation of RelA at three specific residues, T254, S311, and S536. These modifications are required for transcription of collagen I genes and are a consequence of cellular network dynamics. The increase of collagen content is a hallmark of arterial aging, and our work provides a potential mechanistic link between RAGE signaling, NF-κB activation, and aging-associated arterial alterations in structure and function.

Journal ArticleDOI
TL;DR: It is concluded that signaling via cAMP generated by Ca(2+)/CaM-activated AC in SANC lipid raft domains is limited by cAMP degradation by Ca (2+/CaM)-activated PDE1A in non-lipid raft domains.

Journal ArticleDOI
TL;DR: High salt intake in young normotensive rats can induce vascular fibrosis via pressure-independent/MBG-dependent mechanisms, and this remodeling is reduced by immunoneutralization of MBG.

Journal ArticleDOI
TL;DR: SANC automaticity is tightly regulated by ENa, ECa, and ENCX via a complex interplay of numerous key clock components that regulate SANC clock coupling.
Abstract: Coupling of an intracellular Ca(2+) clock to surface membrane ion channels, i.e., a "membrane clock, " via coupling of electrochemical Na(+) and Ca(2+) gradients (ENa and ECa, respectively) has been theorized to regulate sinoatrial nodal cell (SANC) normal automaticity. To test this hypothesis, we measured responses of [Na(+)]i, [Ca(2+)]i, membrane potential, action potential cycle length (APCL), and rhythm in rabbit SANCs to Na(+)/K(+) pump inhibition by the digitalis glycoside, digoxigenin (DG, 10-20 μmol/l). Initial small but significant increases in [Na(+)]i and [Ca(2+)]i and reductions in ENa and ECa in response to DG led to a small reduction in maximum diastolic potential (MDP), significantly enhanced local diastolic Ca(2+) releases (LCRs), and reduced the average APCL. As [Na(+)]i and [Ca(2+)]i continued to increase at longer times following DG exposure, further significant reductions in MDP, ENa, and ECa occurred; LCRs became significantly reduced, and APCL became progressively and significantly prolonged. This was accompanied by increased APCL variability. We also employed a coupled-clock numerical model to simulate changes in ENa and ECa simultaneously with ion currents not measured experimentally. Numerical modeling predicted that, as the ENa and ECa monotonically reduced over time in response to DG, ion currents (ICaL, ICaT, If, IKr, and IbNa) monotonically decreased. In parallel with the biphasic APCL, diastolic INCX manifested biphasic changes; initial INCX increase attributable to enhanced LCR ensemble Ca(2+) signal was followed by INCX reduction as ENCX (ENCX = 3ENa - 2ECa) decreased. Thus SANC automaticity is tightly regulated by ENa, ECa, and ENCX via a complex interplay of numerous key clock components that regulate SANC clock coupling.

Journal ArticleDOI
TL;DR: The consistent association of specific clusters of MetS components with extremely thick (older) large artery cross-culturally suggests that identification of those clusters in clinical practice will facilitate a personalized health care and a better - i.e. more healthy and cost-effective - prevention of major cardiovascular (CV) events.

Book ChapterDOI
01 Jan 2016
TL;DR: In this article, the authors provide an overview of cardiovascular aging in humans and mammalian models, and discuss the recent advances in understanding how different hallmarks of aging play their roles in the pathogenesis of cardiovascular ageing.
Abstract: Cardiovascular diseases are the leading cause of death for population over 65 years of age. With the progressive increase of population in this age group, it is crucial to understand the interaction between aging and cardiovascular diseases. Aging results in structural changes and functional decline of the cardiovascular system and is a major risk factor for cardiovascular diseases. Aging not only increases the prevalence of cardiovascular diseases but is also associated with impaired responses to cardiovascular diseases. This supports the notion that age-related changes in the cardiovascular system superimpose and interact with the pathophysiological mechanisms that lead to cardiovascular disease. This chapter will provide an overview of cardiovascular aging in humans and mammalian models, and discuss the recent advances in understanding how different hallmarks of aging play their roles in the pathogenesis of cardiovascular aging.

Journal ArticleDOI
21 Sep 2016-PLOS ONE
TL;DR: In this article, the authors characterized mRAGEV4 isoreceptor in comparison with the full-length m-RAGE (mRAGEFL) and showed that the two isoforms display very different cellular behaviors.
Abstract: The receptor for advanced glycation end products (RAGE) is a multi-ligand, immunoglobulin-like receptor that has been implicated in aging-associated diseases. Recent studies have demonstrated that both human and murine Ager genes undergo extensive alternative splicing that generates multiple putative transcripts encoding different receptor isoforms. Except for the soluble isoform (esRAGE), the majority of putative RAGE isoforms remain unstudied. Profiling of murine Ager transcripts showed that variant transcript 4 (mRAGE_v4), the second most abundant transcript in lungs and multiple other tissues, encodes a receptor that lacks nine residues located within the C2 extracellular section close to the trans-membrane domain. We therefore characterized mRAGEV4 isoreceptor in comparison with the full-length mRAGE (mRAGEFL). Although differing in only nine residues, mRAGEFL and mRAGEV4 display very different cellular behaviors. While mRAGEFL undergoes constitutive, extensive shedding in the cell to generate sRAGE, mRAGEV4 hardly sheds. In addition, we found that while mRAGEFL can localize to both the plasma membrane and the endosome, mRAGEV4 is exclusively localized to the plasma membrane. These very different cellular localization patterns suggest that, in addition to their roles in sRAGE production, mRAGEFL and mRAGEV4 may play distinct, spatiotemporal roles in signaling and innate immune responses. Compared to mice, humans do not have the v4 transcript. Although hRAGE, like mRAGEFL, also localizes to the plasma membrane and the endosome, its rate of constitutive shedding is significantly lower. These observations provide valuable information regarding RAGE biology, and serve as a reference by which to create mouse models relating to human diseases.

Journal ArticleDOI
TL;DR: The rat carotid balloon injury is a well-established surgical model that has been used to study arterial remodeling and vascular cell proliferation and is also a valuable model system to test, and to evaluate therapeutics and drugs that negate maladaptive remodeling in the vessel.
Abstract: The rat carotid balloon injury is a well-established surgical model that has been used to study arterial remodeling and vascular cell proliferation. It is also a valuable model system to test, and to evaluate therapeutics and drugs that negate maladaptive remodeling in the vessel. The injury, or barotrauma, in the vessel lumen caused by an inflated balloon via an inserted catheter induces subsequent neointimal growth, often leading to hyperplasia or thickening of the vessel wall that narrows, or obstructs the lumen. The method described here is sufficiently sensitive, and the results can be obtained in relatively short time (2 weeks after the surgery). The efficacy of the drug or therapeutic against the induced-remodeling can be evaluated either by the post-mortem pathological and histomorphological analysis, or by ultrasound sonography in live animals. In addition, this model system has also been used to determine the therapeutic window or the time course of the administered drug. These studies can leadto the development of a better administrative strategy and a better therapeutic outcome. The procedure described here provides a tool for translational studies that bring drug and therapeutic candidates from bench research to clinical applications.

Book ChapterDOI
01 Jan 2016
TL;DR: The profile of arterial aging is intertwined with hypertension and atherosclerosis at the molecular, cellular, vascular, and clinical levels, rendering the aged arterial wall a fertile soil for their pathogenesis.
Abstract: A chronic increase in the production of inflammatory signals drives age-associated arterial structural remodeling and functional changes, including diffuse arterial intimal-medial thickening and stiffening. Under the microscope, the aged artery is characterized by increases in vascular smooth muscle cell apoptosis, senescence, invasion, proliferation, inflammatory molecule secretion, endothelial disruption, prothrombosis, glycoxidation, fibrosis, elastin fragmentation, calcification, and amyloidosis. These adverse cellular and molecular events occur within aged arterial wall experimentally in young animals under inflammatory stress conditions, and are attenuated in old animals by interfering with proinflammatory signals. Importantly, the profile of arterial aging is intertwined with hypertension and atherosclerosis at the molecular, cellular, vascular, and clinical levels, rendering the aged arterial wall a fertile soil for their pathogenesis. Thus, early and effective strategies to suppress age-associated arterial proinflammation may be realistic approaches to curb the initiation and progression of age-associated cardiovascular diseases such as hypertension and atherosclerosis.

Journal ArticleDOI
TL;DR: Super-resolution structured illumination microscopy is used achieving approximately double the resolution of conventional microscopy to measure both 3D-distribution of RyRs and 2D-calcium dynamics in isolated rabbit SA-node cells and reveals that LCRs occur mainly within ∼1µm under the cell surface, in agreement with immunofluorescence data on RyR cluster localization.

Journal Article
TL;DR: Arginine-Vasopressin exerts a variety of effects, in addition to its antidiuretic effect via V2 receptors in the kidneys, AVP has vasoconstrictive effects via its action on V1a receptor in the peripheral vasculature.
Abstract: Arginine-Vasopressin (AVP) exerts a variety of effects. in addition to its antidiuretic effect via V2 receptors in the kidneys, AVP has vasoconstrictive effects via its action on V1a receptors in the peripheral vasculature. Whether patients with heart failure and preserved ejection fraction (HFpEF) demonstrate increased levels of AVP, and whether this relates to left ventricular remodeling is unknown. Methods: We studied 26 subjects with heart failure and reduced ejection fraction (HFrEF), 28 subjects with HFpEF, and 95 control subjects without heart failure. AVP was measured ELISA (Cayman Chemicals ; USA). In a subgroup of subjects (n=110) we measured LV mass with magnetic resonance imaging (SSFP cine sequences), with LV segmentation using CMR42 (Circle CV Imaging; Calgary, Canada). Results: AVP levels were significantly greater in HFpEF (1.01; 95%CI=0.84 to 1.19)Compared to either HFrEF (0.72; 95%CI=0.58 to 0.87) or controls without HF (0.69; 95%CI=0.62 to 0.75; ANOVA P<0.0001). After adjustme...