Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Sensors (Basel) ; 23(12)2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37420569

ABSTRACT

In the paper, the effect of spontaneous Brillouin scattering (SpBS) is analyzed as a noise source in distributed acoustic sensors (DAS). The intensity of the SpBS wave fluctuates over time, and these fluctuations increase the noise power in DAS. Based on experimental data, the probability density function (PDF) of the spectrally selected SpBS Stokes wave intensity is negative exponential, which corresponds to the known theoretical conception. Based on this statement, an estimation of the average noise power induced by the SpBS wave is given. This noise power equals the square of the average power of the SpBS Stokes wave, which in turn is approximately 18 dB lower than the Rayleigh backscattering power. The noise composition in DAS is determined for two configurations, the first for the initial backscattering spectrum and the second for the spectrum in which the SpBS Stokes and anti-Stokes waves are rejected. It is established that in the analyzed particular case, the SpBS noise power is dominant and exceeds the powers of the thermal, shot, and phase noises in DAS. Accordingly, by rejecting the SpBS waves at the photodetector input, it is possible to reduce the noise power in DAS. In our case, this rejection is carried out by an asymmetric Mach-Zehnder interferometer (MZI). The rejection of the SpBS wave is most relevant for broadband photodetectors, which are associated with the use of short probing pulses to achieve short gauge lengths in DAS.


Subject(s)
Fertilization , Heart Rate , Likelihood Functions
2.
Sensors (Basel) ; 22(23)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36502184

ABSTRACT

A simple and cost-effective architecture of a distributed acoustic sensor (DAS) or a phase-OTDR for engineering geology is proposed. The architecture is based on the dual-pulse acquisition principle, where the dual probing pulse is formed via an unbalanced Michelson interferometer (MI). The necessary phase shifts between the sub-pulses of the dual-pulse are introduced using a 3 × 3 coupler built into the MI. Laser pulses are generated by direct modulation of the injection current, which obtains optical pulses with a duration of 7 ns. The use of an unbalanced MI for the formation of a dual-pulse reduces the requirements for the coherence of the laser source, as the introduced delay between sub-pulses is compensated in the fiber under test (FUT). Therefore, a laser with a relatively broad spectral linewidth of about 1 GHz can be used. To overcome the fading problem, as well as to ensure the linearity of the DAS response, the averaging of over 16 optical frequencies is used. The performance of the DAS was tested by recording a strong vibration impact on a horizontally buried cable and by the recording of seismic waves in a borehole in the seabed.


Subject(s)
Engineering , Geology , Cost-Benefit Analysis , Heart Rate , Acoustics
3.
Sci Rep ; 12(1): 7314, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35513538

ABSTRACT

Plasmalemmal ATP sensitive potassium (KATP) channels are recognized metabolic sensors, yet their cellular reach is less well understood. Here, transgenic Kir6.2 null hearts devoid of the KATP channel pore underwent multiomics surveillance and systems interrogation versus wildtype counterparts. Despite maintained organ performance, the knockout proteome deviated beyond a discrete loss of constitutive KATP channel subunits. Multidimensional nano-flow liquid chromatography tandem mass spectrometry resolved 111 differentially expressed proteins and their expanded network neighborhood, dominated by metabolic process engagement. Independent multimodal chemometric gas and liquid chromatography mass spectrometry unveiled differential expression of over one quarter of measured metabolites discriminating the Kir6.2 deficient heart metabolome. Supervised class analogy ranking and unsupervised enrichment analysis prioritized nicotinamide adenine dinucleotide (NAD+), affirmed by extensive overrepresentation of NAD+ associated circuitry. The remodeled metabolome and proteome revealed functional convergence and an integrated signature of disease susceptibility. Deciphered cardiac patterns were traceable in the corresponding plasma metabolome, with tissue concordant plasma changes offering surrogate metabolite markers of myocardial latent vulnerability. Thus, Kir6.2 deficit precipitates multiome reorganization, mapping a comprehensive atlas of the KATP channel dependent landscape.


Subject(s)
NAD , Proteome , Adenosine Triphosphate , Heart , KATP Channels/genetics , KATP Channels/metabolism , NAD/metabolism , Proteome/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923625

ABSTRACT

Sarcolemmal α2 adrenoceptors (α2-AR), represented by α2A, α2B and α2C isoforms, can safeguard cardiac muscle under sympathoadrenergic surge by governing Ca2+ handling and contractility of cardiomyocytes. Cardiomyocyte-specific targeting of α2-AR would provide cardiac muscle-delimited stress control and enhance the efficacy of cardiac malfunction treatments. However, little is known about the specific contribution of the α2-AR subtypes in modulating cardiomyocyte functions. Herein, we analyzed the expression profile of α2A, α2B and α2C subtypes in mouse ventricle and conducted electrophysiological antagonist assay evaluating the contribution of these isoforms to the suppression of L-type Ca2+ current (ICaL). Patch-clamp electro-pharmacological studies revealed that the α2-agonist-induced suppression of ICaL involves mainly the α2C, to a lesser extent the α2B, and not the α2A isoforms. RT-qPCR evaluation revealed the presence of adra2b and adra2c (α2B and α2C isoform genes, respectively), but was unable to identify the expression of adra2a (α2A isoform gene) in the mouse left ventricle. Immunoblotting confirmed the presence only of the α2B and the α2C proteins in this tissue. The identified α2-AR isoform-linked regulation of ICaL in the mouse ventricle provides an important molecular substrate for the cardioprotective targeting.


Subject(s)
Action Potentials , Calcium Channels, L-Type/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Animals , Cells, Cultured , Heart Ventricles/cytology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Adrenergic, alpha-2/genetics
5.
Pharmacol Ther ; 197: 179-190, 2019 05.
Article in English | MEDLINE | ID: mdl-30703415

ABSTRACT

α2-adrenoceptor (α2-AR) isoforms, abundant in sympathetic synapses and noradrenergic neurons of the central nervous system, are integral in the presynaptic feed-back loop mechanism that moderates norepinephrine surges. We recently identified that postsynaptic α2-ARs, found in the myocellular sarcolemma, also contribute to a muscle-delimited feedback control capable of attenuating mobilization of intracellular Ca2+ and myocardial contractility. This previously unrecognized α2-AR-dependent rheostat is able to counteract competing adrenergic receptor actions in cardiac muscle. Specifically, in ventricular myocytes, nitric oxide (NO) and cGMP are the intracellular messengers of α2-AR signal transduction pathways that gauge the kinase-phosphatase balance and manage cellular Ca2+ handling preventing catecholamine-induced Ca2+ overload. Moreover, α2-AR signaling counterbalances phospholipase C - PKC-dependent mechanisms underscoring a broader cardioprotective potential under sympathoadrenergic and angiotensinergic challenge. Recruitment of such tissue-specific features of α2-AR under sustained sympathoadrenergic drive may, in principle, be harnessed to mitigate or prevent cardiac malfunction. However, cardiovascular disease may compromise peripheral α2-AR signaling limiting pharmacological targeting of these receptors. Prospective cardiac-specific gene or cell-based therapeutic approaches aimed at repairing or improving stress-protective α2-AR signaling may offer an alternative towards enhanced preservation of cardiac muscle structure and function.


Subject(s)
Heart/physiology , Receptors, Adrenergic, alpha-2/physiology , Sarcolemma/physiology , Animals , Feedback, Physiological , Heart Diseases/drug therapy , Heart Diseases/physiopathology , Humans
6.
Tissue Eng Part A ; 25(1-2): 145-158, 2019 01.
Article in English | MEDLINE | ID: mdl-30047313

ABSTRACT

IMPACT STATEMENT: The M3RNA (microencapsulated modified messenger RNA) platform is an approach to deliver messenger RNA (mRNA) in vivo, achieving a nonintegrating and viral-free approach to gene therapy. This technology was, in this study, tested for its utility in the myocardium, providing a unique avenue for targeted gene delivery into the freshly infarcted myocardial tissue. This study provides the evidentiary basis for the use of M3RNA in the heart through depiction of its performance in cultured cells, healthy rodent myocardium, and acutely injured porcine hearts. By testing the technology in large animal models of infarction, compatibility of M3RNA with current coronary intervention procedures was verified.


Subject(s)
Gene Transfer Techniques , Myocardial Infarction , Myocytes, Cardiac/metabolism , RNA, Messenger , Animals , Disease Models, Animal , HEK293 Cells , Humans , Luciferases/biosynthesis , Luciferases/genetics , Mice , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Swine
7.
PLoS One ; 12(5): e0177469, 2017.
Article in English | MEDLINE | ID: mdl-28531217

ABSTRACT

Hibernators have a distinctive ability to adapt to seasonal changes of body temperature in a range between 37°C and near freezing, exhibiting, among other features, a unique reversibility of cardiac contractility. The adaptation of myocardial contractility in hibernation state relies on alterations of excitation contraction coupling, which becomes less-dependent from extracellular Ca2+ entry and is predominantly controlled by Ca2+ release from sarcoplasmic reticulum, replenished by the Ca2+-ATPase (SERCA). We found that the specific SERCA inhibitor cyclopiazonic acid (CPA), in contrast to its effect in papillary muscles (PM) from rat hearts, did not reduce but rather potentiated contractility of PM from hibernating ground squirrels (GS). In GS ventricles we identified drastically elevated, compared to rats, expression of Orai1, Stim1 and Trpc1/3/4/5/6/7 mRNAs, putative components of store operated Ca2+ channels (SOC). Trpc3 protein levels were found increased in winter compared to summer GS, yet levels of Trpc5, Trpc6 or Trpc7 remained unchanged. Under suppressed voltage-dependent K+, Na+ and Ca2+ currents, the SOC inhibitor 2-aminoethyl diphenylborinate (2-APB) diminished whole-cell membrane currents in isolated cardiomyocytes from hibernating GS, but not from rats. During cooling-reheating cycles (30°C-7°C-30°C) of ground squirrel PM, 2-APB did not affect typical CPA-sensitive elevation of contractile force at low temperatures, but precluded the contractility at 30°C before and after the cooling. Wash-out of 2-APB reversed PM contractility to control values. Thus, we suggest that SOC play a pivotal role in governing the ability of hibernator hearts to maintain their function during the transition in and out of hibernating states.


Subject(s)
Hibernation , Indoles/pharmacology , Papillary Muscles/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/metabolism , Sciuridae/physiology , Animals , Calcium/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Male , Myocardial Contraction/drug effects , Papillary Muscles/drug effects , Rats , Rats, Sprague-Dawley , Sciuridae/metabolism , Signal Transduction/drug effects , Temperature
8.
J Mol Cell Cardiol ; 100: 9-20, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27659409

ABSTRACT

Sustained cardiac adrenergic stimulation has been implicated in the development of heart failure and ventricular dysrhythmia. Conventionally, α2 adrenoceptors (α2-AR) have been assigned to a sympathetic short-loop feedback aimed at attenuating catecholamine release. We have recently revealed the expression of α2-AR in the sarcolemma of cardiomyocytes and identified the ability of α2-AR signaling to suppress spontaneous Ca2+ transients through nitric oxide (NO) dependent pathways. Herein, patch-clamp measurements and serine/threonine phosphatase assay revealed that, in isolated rat cardiomyocytes, activation of α2-AR suppressed L-type Ca2+ current (ICaL) via stimulation of NO synthesis and protein kinase G- (PKG) dependent activation of phosphatase reactions, counteracting isoproterenol-induced ß-adrenergic activation. Under stimulation with norepinephrine (NE), an agonist of ß- and α-adrenoceptors, the α2-AR antagonist yohimbine substantially elevated ICaL at NE levels >10nM. Concomitantly, yohimbine potentiated triggered intracellular Ca2+ dynamics and contractility of cardiac papillary muscles. Therefore, in addition to the α2-AR-mediated feedback suppression of sympathetic and adrenal catecholamine release, α2-AR in cardiomyocytes can govern a previously unrecognized local cardiomyocyte-delimited stress-reactive signaling pathway. We suggest that such aberrant α2-AR signaling may contribute to the development of cardiomyopathy under sustained sympathetic drive. Indeed, in cardiomyocytes of spontaneously hypertensive rats (SHR), an established model of cardiac hypertrophy, α2-AR signaling was dramatically reduced despite increased α2-AR mRNA levels compared to normal cardiomyocytes. Thus, targeting α2-AR signaling mechanisms in cardiomyocytes may find implications in medical strategies against maladaptive cardiac remodeling associated with chronic sympathoadrenal stimulation.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Sarcolemma/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Calcium Signaling/drug effects , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cyclic GMP/metabolism , Disease Models, Animal , Male , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Protein Phosphatase 2/metabolism , Rats , Rats, Inbred SHR , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Sarcolemma/drug effects , Signal Transduction/drug effects
9.
Biochim Biophys Acta ; 1860(10): 2269-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27130881

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterized by the inability of patients to sustain a high level of ventilation resulting in perceived exertional discomfort and limited exercise capacity of leg muscles at average intracellular ATP levels sufficient to support contractility. METHODS: Myosin ATPase activity in biopsy samples from healthy and COPD individuals was implemented as a local nucleotide sensor to determine ATP diffusion coefficients within myofibrils. Ergometric parameters clinically measured during maximal exercise tests in both groups were used to define the rates of myosin ATPase reaction and aerobic ATP re-synthesis. The obtained parameters in combination with AK- and CK-catalyzed reactions were implemented to compute the kinetic and steady-state spatial ATP distributions within control and COPD sarcomeres. RESULTS: The developed reaction-diffusion model of two-dimensional sarcomeric space identified similar, yet extremely low nucleotide diffusion in normal and COPD myofibrils. The corresponding spatio-temporal ATP distributions, constructed during imposed exercise, predicted in COPD sarcomeres a depletion of ATP in the zones of overlap between actin and myosin filaments along the center axis at average cytosolic ATP levels similar to healthy muscles. CONCLUSIONS: ATP-depleted zones can induce rigor tension foci impairing muscle contraction and increase a risk for sarcomere damages. Thus, intra-sarcomeric diffusion restrictions at limited aerobic ATP re-synthesis can be an additional risk factor contributing to the muscle contractile deficiency experienced by COPD patients. GENERAL SIGNIFICANCE: This study demonstrates how restricted substrate mobility within a cellular organelle can provoke an energy imbalance state paradoxically occurring at abounding average metabolic resources.


Subject(s)
Adenosine Triphosphate/metabolism , Myofibrils/metabolism , Myosins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Aged , Biopsy , Cell Compartmentation/genetics , Diffusion , Female , Humans , Male , Middle Aged , Muscle Contraction/physiology , Myofibrils/pathology , Oxygen Consumption/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/therapy , Sarcomeres/metabolism , Sarcomeres/pathology
10.
Circ Cardiovasc Genet ; 7(5): 667-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25077947

ABSTRACT

BACKGROUND: Nuclear reprogramming inculcates pluripotent capacity by which de novo tissue differentiation is enabled. Yet, introduction of ectopic reprogramming factors may desynchronize natural developmental schedules. This study aims to evaluate the effect of imposed transgene load on the cardiogenic competency of induced pluripotent stem (iPS) cells. METHODS AND RESULTS: Targeted inclusion and exclusion of reprogramming transgenes (c-MYC, KLF4, OCT4, and SOX2) was achieved using a drug-inducible and removable cassette according to the piggyBac transposon/transposase system. Pulsed transgene overexpression, before iPS cell differentiation, hindered cardiogenic outcomes. Delayed in counterparts with maintained integrated transgenes, transgene removal enabled proficient differentiation of iPS cells into functional cardiac tissue. Transgene-free iPS cells generated reproducible beating activity with robust expression of cardiac α-actinin, connexin 43, myosin light chain 2a, α/ß-myosin heavy chain, and troponin I. Although operational excitation-contraction coupling was demonstrable in the presence or absence of transgenes, factor-free derivatives exhibited an expedited maturing phenotype with canonical responsiveness to adrenergic stimulation. CONCLUSIONS: A disproportionate stemness load, caused by integrated transgenes, affects the cardiogenic competency of iPS cells. Offload of transgenes in engineered iPS cells ensures integrity of cardiac developmental programs, underscoring the value of nonintegrative nuclear reprogramming for derivation of competent cardiogenic regenerative biologics.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Transgenes , Actinin/biosynthesis , Animals , Cardiac Myosins/biosynthesis , Cell Differentiation , Cell Separation , Cellular Reprogramming , Connexin 43/biosynthesis , Electrophysiology , Fibroblasts/metabolism , Flow Cytometry , Genetic Techniques , Kruppel-Like Factor 4 , Mice , Microscopy, Electron , Myosin Heavy Chains/biosynthesis , Myosin Light Chains/biosynthesis , Troponin I/biosynthesis
11.
J Mol Cell Cardiol ; 68: 66-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412533

ABSTRACT

Evidence suggests that intracellular Ca(2+) levels and contractility of cardiomyocytes can be modulated by targeting receptors other than already identified adrenergic or non-adrenergic sarcolemmal receptors. This study uncovers the presence in myocardial cells of adrenergic α2 (α2-AR) and imidazoline I1 (I1R) receptors. In isolated left ventricular myocytes generating stationary spontaneous Ca(2+) transients in the absence of triggered action potentials, the prototypic agonist of both receptors agmatine can activate corresponding signaling cascades with opposing outcomes on nitric oxide (NO) synthesis and intracellular Ca(2+) handling. Specifically, activation of α2-AR signaling through PI3 kinase and Akt/protein kinase B stimulates NO production and abolishes Ca(2+) transients, while targeting of I1R signaling via phosphatidylcholine-specific phospholipase C (PC-PLC) and protein kinase C (PKC) suppresses NO synthesis and elevates averaged intracellular Ca(2+). We identified that endothelial NO synthase (eNOS) is a major effector for both signaling cascades. According to the established eNOS transitions between active (Akt-dependent) and inactive (PKC-dependent) conformations, we suggest that balance between α2-AR and I1R signaling pathways sets eNOS activity, which by defining operational states of myocellular sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) can adjust Ca(2+) re-uptake and thereby cardiac inotropy. These results indicate that the conventional catalog of cardiomyocyte sarcolemmal receptors should be expanded by the α2-AR and I1R populations, unveiling previously unrecognized targets for endogenous ligands as well as for existing and potential pharmacological agents in cardiovascular medicine.


Subject(s)
Calcium Signaling , Imidazoline Receptors/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Agmatine/pharmacology , Animals , Benzofurans/pharmacology , Cells, Cultured , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Imidazoline Receptors/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
12.
J Mol Biol ; 419(1-2): 41-60, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22406676

ABSTRACT

Most cystic fibrosis is caused by a deletion of a single residue (F508) in CFTR (cystic fibrosis transmembrane conductance regulator) that disrupts the folding and biosynthetic maturation of the ion channel protein. Progress towards understanding the underlying mechanisms and overcoming the defect remains incomplete. Here, we show that the thermal instability of human ΔF508 CFTR channel activity evident in both cell-attached membrane patches and planar phospholipid bilayers is not observed in corresponding mutant CFTRs of several non-mammalian species. These more stable orthologs are distinguished from their mammalian counterparts by the substitution of proline residues at several key dynamic locations in first N-terminal nucleotide-binding domain (NBD1), including the structurally diverse region, the γ-phosphate switch loop, and the regulatory insertion. Molecular dynamics analyses revealed that addition of the prolines could reduce flexibility at these locations and increase the temperatures of unfolding transitions of ΔF508 NBD1 to that of the wild type. Introduction of these prolines experimentally into full-length human ΔF508 CFTR together with the already recognized I539T suppressor mutation, also in the structurally diverse region, restored channel function and thermodynamic stability as well as its trafficking to and lifetime at the cell surface. Thus, while cellular manipulations that circumvent its culling by quality control systems leave ΔF508 CFTR dysfunctional at physiological temperature, restoration of the delicate balance between the dynamic protein's inherent stability and channel activity returns a near-normal state.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Allosteric Site , Amino Acid Substitution , Animals , Anura , Carrier Proteins/metabolism , Cell Line, Transformed , Chickens , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , HEK293 Cells , Humans , Models, Molecular , Molecular Dynamics Simulation , Nucleotides/metabolism , Phosphate-Binding Proteins , Proline/metabolism , Protein Binding , Protein Folding , Protein Stability , Protein Transport/genetics , Protein Transport/physiology , Rabbits , Sharks , Sheep , Thermodynamics
13.
J Mol Cell Cardiol ; 52(2): 401-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21704043

ABSTRACT

Orchestrated excitation-contraction coupling in heart muscle requires adequate spatial arrangement of systems responsible for ion movement and metabolite turnover. Co-localization of regulatory and transporting proteins into macromolecular complexes within an environment of microanatomical cell components raises intracellular diffusion barriers that hamper the mobility of metabolites and signaling molecules. Compared to substrate diffusion in the cytosol, diffusional restrictions underneath the sarcolemma are much larger and could impede ion and nucleotide movement by a factor of 10(3)-10(5). Diffusion barriers thus seclude metabolites within the submembrane space enabling rapid and vectorial effector targeting, yet hinder energy supply from the bulk cytosolic space implicating the necessity for a shunting transfer mechanism. Here, we address principles of membrane protein compartmentation, phosphotransfer enzyme-facilitated interdomain energy transfer, and nucleotide signal dynamics at the subsarcolemma-cytosol interface. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Subject(s)
Cellular Microenvironment , Cytosol/metabolism , Myocytes, Cardiac/metabolism , Nucleotides/metabolism , Sarcolemma/metabolism , Animals , Diffusion , Energy Metabolism/physiology , Humans , Intracellular Space/metabolism , Ion Channels/metabolism , Multiprotein Complexes/metabolism , Protein Transport , Signal Transduction
15.
Crit Rev Biochem Mol Biol ; 45(6): 506-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20925594

ABSTRACT

Uniquely gated by intracellular adenine nucleotides, sarcolemmal ATP-sensitive K(+) (K(ATP)) channels have been typically assigned to protective cellular responses under severe energy insults. More recently, K(ATP) channels have been instituted in the continuous control of muscle energy expenditure under non-stressed, physiological states. These advances raised the question of how K(ATP) channels can process trends in cellular energetics within a milieu where each metabolic system is set to buffer nucleotide pools. Unveiling the mechanistic basis of the K(ATP) channel-driven thermogenic response in muscles thus invites the concepts of intracellular compartmentalization of energy and proteins, along with nucleotide signaling over diffusion barriers. Furthermore, it requires gaining insight into the properties of reversibility of intrinsic ATPase activity associated with K(ATP) channel complexes. Notwithstanding the operational paradigm, the homeostatic role of sarcolemmal K(ATP) channels can be now broadened to a wider range of environmental cues affecting metabolic well-being. In this way, under conditions of energy deficit such as ischemic insult or adrenergic stress, the operation of K(ATP) channel complexes would result in protective energy saving, safeguarding muscle performance and integrity. Under energy surplus, downregulation of K(ATP) channel function may find potential implications in conditions of energy imbalance linked to obesity, cold intolerance and associated metabolic disorders.


Subject(s)
Adenine Nucleotides/metabolism , Energy Metabolism , KATP Channels/metabolism , Muscles/physiology , Signal Transduction , Thermogenesis , Animals , Humans , Ion Channel Gating , Muscles/metabolism , Sarcolemma/physiology , Structure-Activity Relationship
16.
Cell Metab ; 11(1): 58-69, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20074528

ABSTRACT

Metabolic processes that regulate muscle energy use are major determinants of bodily energy balance. Here, we find that sarcolemmal ATP-sensitive K(+) (K(ATP)) channels, which couple membrane excitability with cellular metabolic pathways, set muscle energy expenditure under physiological stimuli. Disruption of K(ATP) channel function provoked, under conditions of unaltered locomotor activity and blood substrate availability, an extra energy cost of cardiac and skeletal muscle performance. Inefficient fuel metabolism in K(ATP) channel-deficient striated muscles reduced glycogen and fat body depots, promoting a lean phenotype. The propensity to lesser body weight imposed by K(ATP) channel deficit persisted under a high-fat diet, yet obesity restriction was achieved at the cost of compromised physical endurance. Thus, sarcolemmal K(ATP) channels govern muscle energy economy, and their downregulation in a tissue-specific manner could present an antiobesity strategy by rendering muscle increasingly thermogenic at rest and less fuel efficient during exercise.


Subject(s)
Body Weight , Energy Metabolism/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Sarcolemma/metabolism , Animals , Dietary Fats , Eating , Mice , Mice, Knockout , Phenotype , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/genetics
17.
Circ Res ; 105(7): 648-56, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19696409

ABSTRACT

RATIONALE: Induced pluripotent stem cells (iPS) allow derivation of pluripotent progenitors from somatic sources. Originally, iPS were induced by a stemness-related gene set that included the c-MYC oncogene. OBJECTIVE: Here, we determined from embryo to adult the cardiogenic proficiency of iPS programmed without c-MYC, a cardiogenicity-associated transcription factor. METHODS AND RESULTS: Transgenic expression of 3 human stemness factors SOX2, OCT4, and KLF4 here reset murine fibroblasts to the pluripotent ground state. Transduction without c-MYC reversed cellular ultrastructure into a primitive archetype and induced stem cell markers generating 3-germ layers, all qualifiers of acquired pluripotency. Three-factor induced iPS (3F-iPS) clones reproducibly demonstrated cardiac differentiation properties characterized by vigorous beating activity of embryoid bodies and robust expression of cardiac Mef2c, alpha-actinin, connexin43, MLC2a, and troponin I. In vitro isolated iPS-derived cardiomyocytes demonstrated functional excitation-contraction coupling. Chimerism with 3F-iPS derived by morula-stage diploid aggregation was sustained during prenatal heart organogenesis and contributed in vivo to normal cardiac structure and overall performance in adult tumor-free offspring. CONCLUSIONS: Thus, 3F-iPS bioengineered without c-MYC achieve highest stringency criteria for bona fide cardiogenesis enabling reprogrammed fibroblasts to yield de novo heart tissue compatible with native counterpart throughout embryological development and into adulthood.


Subject(s)
Cell Transdifferentiation , Fibroblasts/metabolism , Kruppel-Like Transcription Factors/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Actinin/metabolism , Action Potentials , Animals , Calcium Signaling , Cell Lineage , Cell Transdifferentiation/genetics , Cells, Cultured , Chimerism , Connexin 43/metabolism , Embryo Culture Techniques , Female , Fibroblasts/ultrastructure , Gene Expression Regulation, Developmental , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , MEF2 Transcription Factors , Mice , Mice, Inbred C57BL , Mice, Nude , Myocardial Contraction/genetics , Myocytes, Cardiac/ultrastructure , Myogenic Regulatory Factors/metabolism , Myosin Light Chains/metabolism , Octamer Transcription Factor-3/genetics , Organogenesis , Pluripotent Stem Cells/ultrastructure , Pregnancy , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , Tissue Engineering/methods , Transduction, Genetic , Troponin I/metabolism
18.
J Gen Physiol ; 131(3): 185-96, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18299394

ABSTRACT

Allosteric regulation of heteromultimeric ATP-sensitive potassium (K(ATP)) channels is unique among protein systems as it implies transmission of ligand-induced structural adaptation at the regulatory SUR subunit, a member of ATP-binding cassette ABCC family, to the distinct pore-forming K+ (Kir6.x) channel module. Cooperative interaction between nucleotide binding domains (NBDs) of SUR is a prerequisite for K(ATP) channel gating, yet pathways of allosteric intersubunit communication remain uncertain. Here, we analyzed the role of the ED domain, a stretch of 15 negatively charged aspartate/glutamate amino acid residues (948-962) of the SUR2A isoform, in the regulation of cardiac K(ATP) channels. Disruption of the ED domain impeded cooperative NBDs interaction and interrupted the regulation of K(ATP) channel complexes by MgADP, potassium channel openers, and sulfonylurea drugs. Thus, the ED domain is a structural component of the allosteric pathway within the K(ATP) channel complex integrating transduction of diverse nucleotide-dependent states in the regulatory SUR subunit to the open/closed states of the K+-conducting channel pore.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Receptors, Drug/chemistry , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/genetics , Allosteric Regulation/drug effects , Cell Line , Humans , Ion Channel Gating/drug effects , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/genetics , Protein Structure, Tertiary , Receptors, Drug/genetics , Sulfonylurea Compounds/pharmacology , Sulfonylurea Receptors
19.
J Exp Med ; 204(2): 405-20, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17283208

ABSTRACT

Embryonic stem cells have the distinct potential for tissue regeneration, including cardiac repair. Their propensity for multilineage differentiation carries, however, the liability of neoplastic growth, impeding therapeutic application. Here, the tumorigenic threat associated with embryonic stem cell transplantation was suppressed by cardiac-restricted transgenic expression of the reprogramming cytokine TNF-alpha, enhancing the cardiogenic competence of recipient heart. The in vivo aptitude of TNF-alpha to promote cardiac differentiation was recapitulated in embryoid bodies in vitro. The procardiogenic action required an intact endoderm and was mediated by secreted cardio-inductive signals. Resolved TNF-alpha-induced endoderm-derived factors, combined in a cocktail, secured guided differentiation of embryonic stem cells in monolayers produce cardiac progenitors termed cardiopoietic cells. Characterized by a down-regulation of oncogenic markers, up-regulation, and nuclear translocation of cardiac transcription factors, this predetermined population yielded functional cardiomyocyte progeny. Recruited cardiopoietic cells delivered in infarcted hearts generated cardiomyocytes that proliferated into scar tissue, integrating with host myocardium for tumor-free repair. Thus, cardiopoietic programming establishes a strategy to hone stem cell pluripotency, offering a tumor-resistant approach for regeneration.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Heart/physiology , Myocytes, Cardiac/cytology , Regeneration/physiology , Stem Cell Transplantation/methods , Tumor Necrosis Factor-alpha/metabolism , Animals , DNA Primers , Embryonic Stem Cells/transplantation , Gene Expression Regulation , Mice , Mice, Transgenic , Microarray Analysis , Neoplasms/prevention & control , Transcription Factors/metabolism
20.
Nat Clin Pract Cardiovasc Med ; 4(2): 110-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17245405

ABSTRACT

BACKGROUND: A 53-year-old female presented with a 10-year history of paroxysmal atrial fibrillation (AF), precipitated by activity and refractory to medical therapy. In the absence of traditional risk factors for disease, a genetic defect in electrical homeostasis underlying stress-induced AF was explored. INVESTIGATIONS: Echocardiography, cardiac perfusion stress imaging, invasive electrophysiology with isoproterenol provocation, genomic DNA sequencing of K(ATP) channel genes, exclusion of mutation in 2,000 individuals free of AF, reconstitution of channel defect with molecular phenotyping, and verification of pathogenic link in targeted knockout. DIAGNOSIS: K(ATP) channelopathy caused by missense mutation (Thr1547Ile) of the ABCC9 gene conferring predisposition to adrenergic AF originating from the vein of Marshall. MANAGEMENT: Disruption of arrhythmogenic gene-environment substrate at the vein of Marshall by radiofrequency ablation.


Subject(s)
Atrial Fibrillation/genetics , Atrial Fibrillation/surgery , Catheter Ablation/methods , Channelopathies/genetics , Kv1.5 Potassium Channel/genetics , ATP-Binding Cassette Transporters/genetics , Atrial Fibrillation/diagnostic imaging , Channelopathies/therapy , Chronic Disease , Coronary Vessels/physiopathology , Echocardiography, Doppler , Female , Humans , Middle Aged , Mutation, Missense , Risk Assessment , Tachycardia, Paroxysmal/diagnostic imaging , Tachycardia, Paroxysmal/genetics , Tachycardia, Paroxysmal/surgery , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...