Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Brain Res ; 1750: 147154, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33068634

ABSTRACT

Activation of Ca2+/calmodulin kinase II (CaMKII) and the N-Methyl D-aspartate receptor (NMDAR), particularly its GluN2B subunit, contribute to the central sensitization of nociceptive pathways and persistent pain. Using mutant mice wherein the activity-driven binding of CaMKII to S1303 in GluN2B is abrogated (GluN2BKI), this study investigated the importance of this interaction for acute and persistent inflammatory nociception. GluN2BKI, wild type and heterozygote mice did not differ in responses to acute noxious heat stimuli as measured with tail flick, paw flick, or hot plate assays, nor did they differ in their responses to mechanical stimulation with von Frey filaments. Surprisingly, the three genotypes exhibited similar spontaneous pain behaviors and hypersensitivity to heat or mechanical stimuli induced by intraplantar injection of capsaicin; however, GluN2BKI mice did not immediately attend to the paw. WT and GluN2BKI mice also did not differ in the nociceptive behaviors elicited by intraplantar injection of formalin, even though MK801 greatly reduced these behaviors in both genotypes concordant with NMDAR dependence. CaMKII binding to GluN2B at S1303 therefore does not appear to be critical for the development of inflammatory nociception. Finally, intrathecal KN93 reduced formalin-induced nociceptive behaviors in GluN2BKI mice. KN93 does not inhibit CaKMII, but rather binds Ca2+/calmodulin. It has multiple other targets including Ca2+-, Na+- and K+-channels, as well as various kinases. Therefore, the use of GluN2BKI mice provided genetic specificity in assessing the role of CaMKII in inflammatory pain signaling cascades. These results challenge current thinking on the involvement of the CaMKII-NMDAR interaction in inflammatory pain.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Inflammation/metabolism , Pain/metabolism , Animals , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calmodulin/metabolism , Capsaicin/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Nociception/physiology , Pain/physiopathology , Protein Binding , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology
2.
Nat Commun ; 11(1): 4416, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887881

ABSTRACT

Despite the clear association between myocardial injury, heart failure and depressed myocardial energetics, little is known about upstream signals responsible for remodeling myocardial metabolism after pathological stress. Here, we report increased mitochondrial calmodulin kinase II (CaMKII) activation and left ventricular dilation in mice one week after myocardial infarction (MI) surgery. By contrast, mice with genetic mitochondrial CaMKII inhibition are protected from left ventricular dilation and dysfunction after MI. Mice with myocardial and mitochondrial CaMKII overexpression (mtCaMKII) have severe dilated cardiomyopathy and decreased ATP that causes elevated cytoplasmic resting (diastolic) Ca2+ concentration and reduced mechanical performance. We map a metabolic pathway that rescues disease phenotypes in mtCaMKII mice, providing insights into physiological and pathological metabolic consequences of CaMKII signaling in mitochondria. Our findings suggest myocardial dilation, a disease phenotype lacking specific therapies, can be prevented by targeted replacement of mitochondrial creatine kinase or mitochondrial-targeted CaMKII inhibition.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomyopathy, Dilated/metabolism , Myocardial Infarction/physiopathology , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Energy Metabolism/genetics , Energy Metabolism/physiology , Heart Failure/metabolism , Heart Ventricles/physiopathology , Mice , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardial Infarction/surgery , Signal Transduction
3.
Int J Pharm ; 520(1-2): 275-283, 2017 Mar 30.
Article in English | MEDLINE | ID: mdl-28167264

ABSTRACT

An excess of calcium (Ca2+) influx into mitochondria during mitochondrial re-energization is one of the causes of myocardial cell death during ischemic/reperfusion injury. This overload of Ca2+ triggers the mitochondrial permeability transition pore (mPTP) opening which leads to programmed cell death. During the ischemic/reperfusion stage, the activated Ca2+/calmodulin-dependent protein kinase II (CaMKII) enzyme is responsible for Ca2+ influx. To reduce CaMKII-related cell death, sub-micron particles composed of poly(lactic-co-glycolic acid) (PLGA), loaded with a CaMKII inhibitor peptide were fabricated. The CaMKII inhibitor peptide-loaded (CIP) particles were coated with a mitochondria targeting moiety, triphenylphosphonium cation (TPP), which allowed the particles to accumulate and release the peptide inside mitochondria to inhibit CaMKII activity. The fluorescently labeled TPP-CIP was taken up by mitochondria and successfully reduced reactive oxygen species (ROS) caused by Isoprenaline (ISO) in a differentiated rat cardiomyocyte-like cell line. When cells were treated with TPP-CIP prior to ISO exposure, they maintained mitochondrial membrane potential. The TPP-CIP protected cells from ISO-induced ROS production and decreased mitochondrial membrane potential. Thus, TPP-CIP has the potential to be used in protection against ischemia/reperfusion injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Organophosphorus Compounds/chemistry , Peptides/pharmacology , Protective Agents/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Isoproterenol/pharmacology , Lactic Acid/chemistry , Membrane Potential, Mitochondrial/drug effects , Myocytes, Cardiac/cytology , Peptides/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Protective Agents/chemistry , Rats , Reactive Oxygen Species/metabolism , Surface Properties
4.
Proc Natl Acad Sci U S A ; 112(29): 9129-34, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26153425

ABSTRACT

Myocardial mitochondrial Ca(2+) entry enables physiological stress responses but in excess promotes injury and death. However, tissue-specific in vivo systems for testing the role of mitochondrial Ca(2+) are lacking. We developed a mouse model with myocardial delimited transgenic expression of a dominant negative (DN) form of the mitochondrial Ca(2+) uniporter (MCU). DN-MCU mice lack MCU-mediated mitochondrial Ca(2+) entry in myocardium, but, surprisingly, isolated perfused hearts exhibited higher O2 consumption rates (OCR) and impaired pacing induced mechanical performance compared with wild-type (WT) littermate controls. In contrast, OCR in DN-MCU-permeabilized myocardial fibers or isolated mitochondria in low Ca(2+) were not increased compared with WT, suggesting that DN-MCU expression increased OCR by enhanced energetic demands related to extramitochondrial Ca(2+) homeostasis. Consistent with this, we found that DN-MCU ventricular cardiomyocytes exhibited elevated cytoplasmic [Ca(2+)] that was partially reversed by ATP dialysis, suggesting that metabolic defects arising from loss of MCU function impaired physiological intracellular Ca(2+) homeostasis. Mitochondrial Ca(2+) overload is thought to dissipate the inner mitochondrial membrane potential (ΔΨm) and enhance formation of reactive oxygen species (ROS) as a consequence of ischemia-reperfusion injury. Our data show that DN-MCU hearts had preserved ΔΨm and reduced ROS during ischemia reperfusion but were not protected from myocardial death compared with WT. Taken together, our findings show that chronic myocardial MCU inhibition leads to previously unanticipated compensatory changes that affect cytoplasmic Ca(2+) homeostasis, reprogram transcription, increase OCR, reduce performance, and prevent anticipated therapeutic responses to ischemia-reperfusion injury.


Subject(s)
Adaptation, Physiological , Calcium Channels/metabolism , Heart/physiopathology , Mitochondria, Heart/metabolism , Stress, Physiological , Animals , Blood Pressure , Calcium/metabolism , Cardiac Pacing, Artificial , Cellular Reprogramming , Cytosol/drug effects , Cytosol/metabolism , Diastole , Electrocardiography , Genes, Dominant , Glucose/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Mice , Mitochondria, Heart/drug effects , Myocardial Reperfusion , Myocardium/metabolism , Myocardium/pathology , Oxygen Consumption , Prostaglandin-Endoperoxide Synthases/metabolism , Sarcoplasmic Reticulum/metabolism , Transcription, Genetic
6.
Curr Mol Pharmacol ; 8(2): 143-8, 2015.
Article in English | MEDLINE | ID: mdl-25966695

ABSTRACT

Cav1 channels mediate L-type Ca(2+) currents that trigger the exocytotic release of glutamate from the specialized "ribbon" synapse of retinal photoreceptors (PRs) and cochlear inner hair cells (IHCs). Genetic evidence from animal models and humans support a role for Cav1.3 and Cav1.4 as the primary Cav channels in IHCs and PRs, respectively. Because of the unique features of transmission at ribbon synapses, Cav1.3 and Cav1.4 exhibit unusual properties that are well-suited for their physiological roles. These properties may be intrinsic to the channel subunit(s) and/or may be conferred by regulatory interactions with synaptic signaling molecules. This review will cover advances in our understanding of the function of Cav1 channels at sensory ribbon synapses, and how dysregulation of these channels leads to disorders of vision and hearing.


Subject(s)
Calcium Channels, L-Type/physiology , Hearing/physiology , Vision, Ocular/physiology , Animals , Calcium Channels, L-Type/genetics , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/physiology , Hearing/genetics , Humans , Models, Biological , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/physiology , Synapses/genetics , Synapses/physiology , Vision, Ocular/genetics
7.
Nat Commun ; 6: 6081, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25603276

ABSTRACT

Heart rate increases are a fundamental adaptation to physiological stress, while inappropriate heart rate increases are resistant to current therapies. However, the metabolic mechanisms driving heart rate acceleration in cardiac pacemaker cells remain incompletely understood. The mitochondrial calcium uniporter (MCU) facilitates calcium entry into the mitochondrial matrix to stimulate metabolism. We developed mice with myocardial MCU inhibition by transgenic expression of a dominant-negative (DN) MCU. Here, we show that DN-MCU mice had normal resting heart rates but were incapable of physiological fight or flight heart rate acceleration. We found that MCU function was essential for rapidly increasing mitochondrial calcium in pacemaker cells and that MCU-enhanced oxidative phoshorylation was required to accelerate reloading of an intracellular calcium compartment before each heartbeat. Our findings show that MCU is necessary for complete physiological heart rate acceleration and suggest that MCU inhibition could reduce inappropriate heart rate increases without affecting resting heart rate.


Subject(s)
Calcium Channels/physiology , Heart Rate/physiology , Mitochondria/metabolism , Action Potentials , Adenosine Triphosphate/chemistry , Animals , Biological Clocks , Caffeine/chemistry , Calcium/chemistry , Calcium/metabolism , Echocardiography/methods , Electrocardiography/methods , Female , Genes, Dominant , Green Fluorescent Proteins/chemistry , Heart/physiology , In Vitro Techniques , Isoproterenol/chemistry , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Myocytes, Cardiac/cytology , NAD/chemistry , Perfusion , Phosphorylation , Transgenes
8.
Nanomedicine (Lond) ; 9(16): 2531-43, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25490424

ABSTRACT

Mitochondria are a promising therapeutic target for the detection, prevention and treatment of various human diseases such as cancer, neurodegenerative diseases, ischemia-reperfusion injury, diabetes and obesity. To reach mitochondria, therapeutic molecules need to not only gain access to specific organs, but also to overcome multiple barriers such as the cell membrane and the outer and inner mitochondrial membranes. Cellular and mitochondrial barriers can be potentially overcome through the design of mitochondriotropic particulate carriers capable of transporting drug molecules selectively to mitochondria. These particulate carriers or vectors can be made from lipids (liposomes), biodegradable polymers, or metals, protecting the drug cargo from rapid elimination and degradation in vivo. Many formulations can be tailored to target mitochondria by the incorporation of mitochondriotropic agents onto the surface and can be manufactured to desired sizes and molecular charge. Here, we summarize recently reported strategies for delivering therapeutic molecules to mitochondria using various particle-based formulations.


Subject(s)
Drug Carriers/administration & dosage , Drug Delivery Systems , Mitochondria/drug effects , Nanoparticles/administration & dosage , Humans , Liposomes/administration & dosage , Mitochondria/pathology
10.
Front Pharmacol ; 5: 67, 2014.
Article in English | MEDLINE | ID: mdl-24822046

ABSTRACT

CaMKII is a newly discovered resident of mitochondria in the heart. Mitochondrial CaMKII promotes poor outcomes after heart injury from a number of pathological conditions, including myocardial infarction (MI), ischemia reperfusion (IR), and stress from catecholamine stimulation. A study using the inhibitor of CaMKII, CaMKIIN, with expression delimited to myocardial mitochondria, indicates that an underlying cause of heart disease results from the opening of the mitochondrial permeability transition pore (mPTP). Evidence from electrophysiological and other experiments show that CaMKII inhibition likely suppresses mPTP opening by reducing Ca(2+) entry into mitochondria. However, we expect other proteins involved in Ca(2+) signaling in the mitochondria are affected with CaMKII inhibition. Several outstanding questions remain for CaMKII signaling in heart mitochondria. Most importantly, how does CaMKII, without the recognized N-terminal mitochondrial targeting sequence transfer to mitochondria?

12.
Hypertension ; 62(2): 434-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23753415

ABSTRACT

The multifunctional Ca(2+)/calmodulin-dependent kinase II (CaMKII) is activated by vasoconstrictors in vascular smooth muscle cells (VSMC), but its impact on vasoconstriction remains unknown. We hypothesized that CaMKII inhibition in VSMC decreases vasoconstriction. Using novel transgenic mice that express the inhibitor peptide CaMKIIN in smooth muscle (TG SM-CaMKIIN), we investigated the effect of CaMKII inhibition on L-type Ca(2+) channel current (ICa), cytoplasmic and sarcoplasmic reticulum Ca(2+), and vasoconstriction in mesenteric arteries. In mesenteric VSMC, CaMKII inhibition significantly reduced action potential duration and the residual ICa 50 ms after peak amplitude, indicative of loss of L-type Ca(2+) channel-dependent ICa facilitation. Treatment with angiotensin II or phenylephrine increased the intracellular Ca(2+) concentration in wild-type but not TG SM-CaMKIIN VSMC. The difference in intracellular Ca(2+) concentration was abolished by pretreatment with nifedipine, an L-type Ca(2+) channel antagonist. In TG SM-CaMKIIN VSMC, the total sarcoplasmic reticulum Ca(2+) content was reduced as a result of diminished sarcoplasmic reticulum Ca(2+) ATPase activity via impaired derepression of the sarcoplasmic reticulum Ca(2+) ATPase inhibitor phospholamban. Despite the differences in intracellular Ca(2+) concentration, CaMKII inhibition did not alter myogenic tone or vasoconstriction of mesenteric arteries in response to KCl, angiotensin II, and phenylephrine. However, it increased myosin light chain kinase activity. These data suggest that CaMKII activity maintains intracellular calcium homeostasis but is not required for vasoconstriction of mesenteric arteries.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calcium/metabolism , Homeostasis , Angiotensin II/pharmacology , Animals , Benzylamines/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Kinase/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sulfonamides/pharmacology
13.
Circ Res ; 112(2): 309-17, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23192947

ABSTRACT

RATIONALE: The sodium-calcium exchanger 1 (NCX1) is predominantly expressed in the heart and is implicated in controlling automaticity in isolated sinoatrial node (SAN) pacemaker cells, but the potential role of NCX1 in determining heart rate in vivo is unknown. OBJECTIVE: To determine the role of Ncx1 in heart rate. METHODS AND RESULTS: We used global myocardial and SAN-targeted conditional Ncx1 knockout (Ncx1(-/-)) mice to measure the effect of the NCX current on pacemaking activity in vivo, ex vivo, and in isolated SAN cells. We induced conditional Ncx1(-/-) using a Cre/loxP system. Unexpectedly, in vivo and ex vivo hearts and isolated SAN cells showed that basal rates in Ncx1(-/-) (retaining ≈20% of control level NCX current) and control mice were similar, suggesting that physiological NCX1 expression is not required for determining resting heart rate. However, increases in heart rate and SAN cell automaticity in response to isoproterenol or the dihydropyridine Ca(2+) channel agonist BayK8644 were significantly blunted or eliminated in Ncx1(-/-) mice, indicating that NCX1 is important for fight or flight heart rate responses. In contrast, the pacemaker current and L-type Ca(2+) currents were equivalent in control and Ncx1(-/-) SAN cells under resting and isoproterenol-stimulated conditions. Ivabradine, a pacemaker current antagonist with clinical efficacy, reduced basal SAN cell automaticity similarly in control and Ncx1(-/-) mice. However, ivabradine decreased automaticity in SAN cells isolated from Ncx1(-/-) mice more effectively than in control SAN cells after isoproterenol, suggesting that the importance of NCX current in fight or flight rate increases is enhanced after pacemaker current inhibition. CONCLUSIONS: Physiological Ncx1 expression is required for increasing sinus rates in vivo, ex vivo, and in isolated SAN cells, but not for maintaining resting heart rate.


Subject(s)
Heart Rate/physiology , Rest/physiology , Sinoatrial Node/physiology , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Heart Rate/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sodium-Calcium Exchanger/metabolism , Sodium-Calcium Exchanger/physiology
14.
Nature ; 491(7423): 269-73, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23051746

ABSTRACT

Myocardial cell death is initiated by excessive mitochondrial Ca(2+) entry causing Ca(2+) overload, mitochondrial permeability transition pore (mPTP) opening and dissipation of the mitochondrial inner membrane potential (ΔΨm). However, the signalling pathways that control mitochondrial Ca(2+) entry through the inner membrane mitochondrial Ca(2+) uniporter (MCU) are not known. The multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is activated in ischaemia reperfusion, myocardial infarction and neurohumoral injury, common causes of myocardial death and heart failure; these findings suggest that CaMKII could couple disease stress to mitochondrial injury. Here we show that CaMKII promotes mPTP opening and myocardial death by increasing MCU current (I(MCU)). Mitochondrial-targeted CaMKII inhibitory protein or cyclosporin A, an mPTP antagonist with clinical efficacy in ischaemia reperfusion injury, equivalently prevent mPTP opening, ΔΨm deterioration and diminish mitochondrial disruption and programmed cell death in response to ischaemia reperfusion injury. Mice with myocardial and mitochondrial-targeted CaMKII inhibition have reduced I(MCU) and are resistant to ischaemia reperfusion injury, myocardial infarction and neurohumoral injury, suggesting that pathological actions of CaMKII are substantially mediated by increasing I(MCU). Our findings identify CaMKII activity as a central mechanism for mitochondrial Ca(2+) entry in myocardial cell death, and indicate that mitochondrial-targeted CaMKII inhibition could prevent or reduce myocardial death and heart failure in response to common experimental forms of pathophysiological stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/enzymology , Myocardium/pathology , Stress, Physiological , Animals , Apoptosis/drug effects , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Cyclosporine/pharmacology , Female , Heart/drug effects , Heart/physiopathology , Heart Failure/drug therapy , Heart Failure/prevention & control , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Infarction/drug therapy , Myocardial Infarction/prevention & control , Myocardium/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Serine/metabolism , Stress, Physiological/drug effects
15.
Nat Med ; 17(12): 1610-8, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081025

ABSTRACT

Excessive activation of the ß-adrenergic, angiotensin II (Ang II) and aldosterone signaling pathways promotes mortality after myocardial infarction, and antagonists targeting these pathways are core therapies for treating this condition. Catecholamines and Ang II activate the multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), the inhibition of which prevents isoproterenol-mediated and Ang II-mediated cardiomyopathy. Here we show that aldosterone exerts direct toxic actions on myocardium by oxidative activation of CaMKII, causing cardiac rupture and increased mortality in mice after myocardial infarction. Aldosterone induces CaMKII oxidation by recruiting NADPH oxidase, and this oxidized and activated CaMKII promotes matrix metalloproteinase 9 (MMP9) expression in cardiomyocytes. Myocardial CaMKII inhibition, overexpression of methionine sulfoxide reductase A (an enzyme that reduces oxidized CaMKII) or NADPH oxidase deficiency prevented aldosterone-enhanced cardiac rupture after myocardial infarction. These findings show that oxidized myocardial CaMKII mediates the cardiotoxic effects of aldosterone on the cardiac matrix and establish CaMKII as a nodal signal for the neurohumoral pathways associated with poor outcomes after myocardial infarction.


Subject(s)
Aldosterone/adverse effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotoxins/adverse effects , Myocardial Infarction/pathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Heart/drug effects , Humans , Luciferases/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Methionine Sulfoxide Reductases/genetics , Methionine Sulfoxide Reductases/metabolism , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Oxidation-Reduction , Signal Transduction , Up-Regulation
16.
J Clin Invest ; 121(8): 3277-88, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21785215

ABSTRACT

Sinus node dysfunction (SND) is a major public health problem that is associated with sudden cardiac death and requires surgical implantation of artificial pacemakers. However, little is known about the molecular and cellular mechanisms that cause SND. Most SND occurs in the setting of heart failure and hypertension, conditions that are marked by elevated circulating angiotensin II (Ang II) and increased oxidant stress. Here, we show that oxidized calmodulin kinase II (ox-CaMKII) is a biomarker for SND in patients and dogs and a disease determinant in mice. In wild-type mice, Ang II infusion caused sinoatrial nodal (SAN) cell oxidation by activating NADPH oxidase, leading to increased ox-CaMKII, SAN cell apoptosis, and SND. p47-/- mice lacking functional NADPH oxidase and mice with myocardial or SAN-targeted CaMKII inhibition were highly resistant to SAN apoptosis and SND, suggesting that ox-CaMKII-triggered SAN cell death contributed to SND. We developed a computational model of the sinoatrial node that showed that a loss of SAN cells below a critical threshold caused SND by preventing normal impulse formation and propagation. These data provide novel molecular and mechanistic information to understand SND and suggest that targeted CaMKII inhibition may be useful for preventing SND in high-risk patients.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Oxygen/chemistry , Sick Sinus Syndrome/genetics , Sinoatrial Node/pathology , Angiotensin II/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Dogs , Electrocardiography/methods , Humans , Mice , Mice, Transgenic , NADPH Oxidases/genetics , Reactive Oxygen Species , Sick Sinus Syndrome/metabolism
17.
Circ Arrhythm Electrophysiol ; 4(3): 379-87, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21406683

ABSTRACT

BACKGROUND: Catecholamines increase heart rate by augmenting the cAMP-responsive hyperpolarization-activated cyclic nucleotide-gated channel 4 pacemaker current (I(f)) and by promoting inward Na(+)/Ca(2+) exchanger current (I(NCX)) by a "Ca(2+) clock" mechanism in sinoatrial nodal cells (SANCs). The importance, identity, and function of signals that connect I(f) and Ca(2+) clock mechanisms are uncertain and controversial, but the multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is required for physiological heart rate responses to ß-adrenergic receptor (ß-AR) stimulation. The aim of this study was to measure the contribution of the Ca(2+) clock and CaMKII to cardiac pacing independent of ß-AR agonist stimulation. METHODS AND RESULTS: We used the L-type Ca(2+) channel agonist Bay K8644 (BayK) to activate the SANC Ca(2+) clock. BayK and isoproterenol were similarly effective in increasing rates in SANCs and Langendorff-perfused hearts from wild-type control mice. In contrast, SANCs and isolated hearts from mice with CaMKII inhibition by transgenic expression of an inhibitory peptide (AC3-I) were resistant to rate increases by BayK. BayK only activated CaMKII in control SANCs but increased L-type Ca(2+) current (I(Ca)) equally in all SANCs, indicating that increasing I(Ca) was insufficient and suggesting that CaMKII activation was required for heart rate increases by BayK. BayK did not increase I(f) or protein kinase A-dependent phosphorylation of phospholamban (at Ser16), indicating that increased SANC Ca(2+) by BayK did not augment cAMP/protein kinase A signaling at these targets. Late-diastolic intracellular Ca(2+) release and I(NCX) were significantly reduced in AC3-I SANCs, and the response to BayK was eliminated by ryanodine in all groups. CONCLUSIONS: The Ca(2+) clock is capable of supporting physiological fight-or-flight responses, independent of ß-AR stimulation or I(f) increases. Complete Ca(2+) clock and ß-AR stimulation responses require CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/pharmacology , Calcium/pharmacology , Catecholamines/pharmacology , Heart Rate/drug effects , Sinoatrial Node/drug effects , Tachycardia/drug therapy , Animals , Disease Models, Animal , Dogs , Heart Rate/physiology , Mice , Microscopy, Confocal , Myocardium/metabolism , Myocardium/pathology , Sinoatrial Node/metabolism , Tachycardia/metabolism
18.
J Mol Cell Cardiol ; 49(1): 33-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20380837

ABSTRACT

Increasing evidence suggests that cardiac pacemaking is the result of two sinoatrial node (SAN) cell mechanisms: a 'voltage clock' and a Ca(2+) dependent process, or 'Ca(2+) clock.' The voltage clock initiates action potentials (APs) by SAN cell membrane potential depolarization from inward currents, of which the pacemaker current (I(f)) is thought to be particularly important. A Ca(2+) dependent process triggers APs when sarcoplasmic reticulum (SR) Ca(2+) release activates inward current carried by the forward mode of the electrogenic Na(+)/Ca(2+) exchanger (NCX). However, these mechanisms have mostly been defined in rodents or rabbits, but are unexplored in single SAN cells from larger animals. Here, we used patch-clamp and confocal microscope techniques to explore the roles of the voltage and Ca(2+) clock mechanisms in canine SAN pacemaker cells. We found that ZD7288, a selective I(f) antagonist, significantly reduced basal automaticity and induced irregular, arrhythmia-like activity in canine SAN cells. In addition, ZD7288 impaired but did not eliminate the SAN cell rate acceleration by isoproterenol. In contrast, ryanodine significantly reduced the SAN cell acceleration by isoproterenol, while ryanodine reduction of basal automaticity was modest ( approximately 14%) and did not reach statistical significance. Importantly, pretreatment with ryanodine eliminated SR Ca(2+) release, but did not affect basal or isoproterenol-enhanced I(f). Taken together, these results indicate that voltage and Ca(2+) dependent automaticity mechanisms coexist in canine SAN cells, and suggest that I(f) and SR Ca(2+) release cooperate to determine baseline and catecholamine-dependent automaticity in isolated dog SAN cells.


Subject(s)
Calcium/metabolism , Calcium/physiology , Sinoatrial Node , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dogs , Female , Heart , Isoproterenol/metabolism , Isoproterenol/pharmacology , Male , Myocytes, Cardiac/metabolism , Pacemaker, Artificial , Ryanodine/metabolism , Ryanodine/pharmacology , Sarcoplasmic Reticulum/metabolism , Sinoatrial Node/cytology , Sinoatrial Node/metabolism , Sinoatrial Node/physiology
19.
EMBO J ; 29(2): 482-95, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-19942860

ABSTRACT

Central noradrenergic signalling mediates arousal and facilitates learning through unknown molecular mechanisms. Here, we show that the beta(2)-adrenergic receptor (beta(2)AR), the trimeric G(s) protein, adenylyl cyclase, and PKA form a signalling complex with the AMPA-type glutamate receptor subunit GluR1, which is linked to the beta(2)AR through stargazin and PSD-95 and their homologues. Only GluR1 associated with the beta(2)AR is phosphorylated by PKA on beta(2)AR stimulation. Peptides that interfere with the beta(2)AR-GluR1 association prevent this phosphorylation of GluR1. This phosphorylation increases GluR1 surface expression at postsynaptic sites and amplitudes of EPSCs and mEPSCs in prefrontal cortex slices. Assembly of all proteins involved in the classic beta(2)AR-cAMP cascade into a supramolecular signalling complex and thus allows highly localized and selective regulation of one of its major target proteins.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, AMPA/analysis , Receptors, AMPA/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adenylyl Cyclases/analysis , Animals , Calcium Channels/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Disks Large Homolog 4 Protein , Electrophysiology , GTP-Binding Protein alpha Subunits, Gs/analysis , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation , Hippocampus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurons/cytology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, Adrenergic, beta-2/analysis
20.
Fly (Austin) ; 2(6): 269-79, 2008.
Article in English | MEDLINE | ID: mdl-19077536

ABSTRACT

elfless (CG15150, FBgn0032660) maps to polytene region 36DE 5' (left) of reduced ocelli/Pray for Elves (PFE) on chromosome 2L and is predicted to encode a 187 amino acid RING finger E3 ubiquitin ligase that is putatively involved in programmed cell death (PCD, e.g., apoptosis). Several experimental approaches were used to characterize CG15150/elfless and test whether defects in this gene underlie the male sterile phenotype associated with overlapping chromosomal deficiencies of region 36DE. elfless expression is greatly enhanced in the testes and the expression pattern of UAS-elfless-EGFP driven by elfless-Gal4 is restricted to the tail cyst cell nuclei of the testes. Despite this, elfless transgenes failed to rescue the male sterile phenotype in Df/Df flies. Furthermore, null alleles of elfless, generated either by imprecise excision of an upstream P-element or by FLP-FRT deletion between two flanking piggyBac elements, are fertile. In a gain-of-function setting in the eye, we found that elfless genetically interacts with key members of the apoptotic pathway including the initiator caspase Dronc and the ubiquitin conjugating enzyme UbcD1. DIAP1, but not UbcD1, protein levels are increased in heads of flies expressing Elfless-EGFP in the eye, and in testes of flies expressing elfless-Gal4 driven Elfless-EGFP. Based on these findings, we speculate that Elfless may regulate tail cyst cell degradation to provide an advantageous, though not essential, function in the testis.


Subject(s)
Apoptosis , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation , Male , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spermatogenesis/physiology , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...