Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin Cancer Res ; 29(23): 4870-4882, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37733811

ABSTRACT

PURPOSE: Tumors activate protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK, also called EIF2AK3) in response to hypoxia and nutrient deprivation as a stress-mitigation strategy. Here, we tested the hypothesis that inhibiting PERK with HC-5404 enhances the antitumor efficacy of standard-of-care VEGF receptor tyrosine kinase inhibitors (VEGFR-TKI). EXPERIMENTAL DESIGN: HC-5404 was characterized as a potent and selective PERK inhibitor, with favorable in vivo properties. Multiple renal cell carcinoma (RCC) tumor models were then cotreated with both HC-5404 and VEGFR-TKI in vivo, measuring tumor volume across time and evaluating tumor response by protein analysis and IHC. RESULTS: VEGFR-TKI including axitinib, cabozantinib, lenvatinib, and sunitinib induce PERK activation in 786-O RCC xenografts. Cotreatment with HC-5404 inhibited PERK in tumors and significantly increased antitumor effects of VEGFR-TKI across multiple RCC models, resulting in tumor stasis or regression. Analysis of tumor sections revealed that HC-5404 enhanced the antiangiogenic effects of axitinib and lenvatinib by inhibiting both new vasculature and mature tumor blood vessels. Xenografts that progress on axitinib monotherapy remain sensitive to the combination treatment, resulting in ∼20% tumor regression in the combination group. When tested across a panel of 18 RCC patient-derived xenograft (PDX) models, the combination induced greater antitumor effects relative to monotherapies. In this single animal study, nine out of 18 models responded with ≥50% tumor regression from baseline in the combination group. CONCLUSIONS: By disrupting an adaptive stress response evoked by VEGFR-TKI, HC-5404 presents a clinical opportunity to improve the antitumor effects of well-established standard-of-care therapies in RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Humans , Carcinoma, Renal Cell/pathology , Axitinib/pharmacology , Axitinib/therapeutic use , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use
2.
Pharmaceutics ; 14(10)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36297668

ABSTRACT

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) responsible for regulating protein synthesis and alleviating ER stress. PERK has been implicated in tumorigenesis, cancer cell survival as well metabolic diseases such as diabetes. The structure-based design and optimization of a novel mandelamide-derived pyrrolopyrimidine series of PERK inhibitors as described herein, resulted in the identification of compound 26, a potent, selective, and orally bioavailable compound suitable for interrogating PERK pathway biology in vitro and in vivo, with pharmacokinetics suitable for once-a-day oral dosing in mice.

3.
Bioorg Med Chem Lett ; 43: 128058, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33895276

ABSTRACT

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of the three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) that regulates protein synthesis, alleviates cellular ER stress and has been implicated in tumorigenesis and prolonged cancer cell survival. In this study, we report a series of 2-amino-3-amido-5-aryl-pyridines that we have identified as potent, selective, and orally bioavailable PERK inhibitors. Amongst the series studied herein, compound (28) a (R)-2-Amino-5-(4-(2-(3,5-difluorophenyl)-2-hydroxyacetamido)-2-ethylphenyl)-N-isopropylnicotinamide has demonstrated potent biochemical and cellular activity, robust pharmacokinetics and 70% oral bioavailability in mice. Given these data, this compound (28) was studied in the 786-O renal cell carcinoma xenograft model. We observed dose-dependent, statistically significant tumor growth inhibition, supporting the use of this tool compound in additional mechanistic studies.


Subject(s)
Drug Discovery , Pyridines/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Administration, Oral , Biological Availability , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Structure-Activity Relationship , eIF-2 Kinase/metabolism
4.
Curr Mol Pharmacol ; 8(2): 133-42, 2015.
Article in English | MEDLINE | ID: mdl-25981977

ABSTRACT

Voltage gated calcium channels are essential for cardiac physiology by serving as sarcolemma- restricted gatekeepers for calcium in cardiac myocytes. Activation of the L-type voltagegated calcium channel provides the calcium entry required for excitation-contraction coupling and contributes to the plateau phase of the cardiac action potential. Given these critical physiological roles, subtle disturbances in L-type channel function can lead to fatal cardiac arrhythmias. Indeed, numerous human arrhythmia syndromes have been linked to mutations in the L-type channel leading to gain-of-function or loss-of-function mutations. In this review, we discuss the current state of knowledge regarding these mutations present in Timothy Syndrome, Long and Short QT Syndromes, Brugada Syndrome and Early Repolarization Syndrome. We discuss the pathological consequences of the mutations, the biophysical effects of the mutations on the channel as well as possible therapeutic considerations and challenges for future studies.


Subject(s)
Arrhythmias, Cardiac/genetics , Calcium Channels, L-Type/genetics , Genetic Predisposition to Disease/genetics , Mutation , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autistic Disorder/physiopathology , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Brugada Syndrome/physiopathology , Calcium/metabolism , Calcium Channels, L-Type/physiology , Humans , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Long QT Syndrome/physiopathology , Models, Genetic , Syndactyly/genetics , Syndactyly/metabolism , Syndactyly/physiopathology
5.
J Am Coll Cardiol ; 64(1): 66-79, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24998131

ABSTRACT

BACKGROUND: BrS is an inherited sudden cardiac death syndrome. Less than 35% of BrS probands have genetically identified pathogenic variants. Recent evidence has implicated SCN10A, a neuronal sodium channel gene encoding Nav1.8, in the electrical function of the heart. OBJECTIVES: The purpose of this study was to test the hypothesis that SCN10A variants contribute to the development of Brugada syndrome (BrS). METHODS: Clinical analysis and direct sequencing of BrS susceptibility genes were performed for 150 probands and family members as well as >200 healthy controls. Expression and coimmunoprecipitation studies were performed to functionally characterize the putative pathogenic mutations. RESULTS: We identified 17 SCN10A mutations in 25 probands (20 male and 5 female); 23 of the 25 probands (92.0%) displayed overlapping phenotypes. SCN10A mutations were found in 16.7% of BrS probands, approaching our yield for SCN5A mutations (20.1%). Patients with BrS who had SCN10A mutations were more symptomatic and displayed significantly longer PR and QRS intervals compared with SCN10A-negative BrS probands. The majority of mutations localized to the transmembrane-spanning regions. Heterologous coexpression of wild-type (WT) SCN10A with WT-SCN5A in HEK cells caused a near doubling of sodium channel current compared with WT-SCN5A alone. In contrast, coexpression of SCN10A mutants (R14L and R1268Q) with WT-SCN5A caused a 79.4% and 84.4% reduction in sodium channel current, respectively. The coimmunoprecipitation studies provided evidence for the coassociation of Nav1.8 and Nav1.5 in the plasma membrane. CONCLUSIONS: Our study identified SCN10A as a major susceptibility gene for BrS, thus greatly enhancing our ability to genotype and risk stratify probands and family members.


Subject(s)
Brugada Syndrome/diagnosis , Brugada Syndrome/genetics , Genetic Variation/genetics , Mutation, Missense/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
7.
Heart Rhythm ; 10(7): 1054-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23499631

ABSTRACT

BACKGROUND: Wenxin Keli (WK), a Chinese herb extract, is reported to be effective in the treatment of atrial and ventricular cardiac arrhythmias. Recent studies suggest that WK inhibits the transient potassium outward current (I(to)). OBJECTIVE: To examine the effectiveness of WK, alone and in combination with quinidine, to suppress arrhythmogenesis in an experimental model of Brugada syndrome (BrS). METHODS: Action potential and electrocardiographic recordings were obtained from epicardial and endocardial sites of coronary-perfused canine right ventricular wedge preparations. The Ito agonist NS5806 (10-15 µM) was used to pharmacologically mimic a genetic predisposition to BrS. RESULTS: The Ito agonist induced Phase 2 reentry (P2R) in 13/19 preparations and polymorphic ventricular tachycardia (pVT) in 11/19 wedge preparations. WK (10 g/L) suppressed P2R and pVT in 100% (3/3) of preparations. A lower concentration of WK (5 g/L) suppressed P2R in 60% (3/5) and pVT in 50% (2/4), but in combination with a low concentration of quinidine (5 µM), was 100% effective in suppressing P2R and pVT. Quinidine alone suppressed P2R and pVT in 60% (3/5) and 50% (2/4), respectively, and in combination with WK (5 g/L) suppressed P2R and pVT by 80% (4/5) and 75% (3/4), respectively. WK reduced Ito, the L-type calcium current, and contractility in single cardiomyocytes, but dose-dependently increased contractility in intact wedge preparations, an effect mimicked by tyramine. CONCLUSIONS: Our data provide support for the hypothesis that WK, particularly in combination with quinidine, effectively suppresses arrhythmogenesis in an experimental model of BrS via inhibition of Ito and indirect adrenergic sympathomimetic effects.


Subject(s)
Brugada Syndrome/drug therapy , Drugs, Chinese Herbal/pharmacology , Myocytes, Cardiac/drug effects , Quinidine/pharmacology , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Brugada Syndrome/pathology , Brugada Syndrome/physiopathology , Disease Models, Animal , Dogs , Myocytes, Cardiac/pathology , Patch-Clamp Techniques
8.
J Physiol ; 590(24): 6381-7, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23070698

ABSTRACT

Enhancement of contractile force (inotropy) occurs in skeletal muscle following neuroendocrine release of catecholamines and activation of muscle ß-adrenergic receptors. Despite extensive study, the molecular mechanism underlying the inotropic response in skeletal muscle is not well understood. Here we show that phosphorylation of a single serine residue (S2844) in the sarcoplasmic reticulum (SR) Ca(2+) release channel/ryanodine receptor type 1 (RyR1) by protein kinase A (PKA) is critical for skeletal muscle inotropy. Treating fast twitch skeletal muscle from wild-type mice with the ß-receptor agonist isoproterenol (isoprenaline) increased RyR1 PKA phosphorylation, twitch Ca(2+) and force generation. In contrast, the enhanced muscle Ca(2+), force and in vivo muscle strength responses following isoproterenol stimulation were abrogated in RyR1-S2844A mice in which the serine in the PKA site in RyR1 was replaced with alanine. These data suggest that the molecular mechanism underlying skeletal muscle inotropy requires enhanced SR Ca(2+) release due to PKA phosphorylation of S2844 in RyR1.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoproterenol/pharmacology , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Muscle Strength/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium Signaling/drug effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Fast-Twitch/enzymology , Phosphorylation , Point Mutation , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Serine , Time Factors
9.
Cell ; 150(5): 1055-67, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939628

ABSTRACT

The type 2 ryanodine receptor/calcium release channel (RyR2), required for excitation-contraction coupling in the heart, is abundant in the brain. Chronic stress induces catecholamine biosynthesis and release, stimulating ß-adrenergic receptors and activating cAMP signaling pathways in neurons. In a murine chronic restraint stress model, neuronal RyR2 were phosphorylated by protein kinase A (PKA), oxidized, and nitrosylated, resulting in depletion of the stabilizing subunit calstabin2 (FKBP12.6) from the channel complex and intracellular calcium leak. Stress-induced cognitive dysfunction, including deficits in learning and memory, and reduced long-term potentiation (LTP) at the hippocampal CA3-CA1 connection were rescued by oral administration of S107, a compound developed in our laboratory that stabilizes RyR2-calstabin2 interaction, or by genetic ablation of the RyR2 PKA phosphorylation site at serine 2808. Thus, neuronal RyR2 remodeling contributes to stress-induced cognitive dysfunction. Leaky RyR2 could be a therapeutic target for treatment of stress-induced cognitive dysfunction.


Subject(s)
Cognition Disorders/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Stress Disorders, Traumatic/metabolism
10.
Circ Res ; 111(6): 708-17, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22828895

ABSTRACT

RATIONALE: Atrial fibrillation (AF) is the most common cardiac arrhythmia, however the mechanism(s) causing AF remain poorly understood and therapy is suboptimal. The ryanodine receptor (RyR2) is the major calcium (Ca2+) release channel on the sarcoplasmic reticulum (SR) required for excitation-contraction coupling in cardiac muscle. OBJECTIVE: In the present study, we sought to determine whether intracellular diastolic SR Ca2+ leak via RyR2 plays a role in triggering AF and whether inhibiting this leak can prevent AF. METHODS AND RESULTS: We generated 3 knock-in mice with mutations introduced into RyR2 that result in leaky channels and cause exercise induced polymorphic ventricular tachycardia in humans [catecholaminergic polymorphic ventricular tachycardia (CPVT)]. We examined AF susceptibility in these three CPVT mouse models harboring RyR2 mutations to explore the role of diastolic SR Ca2+ leak in AF. AF was stimulated with an intra-esophageal burst pacing protocol in the 3 CPVT mouse models (RyR2-R2474S+/-, 70%; RyR2-N2386I+/-, 60%; RyR2-L433P+/-, 35.71%) but not in wild-type (WT) mice (P<0.05). Consistent with these in vivo results, there was a significant diastolic SR Ca2+ leak in atrial myocytes isolated from the CPVT mouse models. Calstabin2 (FKBP12.6) is an RyR2 subunit that stabilizes the closed state of RyR2 and prevents a Ca2+ leak through the channel. Atrial RyR2 from RyR2-R2474S+/- mice were oxidized, and the RyR2 macromolecular complex was depleted of calstabin2. The Rycal drug S107 stabilizes the closed state of RyR2 by inhibiting the oxidation/phosphorylation induced dissociation of calstabin2 from the channel. S107 reduced the diastolic SR Ca2+ leak in atrial myocytes and decreased burst pacing-induced AF in vivo. S107 did not reduce the increased prevalence of burst pacing-induced AF in calstabin2-deficient mice, confirming that calstabin2 is required for the mechanism of action of the drug. CONCLUSIONS: The present study demonstrates that RyR2-mediated diastolic SR Ca2+ leak in atrial myocytes is associated with AF in CPVT mice. Moreover, the Rycal S107 inhibited diastolic SR Ca2+ leak through RyR2 and pacing-induced AF associated with CPVT mutations.


Subject(s)
Atrial Fibrillation/metabolism , Calcium/metabolism , Disease Models, Animal , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/metabolism , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Caffeine/pharmacology , Cardiac Pacing, Artificial , Cells, Cultured , Electrocardiography/drug effects , Epinephrine/pharmacology , Gene Knock-In Techniques , Heart/drug effects , Heart/physiopathology , Humans , Immunoblotting , Mice , Mice, Knockout , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Physical Conditioning, Animal/physiology , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Thiazepines/pharmacology
11.
Skelet Muscle ; 2(1): 9, 2012 May 28.
Article in English | MEDLINE | ID: mdl-22640601

ABSTRACT

BACKGROUND: Disruption of the sarcolemma-associated dystrophin-glycoprotein complex underlies multiple forms of muscular dystrophy, including Duchenne muscular dystrophy and sarcoglycanopathies. A hallmark of these disorders is muscle weakness. In a murine model of Duchenne muscular dystrophy, mdx mice, cysteine-nitrosylation of the calcium release channel/ryanodine receptor type 1 (RyR1) on the skeletal muscle sarcoplasmic reticulum causes depletion of the stabilizing subunit calstabin1 (FKBP12) from the RyR1 macromolecular complex. This results in a sarcoplasmic reticular calcium leak via defective RyR1 channels. This pathological intracellular calcium leak contributes to reduced calcium release and decreased muscle force production. It is unknown whether RyR1 dysfunction occurs also in other muscular dystrophies. METHODS: To test this we used a murine model of Limb-Girdle muscular dystrophy, deficient in ß-sarcoglycan (Sgcb-/-). RESULTS: Skeletal muscle RyR1 from Sgcb-/- deficient mice were oxidized, nitrosylated, and depleted of the stabilizing subunit calstabin1, which was associated with increased open probability of the RyR1 channels. Sgcb-/- deficient mice exhibited decreased muscle specific force and calcium transients, and displayed reduced exercise capacity. Treating Sgcb-/- mice with the RyR stabilizing compound S107 improved muscle specific force, calcium transients, and exercise capacity. We have previously reported similar findings in mdx mice, a murine model of Duchenne muscular dystrophy. CONCLUSIONS: Our data suggest that leaky RyR1 channels may underlie multiple forms of muscular dystrophy linked to mutations in genes encoding components of the dystrophin-glycoprotein complex. A common underlying abnormality in calcium handling indicates that pharmacological targeting of dysfunctional RyR1 could be a novel therapeutic approach to improve muscle function in Limb-Girdle and Duchenne muscular dystrophies.

12.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G97-G104, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21960523

ABSTRACT

Secretagogue-stimulated intracellular Ca(2+) signals are fundamentally important for initiating the secretion of the fluid and ion component of saliva from parotid acinar cells. The Ca(2+) signals have characteristic spatial and temporal characteristics, which are defined by the specific properties of Ca(2+) release mediated by inositol 1,4,5-trisphosphate receptors (InsP(3)R). In this study we have investigated the role of adenine nucleotides in modulating Ca(2+) release in mouse parotid acinar cells. In permeabilized cells, the Ca(2+) release rate induced by submaximal [InsP(3)] was increased by 5 mM ATP. Enhanced Ca(2+) release was not observed at saturating [InsP(3)]. The EC(50) for the augmented Ca(2+) release was ∼8 µM ATP. The effect was mimicked by nonhydrolysable ATP analogs. ADP and AMP also potentiated Ca(2+) release but were less potent than ATP. In acini isolated from InsP(3)R-2-null transgenic animals, the rate of Ca(2+) release was decreased under all conditions but now enhanced by ATP at all [InsP(3)]. In addition the EC(50) for ATP potentiation increased to ∼500 µM. These characteristics are consistent with the properties of the InsP(3)R-2 dominating the overall features of InsP(3)R-induced Ca(2+) release despite the expression of all isoforms. Finally, Ca(2+) signals were measured in intact parotid lobules by multiphoton microscopy. Consistent with the release data, carbachol-stimulated Ca(2+) signals were reduced in lobules exposed to experimental hypoxia compared with control lobules only at submaximal concentrations. Adenine nucleotide modulation of InsP(3)R in parotid acinar cells likely contributes to the properties of Ca(2+) signals in physiological and pathological conditions.


Subject(s)
Acinar Cells/drug effects , Adenine Nucleotides/pharmacology , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Parotid Gland/drug effects , Acinar Cells/metabolism , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Hypoxia/metabolism , Mice , Parotid Gland/metabolism
13.
Appl Environ Microbiol ; 78(4): 1215-27, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22179247

ABSTRACT

NADH oxidase (Nox) is a flavin-containing enzyme used by Streptococcus mutans to reduce dissolved oxygen encountered during growth in the oral cavity. In this study, we characterized the role of the NADH oxidase in the oxidative and acid stress responses of S. mutans. A nox-defective mutant strain of S. mutans and its parental strain, the genomic type strain UA159, were exposed to various oxygen concentrations at pH values of 5 and 7 to better understand the adaptive mechanisms used by the organism to withstand environmental pressures. With the loss of nox, the activities of oxygen stress response enzymes such as superoxide dismutase and glutathione oxidoreductase were elevated compared to those in controls, resulting in a greater adaptation to oxygen stress. In contrast, the loss of nox led to a decreased ability to grow in a low-pH environment despite an increased resistance to severe acid challenge. Analysis of the membrane fatty acid composition revealed that for both the nox mutant and UA159 parent strain, growth in an oxygen-rich environment resulted in high proportions of unsaturated membrane fatty acids, independent of external pH. The data indicate that S. mutans membrane fatty acid composition is responsive to oxidative stress, as well as changes in environmental pH, as previously reported (E. M. Fozo and R. G. Quivey, Jr., Appl. Environ. Microbiol. 70:929-936, 2004). The heightened ability of the nox strain to survive acidic and oxidative environmental stress suggests a multifaceted response system that is partially dependent on oxygen metabolites.


Subject(s)
Acids/toxicity , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxygen/toxicity , Streptococcus mutans/drug effects , Streptococcus mutans/physiology , Stress, Physiological , Cell Membrane/chemistry , Fatty Acids/analysis , Hydrogen-Ion Concentration , Microbial Viability/drug effects , Multienzyme Complexes/deficiency , Mutation , NADH, NADPH Oxidoreductases/deficiency , Oxidative Stress , Streptococcus mutans/enzymology
14.
Cell Metab ; 14(2): 196-207, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21803290

ABSTRACT

Age-related loss of muscle mass and force (sarcopenia) contributes to disability and increased mortality. Ryanodine receptor 1 (RyR1) is the skeletal muscle sarcoplasmic reticulum calcium release channel required for muscle contraction. RyR1 from aged (24 months) rodents was oxidized, cysteine-nitrosylated, and depleted of the channel-stabilizing subunit calstabin1, compared to RyR1 from younger (3-6 months) adults. This RyR1 channel complex remodeling resulted in "leaky" channels with increased open probability, leading to intracellular calcium leak in skeletal muscle. Similarly, 6-month-old mice harboring leaky RyR1-S2844D mutant channels exhibited skeletal muscle defects comparable to 24-month-old wild-type mice. Treating aged mice with S107 stabilized binding of calstabin1 to RyR1, reduced intracellular calcium leak, decreased reactive oxygen species (ROS), and enhanced tetanic Ca(2+) release, muscle-specific force, and exercise capacity. Taken together, these data indicate that leaky RyR1 contributes to age-related loss of muscle function.


Subject(s)
Aging , Calcium/metabolism , Muscle Weakness/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcopenia/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , Mitochondria/physiology , Motor Activity/drug effects , Motor Activity/physiology , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/blood , Tacrolimus Binding Proteins/deficiency , Tacrolimus Binding Proteins/metabolism , Thiazepines/pharmacology
15.
J Clin Invest ; 120(12): 4375-87, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21099115

ABSTRACT

Increased sarcoplasmic reticulum (SR) Ca2+ leak via the cardiac ryanodine receptor/calcium release channel (RyR2) is thought to play a role in heart failure (HF) progression. Inhibition of this leak is an emerging therapeutic strategy. To explore the role of chronic PKA phosphorylation of RyR2 in HF pathogenesis and treatment, we generated a knockin mouse with aspartic acid replacing serine 2808 (mice are referred to herein as RyR2-S2808D+/+ mice). This mutation mimics constitutive PKA hyperphosphorylation of RyR2, which causes depletion of the stabilizing subunit FKBP12.6 (also known as calstabin2), resulting in leaky RyR2. RyR2-S2808D+/+ mice developed age-dependent cardiomyopathy, elevated RyR2 oxidation and nitrosylation, reduced SR Ca2+ store content, and increased diastolic SR Ca2+ leak. After myocardial infarction, RyR2-S2808D+/+ mice exhibited increased mortality compared with WT littermates. Treatment with S107, a 1,4-benzothiazepine derivative that stabilizes RyR2-calstabin2 interactions, inhibited the RyR2-mediated diastolic SR Ca2+ leak and reduced HF progression in WT and RyR2-S2808D+/+ mice. In contrast, ß-adrenergic receptor blockers improved cardiac function in WT but not in RyR2-S2808D+/+ mice.Thus, chronic PKA hyperphosphorylation of RyR2 results in a diastolic leak that causes cardiac dysfunction. Reversing PKA hyperphosphorylation of RyR2 is an important mechanism underlying the therapeutic action of ß-blocker therapy in HF.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Heart Failure/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Substitution , Animals , Calcium Signaling/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Failure/drug therapy , Heart Failure/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutation, Missense , Myocardial Infarction/metabolism , Myocardium/metabolism , Phosphorylation , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
16.
J Clin Invest ; 120(12): 4388-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21099118

ABSTRACT

During the classic "fight-or-flight" stress response, sympathetic nervous system activation leads to catecholamine release, which increases heart rate and contractility, resulting in enhanced cardiac output. Catecholamines bind to ß-adrenergic receptors, causing cAMP generation and activation of PKA, which phosphorylates multiple targets in cardiac muscle, including the cardiac ryanodine receptor/calcium release channel (RyR2) required for muscle contraction. PKA phosphorylation of RyR2 enhances channel activity by sensitizing the channel to cytosolic calcium (Ca²+). Here, we found that mice harboring RyR2 channels that cannot be PKA phosphorylated (referred to herein as RyR2-S2808A+/+ mice) exhibited blunted heart rate and cardiac contractile responses to catecholamines (isoproterenol). The isoproterenol-induced enhancement of ventricular myocyte Ca²+ transients and fractional shortening (contraction) and the spontaneous beating rate of sinoatrial nodal cells were all blunted in RyR2-S2808A+/+ mice. The blunted cardiac response to catecholamines in RyR2-S2808A+/+ mice resulted in impaired exercise capacity. RyR2-S2808A+/+ mice were protected against chronic catecholaminergic-induced cardiac dysfunction. These studies identify what we believe to be new roles for PKA phosphorylation of RyR2 in both the heart rate and contractile responses to acute catecholaminergic stimulation.


Subject(s)
Heart/physiology , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Substitution , Animals , Calcium Signaling , Catecholamines/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutation, Missense , Myocardial Contraction , Phosphorylation , Receptors, Adrenergic, beta/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Stress, Physiological
17.
J Biol Chem ; 285(48): 37927-38, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20876535

ABSTRACT

Ca(2+) release through inositol 1,4,5-trisphosphate receptors (InsP(3)R) can be modulated by numerous factors, including input from other signal transduction cascades. These events shape the spatio-temporal characteristics of the Ca(2+) signal and provide fidelity essential for the appropriate activation of effectors. In this study, we investigate the regulation of Ca(2+) release via InsP(3)R following activation of cyclic nucleotide-dependent kinases in the presence and absence of expression of a binding partner InsP(3)R-associated cGMP kinase substrate (IRAG). cGMP-dependent kinase (PKG) phosphorylation of only the S2+ InsP(3)R-1 subtype resulted in enhanced Ca(2+) release in the absence of IRAG expression. In contrast, IRAG bound to each InsP(3)R subtype, and phosphorylation of IRAG by PKG attenuated Ca(2+) release through all InsP(3)R subtypes. Surprisingly, simply the expression of IRAG attenuated phosphorylation and inhibited the enhanced Ca(2+) release through InsP(3)R-1 following cAMP-dependent protein kinase (PKA) activation. In contrast, IRAG expression did not influence the PKA-enhanced activity of the InsP(3)R-2. Phosphorylation of IRAG resulted in reduced Ca(2+) release through all InsP(3)R subtypes during concurrent activation of PKA and PKG, indicating that IRAG modulation is dominant under these conditions. These studies yield mechanistic insight into how cells with various complements of proteins integrate and prioritize signals from ubiquitous signaling pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Phosphoproteins/metabolism , Animals , COS Cells , Calcium/metabolism , Cell Line , Chickens , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Proteins , Mice , Phosphoproteins/genetics , Phosphorylation , Rats
18.
Cell Calcium ; 47(6): 469-79, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20510450

ABSTRACT

Great insight has been gained into the structure and function of the inositol 1,4,5 trisphosphate receptor (InsP(3)R) by studies employing mutagenesis of the cDNA encoding the receptor. Notably, early studies using this approach defined the key constituents required for InsP(3) binding in the N-terminus and the membrane spanning regions in the C-terminal domain responsible for channel formation, targeting and function. In this article we evaluate recent studies which have used a similar approach to investigate key residues underlying the in vivo modulation by select regulatory factors. In addition, we review studies defining the structural requirements in the channel domain which comprise the conduction pathway and are suggested to be involved in the gating of the channel.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Calcium Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mutagenesis , Protein Structure, Tertiary
19.
Proc Natl Acad Sci U S A ; 107(22): 10274-9, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20479242

ABSTRACT

The force frequency relationship (FFR), first described by Bowditch 139 years ago as the observation that myocardial contractility increases proportionally with increasing heart rate, is an important mediator of enhanced cardiac output during exercise. Individuals with heart failure have defective positive FFR that impairs their cardiac function in response to stress, and the degree of positive FFR deficiency correlates with heart failure progression. We have identified a mechanism for FFR involving heart rate dependent phosphorylation of the major cardiac sarcoplasmic reticulum calcium release channel/ryanodine receptor (RyR2), at Ser2814, by calcium/calmodulin-dependent serine/threonine kinase-delta (CaMKIIdelta). Mice engineered with an RyR2-S2814A mutation have RyR2 channels that cannot be phosphorylated by CaMKIIdelta, and exhibit a blunted positive FFR. Ex vivo hearts from RyR2-S2814A mice also have blunted positive FFR, and cardiomyocytes isolated from the RyR2-S2814A mice exhibit impaired rate-dependent enhancement of cytosolic calcium levels and fractional shortening. The cardiac RyR2 macromolecular complexes isolated from murine and human failing hearts have reduced CaMKIIdelta levels. These data indicate that CaMKIIdelta phosphorylation of RyR2 plays an important role in mediating positive FFR in the heart, and that defective regulation of RyR2 by CaMKIIdelta-mediated phosphorylation is associated with the loss of positive FFR in failing hearts.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/physiopathology , Heart Rate/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cardiac Output/genetics , Cardiac Output/physiology , DNA Primers/genetics , Heart Rate/genetics , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myocardial Contraction/genetics , Myocytes, Cardiac/physiology , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics
20.
FEBS Lett ; 584(10): 1956-65, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20214899

ABSTRACT

Ryanodine receptors (RyR) regulate intracellular Ca(2+) release in many cell types and have been implicated in a number of inherited human diseases. Over the past 15 years genetically engineered mouse models have been developed to elucidate the role that RyRs play in physiology and pathophysiology. To date these models have implicated RyRs in fundamental biological processes including excitation-contraction coupling and long term plasticity as well as diseases including malignant hyperthermia, cardiac arrhythmias, heart failure, and seizures. In this review we summarize the RyR mouse models and how they have enhanced our understanding of the RyR channels and their roles in cellular physiology and disease.


Subject(s)
Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Brain/cytology , Brain/metabolism , Brain/pathology , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...