Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
Proc Natl Acad Sci U S A ; 119(36): e2206708119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36044551

ABSTRACT

The sinoatrial node (SAN), the leading pacemaker region, generates electrical impulses that propagate throughout the heart. SAN dysfunction with bradyarrhythmia is well documented in heart failure (HF). However, the underlying mechanisms are not completely understood. Mitochondria are critical to cellular processes that determine the life or death of the cell. The release of Ca2+ from the ryanodine receptors 2 (RyR2) on the sarcoplasmic reticulum (SR) at mitochondria-SR microdomains serves as the critical communication to match energy production to meet metabolic demands. Therefore, we tested the hypothesis that alterations in the mitochondria-SR connectomics contribute to SAN dysfunction in HF. We took advantage of a mouse model of chronic pressure overload-induced HF by transverse aortic constriction (TAC) and a SAN-specific CRISPR-Cas9-mediated knockdown of mitofusin-2 (Mfn2), the mitochondria-SR tethering GTPase protein. TAC mice exhibited impaired cardiac function with HF, cardiac fibrosis, and profound SAN dysfunction. Ultrastructural imaging using electron microscope (EM) tomography revealed abnormal mitochondrial structure with increased mitochondria-SR distance. The expression of Mfn2 was significantly down-regulated and showed reduced colocalization with RyR2 in HF SAN cells. Indeed, SAN-specific Mfn2 knockdown led to alterations in the mitochondria-SR microdomains and SAN dysfunction. Finally, disruptions in the mitochondria-SR microdomains resulted in abnormal mitochondrial Ca2+ handling, alterations in localized protein kinase A (PKA) activity, and impaired mitochondrial function in HF SAN cells. The current study provides insights into the role of mitochondria-SR microdomains in SAN automaticity and possible therapeutic targets for SAN dysfunction in HF patients.


Subject(s)
Connectome , Heart Failure , Mitochondria, Heart , Sarcoplasmic Reticulum , Sick Sinus Syndrome , Sinoatrial Node , Animals , Heart Failure/pathology , Heart Failure/physiopathology , Mice , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/pathology , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/physiopathology
2.
Sheng Li Xue Bao ; 74(2): 188-200, 2022 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-35503066

ABSTRACT

Atrial Ca2+ handling abnormalities, mainly involving the dysfunction of ryanodine receptor (RyR) and sarcoplasmic reticulum Ca2+-ATPase (SERCA), play a role in the pathogenesis of atrial fibrillation (AF). Previously, we found that the expression and function of transient receptor potential vanilloid subtype 4 (TRPV4) are upregulated in a sterile pericarditis (SP) rat model of AF, and oral administration of TRPV4 inhibitor GSK2193874 alleviates AF in this animal model. The aim of this study was to investigate whether oral administration of GSK2193874 could alleviate atrial Ca2+ handling abnormalities in SP rats. A SP rat model of AF was established by daubing sterile talcum powder on both atria of Sprague-Dawley (SD) rats after a pericardiotomy, to simulate the pathogenesis of postoperative atrial fibrillation (POAF). On the 3rd postoperative day, Ca2+ signals of atria were collected in isolated perfused hearts by optical mapping. Ca2+ transient duration (CaD), alternan, and the recovery properties of Ca2+ transient (CaT) were quantified and analyzed. GSK2193874 treatment reversed the abnormal prolongation of time to peak (determined mainly by RyR activity) and CaD (determined mainly by SERCA activity), as well as the regional heterogeneity of CaD in SP rats. Furthermore, GSK2193874 treatment relieved alternan in SP rats, and reduced its incidence of discordant alternan (DIS-ALT). More importantly, GSK2193874 treatment prevented the reduction of the S2/S1 CaT ratio (determined mainly by RyR refractoriness) in SP rats, and decreased its regional heterogeneity. Taken together, oral administration of TRPV4 inhibitor alleviates Ca2+ handling abnormalities in SP rats primarily by blocking the TRPV4-Ca2+-RyR pathway, and thus exerts therapeutic effect on POAF.


Subject(s)
Atrial Fibrillation , Pericarditis , Administration, Oral , Animals , Atrial Fibrillation/drug therapy , Atrial Fibrillation/etiology , Calcium/metabolism , Myocytes, Cardiac/metabolism , Pericarditis/complications , Pericarditis/metabolism , Pericarditis/pathology , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/pharmacology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , TRPV Cation Channels
3.
Acta Physiologica Sinica ; (6): 188-200, 2022.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-927594

ABSTRACT

Atrial Ca2+ handling abnormalities, mainly involving the dysfunction of ryanodine receptor (RyR) and sarcoplasmic reticulum Ca2+-ATPase (SERCA), play a role in the pathogenesis of atrial fibrillation (AF). Previously, we found that the expression and function of transient receptor potential vanilloid subtype 4 (TRPV4) are upregulated in a sterile pericarditis (SP) rat model of AF, and oral administration of TRPV4 inhibitor GSK2193874 alleviates AF in this animal model. The aim of this study was to investigate whether oral administration of GSK2193874 could alleviate atrial Ca2+ handling abnormalities in SP rats. A SP rat model of AF was established by daubing sterile talcum powder on both atria of Sprague-Dawley (SD) rats after a pericardiotomy, to simulate the pathogenesis of postoperative atrial fibrillation (POAF). On the 3rd postoperative day, Ca2+ signals of atria were collected in isolated perfused hearts by optical mapping. Ca2+ transient duration (CaD), alternan, and the recovery properties of Ca2+ transient (CaT) were quantified and analyzed. GSK2193874 treatment reversed the abnormal prolongation of time to peak (determined mainly by RyR activity) and CaD (determined mainly by SERCA activity), as well as the regional heterogeneity of CaD in SP rats. Furthermore, GSK2193874 treatment relieved alternan in SP rats, and reduced its incidence of discordant alternan (DIS-ALT). More importantly, GSK2193874 treatment prevented the reduction of the S2/S1 CaT ratio (determined mainly by RyR refractoriness) in SP rats, and decreased its regional heterogeneity. Taken together, oral administration of TRPV4 inhibitor alleviates Ca2+ handling abnormalities in SP rats primarily by blocking the TRPV4-Ca2+-RyR pathway, and thus exerts therapeutic effect on POAF.


Subject(s)
Animals , Rats , Administration, Oral , Atrial Fibrillation/etiology , Calcium/metabolism , Myocytes, Cardiac/metabolism , Pericarditis/pathology , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/pharmacology , Sarcoplasmic Reticulum/pathology , TRPV Cation Channels
4.
J Am Coll Cardiol ; 78(24): 2439-2453, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34886965

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy is associated with progressive deterioration in left ventricular (LV) function. The golden retriever muscular dystrophy (GRMD) dog model recapitulates the pathology and clinical manifestations of Duchenne muscular dystrophy. Importantly, they develop progressive LV dysfunction starting at early age. OBJECTIVES: The authors tested the cardioprotective effect of chronic administration of the ARM036, a small molecule that stabilizes the closed conformation of the cardiac sarcoplasmic reticulum ryanodine receptor/calcium release channel (RyR2) in young GRMD-dogs. METHODS: Two-month-old GRMD-dogs were treated with ARM036 or placebo for 4 months. Healthy-dogs of the same genetic background served as controls. Cardiac function was evaluated by conventional and 2-dimensional speckle-tracking echocardiography. Cardiac cellular and molecular analyses were performed at 6 months old. RESULTS: Conventional echocardiography showed normal LV dimensions and ejection fraction in 6-month-old GRMD dogs. Interestingly, 2-dimensional speckle-tracking echocardiography revealed decreased global longitudinal strain and the presence of hypokinetic segments in placebo-treated GRMD dogs. Single-channel measurements revealed higher RyR2 open probability at low resting Ca2+ in GRMD cardiomyocytes than in controls. ARM036 prevented those in vivo and in vitro dysfunctions in GRMD dogs. Myofilament Ca2+-sensitivity was increased in permeabilized GRMD cardiomyocytes at short sarcomere length. ARM036 had no effect on this parameter. Cross-bridge cycling kinetics were altered in GRMD myocytes and recovered with ARM036 treatment, which coincided with the level of myosin binding protein-C-S glutathionylation. CONCLUSIONS: GRMD-dogs exhibit early LV dysfunction associated with altered myofilament contractile properties. These abnormalities were prevented pharmacologically by stabilizing RyR2 with ARM036.


Subject(s)
Muscular Dystrophy, Duchenne/complications , Ryanodine Receptor Calcium Release Channel/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/physiology , Animals , Biopsy , Disease Models, Animal , Dogs , Echocardiography , Muscular Dystrophy, Duchenne/diagnosis , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Myofibrils/pathology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
5.
Bull Exp Biol Med ; 172(2): 117-120, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855078

ABSTRACT

We compared the expression of Са2+-ATPase (SERCA2a), calsequestrin (CASQ2), ryanodine receptors (RyR2) proteins and their genes (ATP2A2, CASQ2, and RYR2) in coronary heart disease (CHD) patients with and without comorbid type 2 diabetes mellitus. All studies were performed on the right atrial appendages resected during coronary bypass surgeries. Expression of SERCA2a and RyR2 proteins and their ATP2A2 (p=0.046) and RYR2 genes in comorbid pathology was significantly (p=0.042) higher (by 1.2 and 2 times; p=0.025). The expression of CASQ2 protein and its gene did not differ significantly between the groups (p=0.82 and p=0.066, respectively). It was concluded that the expression of SERCA2a and RyR2 proteins and their genes (but not CASQ2 and its gene) is elevated in CHD associated with type 2 diabetes mellitus. Expression of the studied proteins correlated with the expression of their genes. Increased expression of CASQ2 protein and its gene can probably prevent imbalance of the Ca2+-transporting systems in cardiomyocytes and contractile dysfunction of the myocardium, even in CHD associated with type 2 diabetes mellitus.


Subject(s)
Calcium Signaling/genetics , Coronary Disease , Diabetes Mellitus, Type 2 , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Aged , Biological Transport/genetics , Biopsy , Calcium/metabolism , Calsequestrin/genetics , Calsequestrin/metabolism , Case-Control Studies , Coronary Disease/complications , Coronary Disease/genetics , Coronary Disease/metabolism , Coronary Disease/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression , Humans , Middle Aged , Myocardium/metabolism , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
6.
Mol Cell Biochem ; 476(10): 3827-3844, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34114148

ABSTRACT

Metabolic syndrome (MetS) is associated with additional cardiovascular risk in mammalians while there are relationships between hyperglycemia-associated cardiovascular dysfunction and increased platelet P2Y12 receptor activation. Although P2Y12 receptor antagonist ticagrelor (Tica) plays roles in reduction of cardiovascular events, its beneficial mechanism remains poorly understood. Therefore, we aimed to clarify whether Tica can exert a direct protective effect in ventricular cardiomyocytes from high-carbohydrate diet-induced MetS rats, at least, through affecting sarcoplasmic reticulum (SR)-mitochondria (Mit) miscommunication. Tica treatment of MetS rats (150 mg/kg/day for 15 days) significantly reversed the altered parameters of action potentials by reversing sarcolemmal ionic currents carried by voltage-dependent Na+ and K+ channels, and Na+/Ca2+-exchanger in the cells, expressed P2Y12 receptors. The increased basal-cytosolic Ca2+ level and depressed SR Ca2+ load were also reversed in Tica-treated cells, at most, though recoveries in the phosphorylation levels of ryanodine receptors and phospholamban. Moreover, there were marked recoveries in Mit structure and function (including increases in both autophagosomes and fragmentations) together with recoveries in Mit proteins and the factors associated with Ca2+ transfer between SR-Mit. There were further significant recoveries in markers of both ER stress and oxidative stress. Taken into consideration the Tica-induced prevention of ER stress and mitochondrial dysfunction, our data provided an important document on the pleiotropic effects of Tica in the electrical activity of the cardiomyocytes from MetS rats. This protective effect seems through recoveries in SR-Mit miscommunication besides modulation of different sarcolemmal ion-channel activities, independent of P2Y12 receptor antagonism.


Subject(s)
Action Potentials/drug effects , Dietary Carbohydrates/adverse effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Ticagrelor/pharmacology , Animals , Dietary Carbohydrates/pharmacology , Ion Transport/drug effects , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Sarcoplasmic Reticulum/pathology , Signal Transduction/drug effects
7.
Hum Mol Genet ; 30(11): 1006-1019, 2021 05 31.
Article in English | MEDLINE | ID: mdl-33822956

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscular weakness because of the loss of dystrophin. Extracellular Ca2+ flows into the cytoplasm through membrane tears in dystrophin-deficient myofibers, which leads to muscle contracture and necrosis. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) takes up cytosolic Ca2+ into the sarcoplasmic reticulum, but its activity is decreased in dystrophic muscle. Here, we show that an allosteric SERCA activator, CDN1163, ameliorates dystrophic phenotypes in dystrophin-deficient mdx mice. The administration of CDN1163 prevented exercise-induced muscular damage and restored mitochondrial function. In addition, treatment with CDN1163 for 7 weeks enhanced muscular strength and reduced muscular degeneration and fibrosis in mdx mice. Our findings provide preclinical proof-of-concept evidence that pharmacological activation of SERCA could be a promising therapeutic strategy for DMD. Moreover, CDN1163 improved muscular strength surprisingly in wild-type mice, which may pave the new way for the treatment of muscular dysfunction.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Calcium/metabolism , Disease Models, Animal , Dystrophin/deficiency , Humans , Mice , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Dystrophy, Duchenne/pathology , Phenotype , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
8.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33669042

ABSTRACT

Since its first identification as a cardiac transverse tubule (t-tubule) protein, followed by the cloning of the cardiac isoform responsible for t-tubule membrane microdomain formation, cardiac bridging integrator 1 (cBIN1) and its organized microdomains have emerged as a key mechanism in maintaining normal beat-to-beat heart contraction and relaxation. The abnormal remodeling of cBIN1-microdomains occurs in stressed and diseased cardiomyocytes, contributing to the pathophysiology of heart failure. Due to the homeostatic turnover of t-tubule cBIN1-microdomains via microvesicle release into the peripheral circulation, plasma cBIN1 can be assayed as a liquid biopsy of cardiomyocyte health. A new blood test cBIN1 score (CS) has been developed as a dimensionless inverse index derived from plasma cBIN1 concentration with a diagnostic and prognostic power for clinical outcomes in stable ambulatory patients with heart failure with reduced or preserved ejection fraction (HFrEF or HFpEF). Recent evidence further indicates that exogenous cBIN1 introduced by adeno-associated virus 9-based gene therapy can rescue cardiac contraction and relaxation in failing hearts. The therapeutic potential of cBIN1 gene therapy is enormous given its ability to rescue cardiac inotropy and provide lusitropic protection in the meantime. These unprecedented capabilities of cBIN1 gene therapy are shifting the current paradigm of therapy development for heart failure, particularly HFpEF.


Subject(s)
Adaptor Proteins, Signal Transducing/blood , Genetic Therapy/methods , Heart Failure/blood , Myocytes, Cardiac/metabolism , Nuclear Proteins/blood , Sarcoplasmic Reticulum/metabolism , Tumor Suppressor Proteins/blood , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers/blood , Calcium Signaling/physiology , Heart Failure/diagnosis , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Membrane Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Sarcolemma/metabolism , Sarcoplasmic Reticulum/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
FASEB J ; 35(5): e21349, 2021 05.
Article in English | MEDLINE | ID: mdl-33786938

ABSTRACT

Mice with a mutation (D244G, DG) in calsequestrin 1 (CASQ1), analogous to a human mutation in CASQ1 associated with a delayed onset human myopathy (vacuolar aggregate myopathy), display a progressive myopathy characterized by decreased activity, decreased ability of fast twitch muscles to generate force and low body weight after one year of age. The DG mutation causes CASQ1 to partially dissociate from the junctional sarcoplasmic reticulum (SR) and accumulate in the endoplasmic reticulum (ER). Decreased junctional CASQ1 reduces SR Ca2+ release. Muscles from older DG mice display ER stress, ER expansion, increased mTOR signaling, inadequate clearance of aggregated proteins by the proteasomes, and elevation of protein aggregates and lysosomes. This study suggests that the myopathy associated with the D244G mutation in CASQ1 is driven by CASQ1 mislocalization, reduced SR Ca2+ release, CASQ1 misfolding/aggregation and ER stress. The subsequent maladaptive increase in protein synthesis and decreased protein aggregate clearance are likely to contribute to disease progression.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium/metabolism , Endoplasmic Reticulum Stress , Lysosomal Storage Diseases/pathology , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Mutation , Sarcoplasmic Reticulum/pathology , Animals , Calsequestrin , Lysosomal Storage Diseases/etiology , Lysosomal Storage Diseases/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Muscular Diseases/etiology , Muscular Diseases/metabolism , Sarcoplasmic Reticulum/metabolism
10.
J Cell Mol Med ; 25(8): 3922-3934, 2021 04.
Article in English | MEDLINE | ID: mdl-33619882

ABSTRACT

Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an X-linked disease affecting male and rarely adult heterozygous females, resulting in death by the late 20s to early 30s. Previous studies reported depressed left ventricular function in DMD patients which may result from deranged intracellular Ca2+ -handling. To decipher the mechanism(s) underlying the depressed LV function, we tested the hypothesis that iPSC-CMs generated from DMD patients feature blunted positive inotropic response to ß-adrenergic stimulation. To test the hypothesis, [Ca2+ ]i transients and contractions were recorded from healthy and DMD-CMs. While in healthy CMs (HC) isoproterenol caused a prominent positive inotropic effect, DMD-CMs displayed a blunted inotropic response. Next, we tested the functionality of the sarcoplasmic reticulum (SR) by measuring caffeine-induced Ca2+ release. In contrast to HC, DMD-CMs exhibited reduced caffeine-induced Ca2+ signal amplitude and recovery time. In support of the depleted SR Ca2+ stores hypothesis, in DMD-CMs the negative inotropic effects of ryanodine and cyclopiazonic acid were smaller than in HC. RNA-seq analyses demonstrated that in DMD CMs the RNA-expression levels of specific subunits of the L-type calcium channel, the ß1-adrenergic receptor (ADRß1) and adenylate cyclase were down-regulated by 3.5-, 2.8- and 3-fold, respectively, which collectively contribute to the depressed ß-adrenergic responsiveness.


Subject(s)
Adrenergic Agents/pharmacology , Calcium/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/pathology , Muscular Dystrophy, Duchenne/pathology , Myocardial Contraction , Myocytes, Cardiac/pathology , Adult , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Differentiation , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA-Seq , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
11.
J Neuropathol Exp Neurol ; 79(12): 1276-1292, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33184660

ABSTRACT

We investigated the immunohistochemical localization of several proteins related to excitation-contraction coupling and ultrastructural alterations of the sarcotubular system in biopsied muscles from a father and a daughter in a family with permanent myopathy with hypokalemic periodic paralysis (PMPP) due to a mutation in calcium channel CACNA1S; p. R1239H hetero. Immunostaining for L-type calcium channels (LCaC) showed linear hyper-stained regions indicating proliferation of longitudinal t-tubules. The margin of vacuoles was positive for ryanodine receptor, LCaC, calsequestrin (CASQ) 1, CASQ 2, SR/ER Ca2+-ATPase (SERCA) 1, SERCA2, dysferlin, dystrophin, α-actinin, LC3, and LAMP 1. Electron microscopy indicated that the vacuoles mainly originated from the sarcoplasmic reticulum (SR). These findings indicate impairment of the muscle contraction system related to Ca2+ dynamics, remodeling of t-tubules and muscle fiber repair. We speculate that PMPP in patients with a CACNA1S mutation might start with abnormal SR function due to impaired LCaC. Subsequent induction of muscular contractile abnormalities and the vacuoles formed by fused SR in the repair process including autophagy might result in permanent myopathy. Our findings may facilitate prediction of the pathomechanisms of PMPP seen on morphological observation.


Subject(s)
Calcium Channels, L-Type/genetics , Hypokalemic Periodic Paralysis/pathology , Muscle Fibers, Skeletal/pathology , Muscular Diseases/pathology , Mutation , Sarcoplasmic Reticulum/pathology , Adult , Aged , Calcium Channels, L-Type/metabolism , Female , Humans , Hypokalemic Periodic Paralysis/genetics , Hypokalemic Periodic Paralysis/metabolism , Male , Muscle Fibers, Skeletal/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
12.
Int J Mol Sci ; 21(22)2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33238586

ABSTRACT

Risk of cardiovascular disease (CVD) increases considerably as renal function declines in chronic kidney disease (CKD). Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) has emerged as a novel innate immune receptor involved in both CVD and CKD. Following activation, NOD1 undergoes a conformational change that allows the activation of the receptor-interacting serine/threonine protein kinase 2 (RIP2), promoting an inflammatory response. We evaluated whether the genetic deficiency of Nod1 or Rip2 in mice could prevent cardiac Ca2+ mishandling induced by sixth nephrectomy (Nx), a model of CKD. We examined intracellular Ca2+ dynamics in cardiomyocytes from Wild-type (Wt), Nod1-/- and Rip2-/- sham-operated or nephrectomized mice. Compared with Wt cardiomyocytes, Wt-Nx cells showed an impairment in the properties and kinetics of the intracellular Ca2+ transients, a reduction in both cell shortening and sarcoplasmic reticulum Ca2+ load, together with an increase in diastolic Ca2+ leak. Cardiomyocytes from Nod1-/--Nx and Rip2-/--Nx mice showed a significant amelioration in Ca2+ mishandling without modifying the kidney impairment induced by Nx. In conclusion, Nod1 and Rip2 deficiency prevents the intracellular Ca2+ mishandling induced by experimental CKD, unveiling new innate immune targets for the development of innovative therapeutic strategies to reduce cardiac complications in patients with CKD.


Subject(s)
Kidney/metabolism , Nod1 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Renal Insufficiency, Chronic/genetics , Animals , Calcium/metabolism , Calcium Signaling/genetics , Disease Models, Animal , Humans , Kidney/pathology , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/genetics , Nod1 Signaling Adaptor Protein/ultrastructure , Protein Conformation , Receptor-Interacting Protein Serine-Threonine Kinase 2/ultrastructure , Renal Insufficiency, Chronic/pathology , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/pathology
13.
JCI Insight ; 5(19)2020 10 02.
Article in English | MEDLINE | ID: mdl-32897880

ABSTRACT

Huntington's disease (HD) is a progressive, autosomal dominant neurodegenerative disorder affecting striatal neurons beginning in young adults with loss of muscle coordination and cognitive decline. Less appreciated is the fact that patients with HD also exhibit cardiac and respiratory dysfunction, including pulmonary insufficiency and cardiac arrhythmias. The underlying mechanism for these symptoms is poorly understood. In the present study we provide insight into the cause of cardiorespiratory dysfunction in HD and identify a potentially novel therapeutic target. We now show that intracellular calcium (Ca2+) leak via posttranslationally modified ryanodine receptor/intracellular calcium release (RyR) channels plays an important role in HD pathology. RyR channels were oxidized, PKA phosphorylated, and leaky in brain, heart, and diaphragm both in patients with HD and in a murine model of HD (Q175). HD mice (Q175) with endoplasmic reticulum Ca2+ leak exhibited cognitive dysfunction, decreased parasympathetic tone associated with cardiac arrhythmias, and reduced diaphragmatic contractile function resulting in impaired respiratory function. Defects in cognitive, motor, and respiratory functions were ameliorated by treatment with a novel Rycal small-molecule drug (S107) that fixes leaky RyR. Thus, leaky RyRs likely play a role in neuronal, cardiac, and diaphragmatic pathophysiology in HD, and RyRs are a potential novel therapeutic target.


Subject(s)
Arrhythmias, Cardiac/pathology , Calcium Signaling , Calcium/metabolism , Disease Models, Animal , Huntington Disease/complications , Respiratory Insufficiency/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Aged , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Case-Control Studies , Female , Humans , Male , Mice , Middle Aged , Neurons/metabolism , Neurons/pathology , Respiratory Insufficiency/etiology , Respiratory Insufficiency/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
14.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165807, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32353614

ABSTRACT

OBJECTIVES: Investigation of the effect of SGLT2 inhibition by empagliflozin on left ventricular function in a model of diabetic cardiomyopathy. BACKGROUND: SGLT2 inhibition is a new strategy to treat diabetes. In the EMPA-REG Outcome trial empagliflozin treatment reduced cardiovascular and overall mortality in patients with diabetes presumably due to beneficial cardiac effects, leading to reduced heart failure hospitalization. The relevant mechanisms remain currently elusive but might be mediated by a shift in cardiac substrate utilization leading to improved energetic supply to the heart. METHODS: We used db/db mice on high-fat western diet with or without empagliflozin treatment as a model of severe diabetes. Left ventricular function was assessed by pressure catheter with or without dobutamine stress. RESULTS: Treatment with empagliflozin significantly increased glycosuria, improved glucose metabolism, ameliorated left ventricular diastolic function and reduced mortality of mice. This was associated with reduced cardiac glucose concentrations and decreased calcium/calmodulin-dependent protein kinase (CaMKII) activation with subsequent less phosphorylation of the ryanodine receptor (RyR). No change of cardiac ketone bodies or branched-chain amino acid (BCAA) metabolites in serum was detected nor was cardiac expression of relevant catabolic enzymes for these substrates affected. CONCLUSIONS: In a murine model of severe diabetes empagliflozin-dependent SGLT2 inhibition improved diastolic function and reduced mortality. Improvement of diastolic function was likely mediated by reduced spontaneous diastolic sarcoplasmic reticulum (SR) calcium release but independent of changes in cardiac ketone and BCAA metabolism.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/drug therapy , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/genetics , Amino Acids, Branched-Chain/blood , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Clinical Trials as Topic , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/mortality , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/mortality , Diabetic Cardiomyopathies/pathology , Diet, High-Fat/adverse effects , Glucose/metabolism , Humans , Ketone Bodies/blood , Male , Mice , Mice, Transgenic , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Sodium-Glucose Transporter 2/metabolism , Survival Analysis , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
15.
Neuropathol Appl Neurobiol ; 46(6): 579-587, 2020 10.
Article in English | MEDLINE | ID: mdl-32144790

ABSTRACT

AIMS: Nakajo-Nishimura syndrome (NNS) is an autosomal recessive disease caused by biallelic mutations in the PSMB8 gene that encodes the immunoproteasome subunit ß5i. There have been only a limited number of reports on the clinicopathological features of the disease in genetically confirmed cases. METHODS: We studied clinical and pathological features of three NNS patients who all carry the homozygous p.G201V mutations in PSMB8. Patients' muscle specimens were analysed with histology and immunohistochemistry. RESULTS: All patients had episodes of typical periodic fever and skin rash, and later developed progressive muscle weakness and atrophy, similar to previous reports. Oral corticosteroid was used for treatment but showed no obvious efficacy. On muscle pathology, lymphocytes were present in the endomysium surrounding non-necrotic fibres, as well as in the perimysium perivascular area. Nearly all fibres strongly expressed MHC-I in the sarcolemma. In the eldest patient, there were abnormal protein aggregates in the sarcoplasm, immunoreactive to p62, TDP-43 and ubiquitin antibodies. CONCLUSIONS: These results suggest that inflammation, inclusion pathology and aggregation of abnormal proteins underlie the progressive clinical course of the NNS pathomechanism.


Subject(s)
Erythema Nodosum/genetics , Erythema Nodosum/pathology , Fingers/abnormalities , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Myositis/genetics , Myositis/pathology , Sarcoplasmic Reticulum/pathology , Adult , Age of Onset , Child, Preschool , Exanthema/genetics , Exanthema/pathology , Female , Fever/genetics , Fever/pathology , Fingers/pathology , Genes, MHC Class I/genetics , Humans , Infant , Lymphocytes/pathology , Male , Muscle Weakness/genetics , Muscle Weakness/pathology , Mutation/genetics , Nerve Fibers/pathology , Proteasome Endopeptidase Complex/genetics , Sarcolemma/pathology , Young Adult
16.
Nat Commun ; 11(1): 965, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32075961

ABSTRACT

The sarco-endoplasmic reticulum (SR/ER) plays an important role in the development and progression of many heart diseases. However, many aspects of its structural organization remain largely unknown, particularly in cells with a highly differentiated SR/ER network. Here, we report a cardiac enriched, SR/ER membrane protein, REEP5 that is centrally involved in regulating SR/ER organization and cellular stress responses in cardiac myocytes. In vitro REEP5 depletion in mouse cardiac myocytes results in SR/ER membrane destabilization and luminal vacuolization along with decreased myocyte contractility and disrupted Ca2+ cycling. Further, in vivo CRISPR/Cas9-mediated REEP5 loss-of-function zebrafish mutants show sensitized cardiac dysfunction upon short-term verapamil treatment. Additionally, in vivo adeno-associated viral (AAV9)-induced REEP5 depletion in the mouse demonstrates cardiac dysfunction. These results demonstrate the critical role of REEP5 in SR/ER organization and function as well as normal heart function and development.


Subject(s)
Heart/physiopathology , Membrane Proteins/deficiency , Sarcoplasmic Reticulum/pathology , Animals , Calcium/metabolism , Cells, Cultured , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Gene Silencing , Heart/growth & development , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Zebrafish
17.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118502, 2020 03.
Article in English | MEDLINE | ID: mdl-31269418

ABSTRACT

This work discusses active and passive electrical properties of transverse (T-)tubules in ventricular cardiomyocytes to understand the physiological roles of T-tubules. T-tubules are invaginations of the lateral membrane that provide a large surface for calcium-handling proteins to facilitate sarcomere shortening. Higher heart rates correlate with higher T-tubular densities in mammalian ventricular cardiomyocytes. We assess ion dynamics in T-tubules and the effects of sodium current in T-tubules on the extracellular potential, which leads to a partial reduction of the sodium current in deep segments of a T-tubule. We moreover reflect on the impact of T-tubules on macroscopic conduction velocity, integrating fundamental principles of action potential propagation and conduction. We also theoretically assess how the conduction velocity is affected by different T-tubular sodium current densities. Lastly, we critically assess literature on ion channel expression to determine whether action potentials can be initiated in T-tubules.


Subject(s)
Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Sarcomeres/metabolism , Action Potentials/physiology , Calcium Signaling/genetics , Electromagnetic Phenomena , Heart Ventricles/pathology , Humans , Myocytes, Cardiac/pathology , Sarcolemma/pathology , Sarcomeres/pathology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Sodium/metabolism
18.
Cardiovasc Res ; 116(5): 1021-1031, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31373602

ABSTRACT

AIMS: Matrix metalloproteinase-2 (MMP-2) is a zinc-dependent protease which contributes to cardiac contractile dysfunction when activated during myocardial ischaemia-reperfusion (IR) injury. MMP-2 is localized to several subcellular sites inside cardiac myocytes; however, its role in the sarcoplasmic reticulum (SR) is unknown. The Ca2+ ATPase SERCA2a, which pumps cytosolic Ca2+ into the SR to facilitate muscle relaxation, is degraded in cardiac IR injury; however, the protease responsible for this is unclear. We hypothesized that MMP-2 contributes to cardiac contractile dysfunction by proteolyzing SERCA2a, thereby impairing its activity in IR injury. METHODS AND RESULTS: Isolated rat hearts were subjected to IR injury in the presence or absence of the selective MMP inhibitor ARP-100, or perfused aerobically as a control. Inhibition of MMP activity with ARP-100 significantly improved the recovery of cardiac mechanical function and prevented the increase of a 70 kDa SERCA2a degradation fragment following IR injury, although 110 kDa SERCA2a and phospholamban levels appeared unchanged. Electrophoresis of IR heart samples followed by LC-MS/MS confirmed the presence of a SERCA2a fragment of ∼70 kDa. MMP-2 activity co-purified with SR-enriched microsomes prepared from the isolated rat hearts. Endogenous SERCA2a in SR-enriched microsomes was proteolyzed to ∼70 kDa products when incubated in vitro with exogenous MMP-2. MMP-2 also cleaved purified porcine SERCA2a in vitro. SERCA activity in SR-enriched microsomes was decreased by IR injury; however, this was not prevented with ARP-100. CONCLUSION: This study shows that MMP-2 activity is found in SR-enriched microsomes from heart muscle and that SERCA2a is proteolyzed by MMP-2. The cardioprotective actions of MMP inhibition in myocardial IR injury may include the prevention of SERCA2a degradation.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Myocardial Contraction , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/enzymology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Hydroxamic Acids/pharmacology , Isolated Heart Preparation , Male , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Proteolysis , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/pathology , Sulfones/pharmacology
19.
J Physiol ; 598(4): 773-787, 2020 02.
Article in English | MEDLINE | ID: mdl-31785106

ABSTRACT

KEY POINTS: Changes in intramuscular Ca2+ handling contribute to development of fatigue and disease-related loss of muscle mass and function. To date, no data on human intact living muscle fibres have been described. We manually dissected intact single fibres from human intercostal muscle and simultaneously measured force and myoplasmic free [Ca2+ ] at physiological temperature. Based on their fatigue resistance, two distinct groups of fibres were distinguished: fatigue sensitive and fatigue resistant. Force depression in fatigue and during recovery was due to impaired sarcoplasmic reticulum Ca2+ release in both groups of fibres. Acidification did not affect force production in unfatigued fibres and did not affect fatigue development in fatigue-resistant fibres. The current study provides novel insight into the mechanisms of fatigue in human intercostal muscle. ABSTRACT: Changes in intracellular Ca2+ handling of individual skeletal muscle fibres cause a force depression following physical activity and are also implicated in disease-related loss of function. The relation of intracellular Ca2+ handling with muscle force production and fatigue tolerance is best studied in intact living single fibres that allow continuous measurements of force and myoplasmic free [Ca2+ ] during repeated contractions. To this end, manual dissections of human intercostal muscle biopsies were performed to isolate intact single fibres. Based on the ability to maintain tetanic force at >40% of the initial value during 500 fatiguing contractions, fibres were classified as either fatigue sensitive or fatigue resistant. Following fatigue all fibres demonstrated a marked reduction in sarcoplasmic reticulum Ca2+ release, while myofibrillar Ca2+ sensitivity was either unaltered or increased. In unfatigued fibres, acidosis caused a reduction in myofibrillar Ca2+ sensitivity that was offset by increased tetanic myoplasmic free [Ca2+ ] so that force remained unaffected. Acidification did not affect the fatigue tolerance of fatigue-resistant fibres, whereas uncertainties remain whether or not fatigue-sensitive fibres were affected. Following fatigue, a prolonged force depression at preferentially low-frequency stimulation was evident in fatigue-sensitive fibres and this was caused exclusively by an impaired sarcoplasmic reticulum Ca2+ release. We conclude that impaired sarcoplasmic reticulum Ca2+ release is the predominant mechanism of force depression both in the development of, and recovery from, fatigue in human intercostal muscle.


Subject(s)
Calcium Signaling , Intercostal Muscles/physiopathology , Muscle Fatigue , Muscle Fibers, Skeletal/pathology , Sarcoplasmic Reticulum/pathology , Calcium/physiology , Humans , In Vitro Techniques , Muscle Contraction
20.
J Clin Invest ; 129(11): 4937-4950, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31609246

ABSTRACT

Atrial fibrillation (AF), defined by disorganized atrial cardiac rhythm, is the most prevalent cardiac arrhythmia worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with AF risk, including the cardiogenic transcription factor genes TBX5, GATA4, and NKX2-5. We report that Tbx5 and Gata4 interact with opposite signs for atrial rhythm controls compared with cardiac development. Using mouse genetics, we found that AF pathophysiology caused by Tbx5 haploinsufficiency, including atrial arrhythmia susceptibility, prolonged action potential duration, and ectopic cardiomyocyte depolarizations, were all rescued by Gata4 haploinsufficiency. In contrast, Nkx2-5 haploinsufficiency showed no combinatorial effect. The molecular basis of the TBX5/GATA4 interaction included normalization of intra-cardiomyocyte calcium flux and expression of calcium channel genes Atp2a2 and Ryr2. Furthermore, GATA4 and TBX5 showed antagonistic interactions on an Ryr2 enhancer. Atrial rhythm instability caused by Tbx5 haploinsufficiency was rescued by a decreased dose of phospholamban, a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, consistent with a role for decreased sarcoplasmic reticulum calcium flux in Tbx5-dependent AF susceptibility. This work defines a link between Tbx5 dose, sarcoplasmic reticulum calcium flux, and AF propensity. The unexpected interactions between Tbx5 and Gata4 in atrial rhythm control suggest that evaluating specific interactions between genetic risk loci will be necessary for ascertaining personalized risk from genetic association data.


Subject(s)
Atrial Fibrillation , Calcium Signaling/genetics , Calcium/metabolism , Genetic Loci , Homeostasis/genetics , Sarcoplasmic Reticulum , Transcription Factors , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Genome-Wide Association Study , Heart Atria/metabolism , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Mice , Risk Factors , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...