Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.512
Filter
1.
Sci Rep ; 14(1): 11241, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755246

ABSTRACT

Current density, the membrane current value divided by membrane capacitance (Cm), is widely used in cellular electrophysiology. Comparing current densities obtained in different cell populations assume that Cm and ion current magnitudes are linearly related, however data is scarce about this in cardiomyocytes. Therefore, we statistically analyzed the distributions, and the relationship between parameters of canine cardiac ion currents and Cm, and tested if dividing original parameters with Cm had any effect. Under conventional voltage clamp conditions, correlations were high for IK1, moderate for IKr and ICa,L, while negligible for IKs. Correlation between Ito1 peak amplitude and Cm was negligible when analyzing all cells together, however, the analysis showed high correlations when cells of subepicardial, subendocardial or midmyocardial origin were analyzed separately. In action potential voltage clamp experiments IK1, IKr and ICa,L parameters showed high correlations with Cm. For INCX, INa,late and IKs there were low-to-moderate correlations between Cm and these current parameters. Dividing the original current parameters with Cm reduced both the coefficient of variation, and the deviation from normal distribution. The level of correlation between ion currents and Cm varies depending on the ion current studied. This must be considered when evaluating ion current densities in cardiac cells.


Subject(s)
Action Potentials , Electric Capacitance , Heart Ventricles , Myocytes, Cardiac , Patch-Clamp Techniques , Animals , Dogs , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Heart Ventricles/cytology , Heart Ventricles/metabolism , Action Potentials/physiology , Membrane Potentials/physiology , Ion Channels/metabolism , Cell Membrane/metabolism
2.
Int J Mol Sci ; 25(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38791311

ABSTRACT

Doxorubicin (DOX), widely used as a chemotherapeutic agent for various cancers, is limited in its clinical utility by its cardiotoxic effects. Despite its widespread use, the precise mechanisms underlying DOX-induced cardiotoxicity at the cellular and molecular levels remain unclear, hindering the development of preventive and early detection strategies. To characterize the cytotoxic effects of DOX on isolated ventricular cardiomyocytes, focusing on the expression of specific microRNAs (miRNAs) and their molecular targets associated with endogenous cardioprotective mechanisms such as the ATP-sensitive potassium channel (KATP), Sirtuin 1 (SIRT1), FOXO1, and GSK3ß. We isolated Guinea pig ventricular cardiomyocytes by retrograde perfusion and enzymatic dissociation. We assessed cell morphology, Reactive Oxygen Species (ROS) levels, intracellular calcium, and mitochondrial membrane potential using light microscopy and specific probes. We determined the miRNA expression profile using small RNAseq and validated it using stem-loop qRT-PCR. We quantified mRNA levels of some predicted and validated molecular targets using qRT-PCR and analyzed protein expression using Western blot. Exposure to 10 µM DOX resulted in cardiomyocyte shortening, increased ROS and intracellular calcium levels, mitochondrial membrane potential depolarization, and changes in specific miRNA expression. Additionally, we observed the differential expression of KATP subunits (ABCC9, KCNJ8, and KCNJ11), FOXO1, SIRT1, and GSK3ß molecules associated with endogenous cardioprotective mechanisms. Supported by miRNA gene regulatory networks and functional enrichment analysis, these findings suggest that DOX-induced cardiotoxicity disrupts biological processes associated with cardioprotective mechanisms. Further research must clarify their specific molecular changes in DOX-induced cardiac dysfunction and investigate their diagnostic biomarkers and therapeutic potential.


Subject(s)
Cardiotoxicity , Doxorubicin , MicroRNAs , Myocytes, Cardiac , Reactive Oxygen Species , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Animals , Doxorubicin/adverse effects , Doxorubicin/toxicity , Cardiotoxicity/etiology , MicroRNAs/genetics , MicroRNAs/metabolism , Reactive Oxygen Species/metabolism , Guinea Pigs , Membrane Potential, Mitochondrial/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Male , Calcium/metabolism , Gene Expression Regulation/drug effects
3.
Article in English | MEDLINE | ID: mdl-38723703

ABSTRACT

Mollusks, including snails, possess two chambered hearts. The heart and cardiomyocytes of snails have many similarities with those of mammals. Also, the biophysics and pharmacology of Ca, K, and Na ion channels resemble. Similar to mammals, in mollusks, the ventricular cardiomyocytes and K channels are often studied, which are selectively sensitive to antagonists such as 4-AP, E-4031, and TEA. Since the physiological properties of the ventricular cardiac cells of snails are well characterized, the enzymatically dissociated atrial cardiomyocytes of Cornu aspersum (Müller, 1774) were studied using the whole-cell patch-clamp technique for detailed comparisons with mice, Mus musculus. The incubation of tissues in a solution simultaneously containing two enzymes, collagenase and papain, enabled the isolation of single cells. Recordings in the atrial cardiomyocytes of snails revealed outward K+ currents closely resembling those of the ventricle. The latter was consistent, whether the voltage ramp or steps and long or short pulses were used. Interestingly, under identical conditions, the current waveforms of atrial cardiomyocytes in snails were similar to those of mice left ventricles, albeit the kinetics and the absence of inward rectifier K channel (IK1) activation. Therefore, the heart of mollusks could be used as a simple and accessible experimental model, particularly for pharmacology and toxicology studies.


Subject(s)
Heart Atria , Heart Ventricles , Myocytes, Cardiac , Animals , Heart Atria/drug effects , Heart Atria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Mice , Patch-Clamp Techniques , Potassium Channels/metabolism , Snails
5.
Nature ; 627(8005): 854-864, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480880

ABSTRACT

The heart, which is the first organ to develop, is highly dependent on its form to function1,2. However, how diverse cardiac cell types spatially coordinate to create the complex morphological structures that are crucial for heart function remains unclear. Here we integrated single-cell RNA-sequencing with high-resolution multiplexed error-robust fluorescence in situ hybridization to resolve the identity of the cardiac cell types that develop the human heart. This approach also provided a spatial mapping of individual cells that enables illumination of their organization into cellular communities that form distinct cardiac structures. We discovered that many of these cardiac cell types further specified into subpopulations exclusive to specific communities, which support their specialization according to the cellular ecosystem and anatomical region. In particular, ventricular cardiomyocyte subpopulations displayed an unexpected complex laminar organization across the ventricular wall and formed, with other cell subpopulations, several cellular communities. Interrogating cell-cell interactions within these communities using in vivo conditional genetic mouse models and in vitro human pluripotent stem cell systems revealed multicellular signalling pathways that orchestrate the spatial organization of cardiac cell subpopulations during ventricular wall morphogenesis. These detailed findings into the cellular social interactions and specialization of cardiac cell types constructing and remodelling the human heart offer new insights into structural heart diseases and the engineering of complex multicellular tissues for human heart repair.


Subject(s)
Body Patterning , Heart , Myocardium , Animals , Humans , Mice , Heart/anatomy & histology , Heart/embryology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/embryology , In Situ Hybridization, Fluorescence , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Gene Expression Analysis
6.
Biomolecules ; 13(8)2023 08 10.
Article in English | MEDLINE | ID: mdl-37627297

ABSTRACT

The extent of heavy-metal-induced cardiotoxicity is proportional to the levels of metal bioaccumulation, and it was previously assumed that heavy metals accumulate uniformly in the myocardium. Therefore, the aim of this study was to investigate concentrations of metals and metalloids in two distant regions of the left ventricle (LV), the base of the LV, and apex of the LV using inductively coupled plasma mass spectrometry (ICP-MS). We also examined the potential correlation between metal levels and the thickness of the interventricular septum in twenty LV specimens (ten from the base of LV and ten from the apex of LV) from 10 individuals (mean age 75 ± 6 years). We found significantly higher concentrations of arsenic and lead in the LV apex compared to the base of the LV. We also found a positive correlation between the concentrations of arsenic in the myocardium of LV and the thickness of the interventricular septum. Our results indicate that arsenic and lead accumulate to a higher extent in the apex of the LV compared to the base of the LV. Therefore, future studies designed to measure levels of metals in heart muscle should consider non-uniform accumulation of metals in the myocardium.


Subject(s)
Arsenic , Bioaccumulation , Heart Ventricles , Lead , Aged , Female , Humans , Male , Arsenic/metabolism , Arsenic/pharmacokinetics , Arsenic/toxicity , Autopsy , Cardiotoxicity/metabolism , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Lead/metabolism , Lead/pharmacokinetics , Lead/toxicity , Ventricular Septum/cytology , Ventricular Septum/drug effects , Ventricular Septum/metabolism , Ventricular Septum/pathology , Aging/metabolism
7.
Cells ; 11(2)2022 01 06.
Article in English | MEDLINE | ID: mdl-35053304

ABSTRACT

Intracellular Ca2+ overload secondary to chronic hemodynamic stimuli promotes the recruitment of Ca2+-dependent signaling implicated in cardiomyocyte hypertrophy. The present study tested the hypothesis that sympathetic-mediated hypertrophy of neonatal rat ventricular cardiomyocytes (NRVMs) translated to an increase in calcium influx secondary to the upregulation of CaV1.2 channel subunits. Confocal imaging of norepinephrine (NE)-treated NRVMs revealed a hypertrophic response compared to untreated NRVMs. L-type CaV1.2 peak current density was increased 4-fold following a 24-h stimulation with NE. NE-treated NRVMs exhibited a significant upregulation of CaVα2δ1 and CaVß3 protein levels without significant changes of CaVα1C and CaVß2 protein levels. Pre-treatment with the ß1-blocker metoprolol failed to inhibit hypertrophy or CaVß3 upregulation whereas CaVα2δ1 protein levels were significantly reduced. NE promoted the phosphorylation of ERK 1/2, and the response was attenuated by the ß1-blocker. U0126 pre-treatment suppressed NE-induced ERK1/2 phosphorylation but failed to attenuate hypertrophy. U0126 inhibition of ERK1/2 phosphorylation prevented NE-mediated upregulation of CaVα2δ1, whereas CaVß3 protein levels remained elevated. Thus, ß1-adrenergic receptor-mediated recruitment of the ERK1/2 plays a seminal role in the upregulation of CaVα2δ1 in NRVMs independent of the concomitant hypertrophic response. However, the upregulation of CaVß3 protein levels may be directly dependent on the hypertrophic response of NRVMs.


Subject(s)
Calcium Channels, L-Type/metabolism , Heart Ventricles/cytology , MAP Kinase Signaling System , Myocytes, Cardiac/metabolism , Protein Subunits/metabolism , Receptors, Adrenergic, beta-1/metabolism , Sympathetic Nervous System/metabolism , Up-Regulation , Animals , Animals, Newborn , Calcium/metabolism , Hypertrophy , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/drug effects , Norepinephrine/pharmacology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Up-Regulation/drug effects
8.
Cells ; 11(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-35053351

ABSTRACT

Freshly isolated primary cardiomyocytes (CM) are indispensable for cardiac research. Experimental CM research is generally incompatible with life of the donor animal, while human heart samples are usually small and scarce. CM isolation from animal hearts, traditionally performed by coronary artery perfusion of enzymes, liberates millions of cells from the heart. However, due to progressive cell remodeling following isolation, freshly isolated primary CM need to be used within 4-8 h post-isolation for most functional assays, meaning that the majority of cells is essentially wasted. In addition, coronary perfusion-based isolation cannot easily be applied to human tissue biopsies, and it does not straightforwardly allow for assessment of regional differences in CM function within the same heart. Here, we provide a method of multi-day CM isolation from one animal heart, yielding calcium-tolerant ventricular and atrial CM. This is based on cell isolation from cardiac tissue slices following repeated (usually overnight) storage of the tissue under conditions that prolong CM viability beyond the day of organ excision by two additional days. The maintenance of cells in their near-native microenvironment slows the otherwise rapid structural and functional decline seen in isolated CM during attempts for prolonged storage or culture. Multi-day slice-based CM isolation increases the amount of useful information gained per animal heart, improving reproducibility and reducing the number of experimental animals required in basic cardiac research. It also opens the doors to novel experimental designs, including exploring same-heart regional differences.


Subject(s)
Biomedical Research , Heart Atria/cytology , Heart Ventricles/cytology , Myocytes, Cardiac/cytology , Animals , Calcium/pharmacology , Cell Separation , Cell Shape/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Membrane Potentials/drug effects , Rabbits , Vasoconstriction/drug effects , Vasodilation/drug effects
9.
J Cell Biochem ; 123(1): 128-141, 2022 01.
Article in English | MEDLINE | ID: mdl-34487557

ABSTRACT

The co-chaperone Bcl2-associated athanogene-3 (BAG3) maintains cellular protein quality control through the regulation of heat shock protein 70 (HSP70). Cancer cells manipulate BAG3-HSP70-regulated pathways for tumor initiation and proliferation, which has led to the development of promising small molecule therapies, such as JG-98, which inhibit the BAG3-HSP70 interaction and mitigate tumor growth. However, it is not known how these broad therapies impact cardiomyocytes, where the BAG3-HSP70 complex is a key regulator of protein turnover and contractility. Here, we show that JG-98 exposure is toxic in neonatal rat ventricular myocytes (NRVMs). Using immunofluorescence microscopy to assess cell death, we found that apoptosis increased in NRVMs treated with JG-98 doses as low as 10 nM. JG-98 treatment also reduced autophagy flux and altered expression of BAG3 and several binding partners involved in BAG3-dependent autophagy, including SYNPO2 and HSPB8. We next assessed protein half-life with disruption of the BAG3-HSP70 complex by treating with JG-98 in the presence of cycloheximide and found BAG3, HSPB5, and HSPB8 half-lives were reduced, indicating that complex formation with HSP70 is important for their stability. Next, we assessed sarcomere structure using super-resolution microscopy and found that disrupting the interaction with HSP70 leads to sarcomere structural disintegration. To determine whether the effects of JG-98 could be mitigated by pharmacological autophagy induction, we cotreated NRVMs with rapamycin, which partially reduced the extent of apoptosis and sarcomere disarray. Finally, we investigated whether the effects of JG-98 extended to skeletal myocytes using C2C12 myotubes and found again increased apoptosis and reduced autophagic flux. Together, our data suggest that nonspecific targeting of the BAG3-HSP70 complex to treat cancer may be detrimental for cardiac and skeletal myocytes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/adverse effects , Apoptosis Regulatory Proteins/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , Sarcomeres/drug effects , Sarcomeres/metabolism , Signal Transduction/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Autophagy/drug effects , Cell Line , Cell Survival/drug effects , Heart Ventricles/cytology , Mice , Myoblasts/drug effects , Myoblasts/metabolism , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley
10.
Mol Pharmacol ; 101(1): 13-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34764211

ABSTRACT

Naturally found chrysosplenol-C (4',5,6-trihydroxy-3,3',7-trimethoxyflavone) increases the contractility of cardiac myocytes independent of ß-adrenergic signaling. We investigated the cellular mechanism for chrysosplenol-C-induced positive inotropy. Global and local Ca2+ signals, L-type Ca2+ current (ICa), and contraction were measured from adult rat ventricular myocytes using two-dimensional confocal Ca2+ imaging, the whole-cell patch-clamp technique, and video-edge detection, respectively. Application of chrysosplenol-C reversibly increased Ca2+ transient magnitude with a maximal increase of ∼55% within 2- to 3-minute exposures (EC50 ≅ 21 µM). This chemical did not alter ICa and slightly increased diastolic Ca2+ level. The frequency and size of resting Ca2+ sparks were increased by chrysosplenol-C. Chrysosplenol-C significantly increased sarcoplasmic reticulum (SR) Ca2+ content but not fractional release. Pretreatment of protein kinase C (PKC) inhibitor but not Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor abolished the stimulatory effects of chrysosplenol-C on Ca2+ transients and Ca2+ sparks. Chrysosplenol-C-induced positive inotropy was removed by the inhibition of PKC but not CaMKII or phospholipase C. Western blotting assessment revealed that PKC-δ protein level in the membrane fractions significantly increase within 2 minutes after chrysosplenol-C exposure with a delayed (5-minute) increase in PKC-α levels in insoluble membrane. These results suggest that chrysosplenol-C enhances contractility via PKC (most likely PKC-δ)-dependent enhancement of SR Ca2+ releases in ventricular myocytes. SIGNIFICANCE STATEMENT: Study shows that chrysosplenol-C, a natural flavone showing a positive inotropic effect, increases SR Ca2+ releases on depolarizations and Ca2+ sparks with an increase of SR Ca2+ loading but not L-type Ca2+ current in ventricular myocytes. Chrysosplenol-C-induced enhancement in contraction is eliminated by PKC inhibition, and it is associated with redistributions of PKC to the membrane. These indicate that chrysosplenol-C enhances contraction via PKC-dependent augmentations of SR Ca2+ release and Ca2+ loading during action potentials.


Subject(s)
Calcium/metabolism , Flavonoids/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Dose-Response Relationship, Drug , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/drug effects
11.
Circ Res ; 130(2): 234-248, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34875852

ABSTRACT

BACKGROUND: During the development of heart failure, a fetal cardiac gene program is reactivated and accelerates pathological cardiac remodeling. We previously reported that a transcriptional repressor, NRSF (neuron restrictive silencer factor), suppresses the fetal cardiac gene program, thereby maintaining cardiac integrity. The underlying molecular mechanisms remain to be determined, however. METHODS: We aim to elucidate molecular mechanisms by which NRSF maintains normal cardiac function. We generated cardiac-specific NRSF knockout mice and analyzed cardiac gene expression profiles in those mice and mice cardiac-specifically expressing a dominant-negative NRSF mutant. RESULTS: We found that cardiac expression of Gαo, an inhibitory G protein encoded in humans by GNAO1, is transcriptionally regulated by NRSF and is increased in the ventricles of several mouse models of heart failure. Genetic knockdown of Gnao1 ameliorated the cardiac dysfunction and prolonged survival rates in these mouse heart failure models. Conversely, cardiac-specific overexpression of GNAO1 in mice was sufficient to induce cardiac dysfunction. Mechanistically, we observed that increasing Gαo expression increased surface sarcolemmal L-type Ca2+ channel activity, activated CaMKII (calcium/calmodulin-dependent kinase-II) signaling, and impaired Ca2+ handling in ventricular myocytes, which led to cardiac dysfunction. CONCLUSIONS: These findings shed light on a novel function of Gαo in the regulation of cardiac Ca2+ homeostasis and systolic function and suggest Gαo may be an effective therapeutic target for the treatment of heart failure.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Repressor Proteins/metabolism , Animals , Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Heart Ventricles/cytology , Heart Ventricles/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics
12.
Cells ; 10(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34831329

ABSTRACT

Spontaneous AP (action potential) firing of sinoatrial nodal cells (SANC) is critically dependent on protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent protein phosphorylation, which are required for the generation of spontaneous, diastolic local Ca2+ releases (LCRs). Although phosphoprotein phosphatases (PP) regulate protein phosphorylation, the expression level of PPs and phosphatase inhibitors in SANC and the impact of phosphatase inhibition on the spontaneous LCRs and other players of the oscillatory coupled-clock system is unknown. Here, we show that rabbit SANC express both PP1, PP2A, and endogenous PP inhibitors I-1 (PPI-1), dopamine and cyclic adenosine 3',5'-monophosphate (cAMP)-regulated phosphoprotein (DARPP-32), kinase C-enhanced PP1 inhibitor (KEPI). Application of Calyculin A, (CyA), a PPs inhibitor, to intact, freshly isolated single SANC: (1) significantly increased phospholamban (PLB) phosphorylation (by 2-3-fold) at both CaMKII-dependent Thr17 and PKA-dependent Ser16 sites, in a time and concentration dependent manner; (2) increased ryanodine receptor (RyR) phosphorylation at the Ser2809 site; (3) substantially increased sarcoplasmic reticulum (SR) Ca2+ load; (4) augmented L-type Ca2+ current amplitude; (5) augmented LCR's characteristics and decreased LCR period in intact and permeabilized SANC, and (6) increased the spontaneous basal AP firing rate. In contrast, the selective PP2A inhibitor okadaic acid (100 nmol/L) had no significant effect on spontaneous AP firing, LCR parameters, or PLB phosphorylation. Application of purified PP1 to permeabilized SANC suppressed LCR, whereas purified PP2A had no effect on LCR characteristics. Our numerical model simulations demonstrated that PP inhibition increases AP firing rate via a coupled-clock mechanism, including respective increases in the SR Ca2+ pumping rate, L-type Ca2+ current, and Na+/Ca2+-exchanger current. Thus, PP1 and its endogenous inhibitors modulate the basal spontaneous firing rate of cardiac pacemaker cells by suppressing SR Ca2+ cycling protein phosphorylation, the SR Ca2+ load and LCRs, and L-type Ca2+ current.


Subject(s)
Biological Clocks , Phosphoprotein Phosphatases/metabolism , Sinoatrial Node/cytology , Action Potentials/drug effects , Animals , Biological Clocks/drug effects , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium-Binding Proteins/metabolism , Cell Membrane Permeability/drug effects , Computer Simulation , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/cytology , Marine Toxins/pharmacology , Models, Biological , Oxazoles/pharmacology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits
13.
STAR Protoc ; 2(4): 100950, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34820638

ABSTRACT

Isolation and culture of ventricular cardiomyocytes from neonatal rats (NRVMs) is a powerful model to study neonatal cardiac development, cell cycle regulation, and cardiac physiology and pathology in vitro. Here, we present our modified enzymatic digestion protocol followed by two-step discontinuous Percoll gradient centrifugation to isolate a high yield of viable ventricular cardiomyocytes from neonatal rats. Finally, here we describe an immunostaining protocol for cytosolic and nuclear staining of NRVMs. For complete details on the use and execution of this protocol, please refer to Pereira et al. (2020).


Subject(s)
Cell Culture Techniques/methods , Heart Ventricles/cytology , Immunohistochemistry/methods , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Separation/methods , Cells, Cultured , Rats
14.
Stem Cell Reports ; 16(12): 3036-3049, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34739849

ABSTRACT

A library of well-characterized human induced pluripotent stem cell (hiPSC) lines from clinically healthy human subjects could serve as a useful resource of normal controls for in vitro human development, disease modeling, genotype-phenotype association studies, and drug response evaluation. We report generation and extensive characterization of a gender-balanced, racially/ethnically diverse library of hiPSC lines from 40 clinically healthy human individuals who range in age from 22 to 61 years. The hiPSCs match the karyotype and short tandem repeat identities of their parental fibroblasts, and have a transcription profile characteristic of pluripotent stem cells. We provide whole-genome sequencing data for one hiPSC clone from each individual, genomic ancestry determination, and analysis of mendelian disease genes and risks. We document similar transcriptomic profiles, single-cell RNA-sequencing-derived cell clusters, and physiology of cardiomyocytes differentiated from multiple independent hiPSC lines. This extensive characterization makes this hiPSC library a valuable resource for many studies on human biology.


Subject(s)
Health , Induced Pluripotent Stem Cells/cytology , Adult , Calcium Signaling , Cell Differentiation , Cell Line , Clone Cells , Ethnicity , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Variation , Heart Atria/cytology , Heart Ventricles/cytology , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Risk Factors , Young Adult
15.
Nutrients ; 13(11)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34835942

ABSTRACT

Although insulin-induced cardiac hypertrophy is reported, very little information is available on the hypertrophic effect of insulin on ventricular cardiomyocytes and the regulation of sodium and calcium homeostasis. Taurine is a non-essential amino acid synthesized by cardiomyocytes and the brain and is present in low quantities in many foods, particularly seafood. The purpose of this study was to investigate whether chronic exposure to insulin induces hypertrophy of ventricular cardiomyocytes that are associated with changes in Na+ and Ca2+ homeostasis and whether taurine pre-treatment prevents these effects. Our results showed that chronic treatment with insulin leads to cardiomyocyte hypertrophy that is associated with an increase in basal intracellular Na+ and Ca2+ levels. Furthermore, long-term taurine treatment prevents morphological and ionic remodeling induced by insulin. In addition, blocking the Na+-taurine co-transporter prevented the taurine antihypertrophic effect. Finally, the insulin-induced remodeling of cardiomyocytes was associated with a decrease in the ratio of phospho-CREB (pCREB) to total cAMP response element binding protein (CREB); taurine prevented this effect. In conclusion, our results show that insulin induces ventricular cardiomyocyte hypertrophy via downregulation of the pCREB/tCREB level and that chronic taurine treatment prevents this effect.


Subject(s)
Cardiomegaly/prevention & control , Myocytes, Cardiac/drug effects , Taurine/pharmacology , Animals , Calcium/metabolism , Cardiomegaly/chemically induced , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/drug effects , Heart Ventricles/cytology , Homeostasis , Insulin/metabolism , Male , Rats , Sodium/metabolism , Symporters/metabolism , Ventricular Remodeling/drug effects , beta-Alanine/metabolism
16.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681711

ABSTRACT

Congenital heart disease (CHD) is one of the most common birth defects in humans, present in around 40% of newborns with Down's syndrome (DS). The SH3 domain-binding glutamic acid-rich (SH3BGR) gene, which maps to the DS region, belongs to a gene family encoding a cluster of small thioredoxin-like proteins sharing SH3 domains. Although its expression is confined to the cardiac and skeletal muscle, the physiological role of SH3BGR in the heart is poorly understood. Interestingly, we observed a significant upregulation of SH3BGR in failing hearts of mice and human patients with hypertrophic cardiomyopathy. Along these lines, the overexpression of SH3BGR exhibited a significant increase in the expression of hypertrophic markers (Nppa and Nppb) and increased cell surface area in neonatal rat ventricular cardiomyocytes (NRVCMs), whereas its knockdown attenuated cellular hypertrophy. Mechanistically, using serum response factor (SRF) response element-driven luciferase assays in the presence or the absence of RhoA or its inhibitor, we found that the pro-hypertrophic effects of SH3BGR are mediated via the RhoA-SRF axis. Furthermore, SH3BGR knockdown resulted in the induction of apoptosis and reduced cell viability in NRVCMs via apoptotic Hippo-YAP signaling. Taking these results together, we here show that SH3BGR is vital for maintaining cytoskeletal integrity and cellular viability in NRVCMs through its modulation of the SRF/YAP signaling pathways.


Subject(s)
Apoptosis , Muscle Proteins/genetics , Actinin/metabolism , Animals , Animals, Newborn , Cells, Cultured , Heart Ventricles/cytology , Hippo Signaling Pathway , Muscle Proteins/deficiency , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Serum Response Factor/genetics , Serum Response Factor/metabolism , YAP-Signaling Proteins/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
17.
Oxid Med Cell Longev ; 2021: 1257283, 2021.
Article in English | MEDLINE | ID: mdl-34630844

ABSTRACT

Heart failure (HF) is a clinical syndrome caused by impairment of ventricular filling, ejection of blood, or both and is categorized as HF with reduced ejection fraction (HFrEF) or HF with preserved ejection fraction (HFpEF) based on left ventricular function. Cardiac fibrosis contributes to left ventricular dysfunction and leads to the development of HF. Ivabradine, an If current selective specific inhibitor, has been shown to improve the prognosis of patients with HF. However, the effects of ivabradine on cardiac function and fibrosis in HFpEF and HFrEF and the underlying mechanism remain unclear. In the present study, we utilized mouse models to mimic HFpEF and HFrEF and evaluated the therapeutic effects of ivabradine. By treating mice with different doses (10 mg/kg/d and 20 mg/kg/d) of ivabradine for 4 or 8 weeks, we found that a high dose of ivabradine improved cardiac diastolic function in HFpEF mice and ameliorated cardiac diastolic and systolic function and ventricular tachycardia incidence in HFrEF mice. Moreover, ivabradine significantly reduced the activation of cardiac fibroblasts and myocardial fibrosis in mice. Mechanistically, microRNA-133a, which was upregulated by ivabradine, targeted connective tissue growth factor and collagen 1 in cardiac fibroblasts and might contribute to the protective role of ivabradine. Together, our work utilized mouse models to study HFpEF and HFrEF, demonstrated the protective role of ivabradine in HFpEF and HFrEF, and elucidated the potential underlying mechanism, which provides an effective strategy for related diseases.


Subject(s)
Cardiotonic Agents/administration & dosage , Heart Failure/drug therapy , Heart Failure/metabolism , Ivabradine/administration & dosage , MicroRNAs/metabolism , Stroke Volume/drug effects , Up-Regulation/drug effects , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/metabolism , Animals , Animals, Newborn , Cells, Cultured , Diastole/drug effects , Disease Models, Animal , Fibroblasts/metabolism , Heart Ventricles/cytology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Systole/drug effects , Transfection , Treatment Outcome
18.
J Mater Sci Mater Med ; 32(9): 121, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34499229

ABSTRACT

Cardiovascular diseases are the leading cause of death in the world, cell therapies have been shown to recover cardiac function in animal models. Biomaterials used as scaffolds can solve some of the problems that cell therapies currently have, plasma polymerized pyrrole (PPPy) is a biomaterial that has been shown to promote cell adhesion and survival. The present research aimed to study PPPy nanoparticles (PPPyN) interaction with adult rat ventricular cardiomyocytes (ARVC), to explore whether PPPyN could be employed as a nanoscaffold and develop cardiac microtissues. PPPyN with a mean diameter of 330 nm were obtained, the infrared spectrum showed that some pyrrole rings are fragmented and that some fragments of the ring can be dehydrogenated during plasma synthesis, it also showed the presence of amino groups in the structure of PPPyN. PPPyN had a significant impact on the ARVC´s shape, delaying dedifferentiation, necrosis, and apoptosis processes, moreover, the cardiomyocytes formed cell aggregates up to 1.12 mm2 with some aligned cardiomyocytes and generated fibers on its surface similar to cardiac extracellular matrix. PPPyN served as a scaffold for adult ARVC. Our results indicate that PPPyN-scaffold is a biomaterial that could have potential application in cardiac cell therapy (CCT).


Subject(s)
Myocytes, Cardiac/drug effects , Nanoparticles/chemistry , Pyrroles/pharmacology , Animals , Cell Dedifferentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Male , Materials Testing , Myocytes, Cardiac/physiology , Plasma Gases/pharmacology , Polymerization/drug effects , Pyrroles/chemistry , Rats , Rats, Wistar
19.
Physiol Rep ; 9(17): e15015, 2021 09.
Article in English | MEDLINE | ID: mdl-34514737

ABSTRACT

Compartmentation of cAMP signaling is a critical factor for maintaining the integrity of receptor-specific responses in cardiac myocytes. This phenomenon relies on various factors limiting cAMP diffusion. Our previous work in adult rat ventricular myocytes (ARVMs) indicates that PKA regulatory subunits anchored to the outer membrane of mitochondria play a key role in buffering the movement of cytosolic cAMP. PKA can be targeted to discrete subcellular locations through the interaction of both type I and type II regulatory subunits with A-kinase anchoring proteins (AKAPs). The purpose of this study is to identify which AKAPs and PKA regulatory subunit isoforms are associated with mitochondria in ARVMs. Quantitative PCR data demonstrate that mRNA for dual specific AKAP1 and 2 (D-AKAP1 & D-AKAP2), acyl-CoA-binding domain-containing 3 (ACBD3), optic atrophy 1 (OPA1) are most abundant, while Rab32, WAVE-1, and sphingosine kinase type 1 interacting protein (SPHKAP) were barely detectable. Biochemical and immunocytochemical analysis suggests that D-AKAP1, D-AKAP2, and ACBD3 are the predominant mitochondrial AKAPs exposed to the cytosolic compartment in these cells. Furthermore, we show that both type I and type II regulatory subunits of PKA are associated with mitochondria. Taken together, these data suggest that D-AKAP1, D-AKAP2, and ACBD3 may be responsible for tethering both type I and type II PKA regulatory subunits to the outer mitochondrial membrane in ARVMs. In addition to regulating PKA-dependent mitochondrial function, these AKAPs may play an important role by buffering the movement of cAMP necessary for compartmentation.


Subject(s)
A Kinase Anchor Proteins/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Heart Ventricles/enzymology , Mitochondria/enzymology , Myocytes, Cardiac/enzymology , Animals , Cells, Cultured , Heart Ventricles/cytology , Male , Rats , Rats, Sprague-Dawley
20.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166237, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34339838

ABSTRACT

R-loops are naturally occurring transcriptional intermediates containing RNA/DNA hybrids. Excessive R-loops cause genomic instability, DNA damage, and replication stress. Senataxin-associated exonuclease (San1) is a protein that interacts with Senataxin (SETX), a helicase resolving R-loops. It remains unknown if R-loops-induced DNA damage plays a role in the heart, especially in the proliferative neonatal cardiomyocytes (CMs). San1-/- mice were generated using the CRISPR/Cas9 technique. The newborn San1-/- mice show no overt phenotype, but their hearts were smaller with larger, yet fewer CMs. CM proliferation was impaired with reduced cell cycle-related transcripts and proteins. S9.6 staining revealed that excessive R-loops accumulated in the nucleus of neonatal San1-/- CMs. Increased γH2AX staining on newborn and adult heart sections exhibited increased DNA damage. Similarly, San1-/- AC16-cardiomyocytes showed cumulative R-loops and DNA damage, leading to the activation of cell cycle checkpoint kinase ATR and PARP1 hyperactivity, arresting G2/M cell-cycle and CM proliferation. Together, the present study uncovers an essential role of San1 in resolving excessive R-loops that lead to DNA damage and repressing CM proliferation, providing new insights into a novel biological function of San1 in the neonatal heart. San1 may serve as a novel therapeutic target for the treatment of hypoplastic cardiac disorders.


Subject(s)
Cardiomyopathies/genetics , Exodeoxyribonucleases/deficiency , Heart Failure/genetics , Heart Ventricles/pathology , Trans-Activators/deficiency , Animals , Cardiomyopathies/complications , Cardiomyopathies/pathology , Cell Line , DNA Damage , Disease Models, Animal , Exodeoxyribonucleases/genetics , Gene Knockout Techniques , Heart Failure/pathology , Heart Ventricles/cytology , Humans , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Primary Cell Culture , R-Loop Structures , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...