Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Biol Chem ; : 107470, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38879012

ABSTRACT

Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodelling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the ß-adrenergic pathways. Using fluorescence resonance energy transfer-based assays we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.

2.
Nat Commun ; 14(1): 1722, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37012244

ABSTRACT

Cardiogenesis relies on the precise spatiotemporal coordination of multiple progenitor populations. Understanding the specification and differentiation of these distinct progenitor pools during human embryonic development is crucial for advancing our knowledge of congenital cardiac malformations and designing new regenerative therapies. By combining genetic labelling, single-cell transcriptomics, and ex vivo human-mouse embryonic chimeras we uncovered that modulation of retinoic acid signaling instructs human pluripotent stem cells to form heart field-specific progenitors with distinct fate potentials. In addition to the classical first and second heart fields, we observed the appearance of juxta-cardiac field progenitors giving rise to both myocardial and epicardial cells. Applying these findings to stem-cell based disease modelling we identified specific transcriptional dysregulation in first and second heart field progenitors derived from stem cells of patients with hypoplastic left heart syndrome. This highlights the suitability of our in vitro differentiation platform for studying human cardiac development and disease.


Subject(s)
Pluripotent Stem Cells , Tretinoin , Humans , Animals , Mice , Tretinoin/pharmacology , Heart , Myocardium , Cell Differentiation , Myocytes, Cardiac
3.
Neuromuscul Disord ; 32(11-12): 908-922, 2022 12.
Article in English | MEDLINE | ID: mdl-36418198

ABSTRACT

Mutations in the dystrophin gene cause the most common and currently incurable Duchenne muscular dystrophy (DMD) characterized by progressive muscle wasting. Although abnormal Ca2+ handling is a pathological feature of DMD, mechanisms underlying defective Ca2+ homeostasis remain unclear. Here we generate a novel DMD patient-derived pluripotent stem cell (PSC) model of skeletal muscle with an isogenic control using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated precise gene correction. Transcriptome analysis identifies dysregulated gene sets in the absence of dystrophin, including genes involved in Ca2+ handling, excitation-contraction coupling and muscle contraction. Specifically, analysis of intracellular Ca2+ transients and mathematical modeling of Ca2+ dynamics reveal significantly reduced cytosolic Ca2+ clearance rates in DMD-PSC derived myotubes. Pharmacological assays demonstrate Ca2+ flux in myotubes is determined by both intracellular and extracellular sources. DMD-PSC derived myotubes display significantly reduced velocity of contractility. Compared with a non-isogenic wildtype PSC line, these pathophysiological defects could be rescued by CRISPR-mediated precise gene correction. Our study provides new insights into abnormal Ca2+ homeostasis in DMD and suggests that Ca2+ signaling pathways amenable to pharmacological modulation are potential therapeutic targets. Importantly, we have established a human physiology-relevant in vitro model enabling rapid pre-clinical testing of potential therapies for DMD.


Subject(s)
Muscular Dystrophy, Duchenne , Pluripotent Stem Cells , Humans , Dystrophin/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Cas Systems , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology
4.
Physiol Genomics ; 53(4): 150-159, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33719582

ABSTRACT

We have assessed the role of ric-b8 in the control of heart rate after the gene was implicated in a recent genome-wide association study of resting heart rate. We developed a novel murine model in which it was possible to conditionally delete ric-8b in the sinoatrial (SA) node after the addition of tamoxifen. Despite this, we were unable to obtain homozygotes and thus studied heterozygotes. Haploinsufficiency of ric-8b in the sinoatrial node induced by the addition of tamoxifen in adult animals leads to mice with a reduced heart rate. However, other electrocardiographic intervals (e.g., PR and QRS) were normal, and there was no apparent arrhythmia such as heart block. The positive chronotropic response to isoprenaline was abrogated, whereas the response to carbachol was unchanged. The pacemaker current If (funny current) has an important role in regulating heart rate, and its function is modulated by both isoprenaline and carbachol. Using a heterologous system expressing HCN4, we show that ric-8b can modulate the HCN4 current. Overexpression of ric-8b led to larger HCN4 currents, whereas silencing ric-8b led to smaller currents. Ric-8b modulates heart rate responses in vivo likely via its actions on the stimulatory G-protein.


Subject(s)
Guanine Nucleotide Exchange Factors , Heart Rate , Animals , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/genetics , Mice
5.
Bio Protoc ; 10(1): e3477, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-33654710

ABSTRACT

The cardiac conduction system allows the synchronized propagation of electrical activity through heart muscle. This is initiated by the spontaneous activity of the specialized pacemaker cells of the sino-atrial node (SAN). The SAN region underlies automaticity in mammals and therefore has a crucial role in the pathogenesis of cardiac disorders such as arrhythmia. Isolation of SAN tissue and SAN cells is critical to advance our understanding of SAN structure and function in health and disease. Initially, isolation of SAN tissue and SAN cells was carried out in the rabbit owing to its larger size and similar electrical properties to human. This protocol was optimized by Mangoni and Nargeot (2001) for use in mice to take advantage of advancements in transgenic models. Here, we provide a step-by-step guide to dissecting the SAN tissue and isolating pacemaker cardiomyocytes from mouse hearts using an enzyme digestion approach.

6.
Bio Protoc ; 10(1): e3478, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-33654711

ABSTRACT

Cardiac pacemaker cells of the sino-atrial node are responsible for the initiation of the heart beat and express an array of ion channels. The patch-clamp technique is the gold standard method for investigating the function of ion channels expressed in electrically active cells. Conventional whole-cell and perforated patch-clamp techniques can be used to investigate ionic currents in the voltage-clamp mode and changes in membrane potential (e.g., action potential) in the current-clamp mode. Here, we provide details of protocols used to measure spontaneous and triggered action potentials and whole-cell funny current If (HCN4) in single cardiomyocytes isolated from the mouse sino-atrial node (SAN).

7.
Physiol Genomics ; 51(8): 323-332, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31172864

ABSTRACT

Atrial fibrillation is a significant worldwide contributor to cardiovascular morbidity and mortality. Few studies have investigated the differences in gene expression between the left and right atrial appendages, leaving their characterization largely unexplored. In this study, differential gene expression was investigated in atrial fibrillation and sinus rhythm using left and right atrial appendages from the same patients. RNA sequencing was performed on the left and right atrial appendages from five sinus rhythm (SR) control patients and five permanent AF case patients. Differential gene expression in both the left and right atrial appendages was analyzed using the Bioconductor package edgeR. A selection of differentially expressed genes, with relevance to atrial fibrillation, were further validated using quantitative RT-PCR. The distribution of the samples assessed through principal component analysis showed distinct grouping between left and right atrial appendages and between SR controls and AF cases. Overall 157 differentially expressed genes were identified to be downregulated and 90 genes upregulated in AF. Pathway enrichment analysis indicated a greater involvement of left atrial genes in the Wnt signaling pathway whereas right atrial genes were involved in clathrin-coated vesicle and collagen formation. The differing expression of genes in both left and right atrial appendages indicate that there are different mechanisms for development, support and remodeling of AF within the left and right atria.


Subject(s)
Atrial Appendage/physiopathology , Atrial Fibrillation/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Aged , Aged, 80 and over , Atrial Fibrillation/pathology , Clathrin-Coated Vesicles/metabolism , Cohort Studies , Collagen/metabolism , Coronary Artery Bypass , Down-Regulation/genetics , Female , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Up-Regulation/genetics , Wnt Signaling Pathway/genetics
9.
Am J Physiol Cell Physiol ; 314(5): C616-C626, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29342363

ABSTRACT

G protein-gated inwardly rectifying K+ (GIRK) channels are the major inwardly rectifying K+ currents in cardiac atrial myocytes and an important determinant of atrial electrophysiology. Inhibitory G protein α-subunits can both mediate activation via acetylcholine but can also suppress basal currents in the absence of agonist. We studied this phenomenon using whole cell patch clamping in murine atria from mice with global genetic deletion of Gαi2, combined deletion of Gαi1/Gαi3, and littermate controls. We found that mice with deletion of Gαi2 had increased basal and agonist-activated currents, particularly in the right atria while in contrast those with Gαi1/Gαi3 deletion had reduced currents. Mice with global genetic deletion of Gαi2 had decreased action potential duration. Tissue preparations of the left atria studied with a multielectrode array from Gαi2 knockout mice showed a shorter effective refractory period, with no change in conduction velocity, than littermate controls. Transcriptional studies revealed increased expression of GIRK channel subunit genes in Gαi2 knockout mice. Thus different G protein isoforms have differential effects on GIRK channel behavior and paradoxically Gαi2 act to increase basal and agonist-activated GIRK currents. Deletion of Gαi2 is potentially proarrhythmic in the atria.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Heart Atria/metabolism , Ion Channel Gating , Potassium/metabolism , Action Potentials , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Atrial Function, Left , Atrial Function, Right , Female , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits/deficiency , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Rate , Kinetics , Male , Mice, 129 Strain , Mice, Knockout , Refractory Period, Electrophysiological
10.
Stem Cell Res ; 25: 233-244, 2017 12.
Article in English | MEDLINE | ID: mdl-29172153

ABSTRACT

The class Ia anti-arrhythmic drug ajmaline is used clinically to unmask latent type I ECG in Brugada syndrome (BrS) patients, although its mode of action is poorly characterised. Our aims were to identify ajmaline's mode of action in human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), and establish a simple BrS hiPSC platform to test whether differences in ajmaline response could be determined between BrS patients and controls. Control hiPSCs were differentiated into spontaneously contracting cardiac clusters. It was found using multi electrode array (MEA) that ajmaline treatment significantly lengthened cluster activation-recovery interval. Patch clamping of single CMs isolated from clusters revealed that ajmaline can block both INa and IKr. Following generation of hiPSC lines from BrS patients (absent of pathogenic SCN5A sodium channel mutations), analysis of hiPSC-CMs from patients and controls revealed that differentiation and action potential parameters were similar. Comparison of cardiac clusters by MEA showed that ajmaline lengthened activation-recovery interval consistently across all lines. We conclude that ajmaline can block both depolarisation and repolarisation of hiPSC-CMs at the cellular level, but that a more refined integrated tissue model may be necessary to elicit differences in its effect between BrS patients and controls.


Subject(s)
Ajmaline/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Brugada Syndrome/drug therapy , Heart/drug effects , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Adult , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Brugada Syndrome/physiopathology , Cell Differentiation/drug effects , Heart/physiopathology , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
11.
Mol Cell Endocrinol ; 447: 12-22, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28232089

ABSTRACT

GnRH neurons are fundamental for reproduction in all vertebrates, integrating all reproductive inputs. The inaccessibility of human GnRH-neurons has been a major impediment to studying the central control of reproduction and its disorders. Here, we report the efficient generation of kisspeptin responsive GnRH-secreting neurons by directed differentiation of human Embryonic Stem Cells and induced-Pluripotent Stem Cells derived from a Kallman Syndrome patient and a healthy family member. The protocol involves the generation of intermediate Neural Progenitor Cells (NPCs) through long-term Bone morphogenetic protein 4 inhibition, followed by terminal specification of these NPCs in media containing Fibroblast Growth Factor 8 and a NOTCH inhibitor. The resulting GnRH-expressing and -secreting neurons display a neuroendocrine gene expression pattern and present spontaneous calcium transients that can be stimulated by kisspeptin. These in vitro generated GnRH expressing cells provide a new resource for studying the molecular mechanisms underlying the development and function of GnRH neurons.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Calcium/metabolism , Cell Differentiation , Cell Line , Gene Expression Profiling , Gene Expression Regulation , Human Embryonic Stem Cells/metabolism , Humans , Neural Stem Cells/metabolism
12.
Pharmacol Res ; 105: 54-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26773203

ABSTRACT

Atrial fibrillation is the commonest cardiac arrhythmia and leads to significant clinical morbidity and mortality. It has a complex pathophysiology but is often initiated by atrial ectopic beats and because of atrial remodelling once it occurs it can become established. Thus therapeutic interventions designed to prevent the initial occurrence of the arrhythmia are particularly needed. At the cellular level, these ectopic beats arise because of abnormal calcium release events from the sarcoplasmic reticulum leading to an inward current mediated by the sodium-calcium exchanger. There has been considerable interest in this over the last few years largely focused on the ryanodine receptor and related signalling pathways. However, atrial myocytes also possess a well-developed inositol trisphosphate (IP3) dependent calcium release system and this has been less studied. In this review we focus on pathways and molecules that couple via the Gq\11 family of G-proteins including regulators of G-protein signalling that may influence IP3 mediated calcium release and atrial fibrillation.


Subject(s)
Atrial Fibrillation/metabolism , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Inositol Phosphates/metabolism , Signal Transduction , Animals , Atrial Fibrillation/drug therapy , Atrial Fibrillation/pathology , Drug Discovery , Heart Atria/drug effects , Heart Atria/metabolism , Heart Atria/pathology , Humans , Molecular Targeted Therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects
13.
Org Biomol Chem ; 14(2): 455-459, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26603469

ABSTRACT

Described in this work is a novel method for photochemically manipulating peptides and proteins via the installation of cysteine-selective photoactive tags. Thiomaleimides, generated simply by the addition of bromomaleimides to reduced disulfide bonds, undergo [2 + 2] photocycloadditions to reconnect the crosslink between the two cysteine residues. This methodology is demonstrated to enable photoactivation of a peptide by macrocyclisation, and reconnection of the heavy and light chains in an antibody fragment to form thiol stable conjugates. Finally we report on an intriguing thiomaleimide mediated photochemical decarboxylation of C-terminal cysteines, discovered during this study.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Maleimides/chemistry , Cyclization , Decarboxylation , Maleimides/chemical synthesis , Molecular Structure , Photochemical Processes
14.
Org Biomol Chem ; 13(37): 9559-63, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26299430

ABSTRACT

We report a concise approach to a multimodal imaging reagent for peptide labelling via disulfide bridging. The reagent is constructed using a one pot, three component, [3 + 2] cycloaddition of a fluorescent azide with a dithiomaleimide-alkyne, with concomitant incorporation of (125)I. The dithiomaleimide handle then enables site selective conjugation to a disulfide bond of a peptide whilst retaining the key structural bridging functionality, as exemplified on the therapeutic peptide octreotide.


Subject(s)
Disulfides/chemistry , Nuclear Medicine/methods , Optical Imaging/methods , Peptides/chemistry , Cycloaddition Reaction , HEK293 Cells , Humans , Multimodal Imaging , Staining and Labeling
15.
J Biol Chem ; 290(31): 19233-44, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26088132

ABSTRACT

The description of potential molecular substrates for predisposition to atrial fibrillation (AF) is incomplete, and it is unknown what role regulators of G-protein signaling might play. We address whether the attenuation of RGS4 function may promote AF and the mechanism through which this occurs. For this purpose, we studied a mouse with global genetic deletion of RGS4 (RGS4(-/-)) and the normal littermate controls (RGS4(+/+)). In vivo electrophysiology using atrial burst pacing revealed that mice with global RGS4 deletion developed AF more frequently than control littermates. Isolated atrial cells from RGS4(-/-) mice show an increase in Ca(2+) spark frequency under basal conditions and after the addition of endothelin-1 and abnormal spontaneous Ca(2+) release events after field stimulation. Isolated left atria studied on a multielectrode array revealed modest changes in path length for re-entry but abnormal electrical events after a pacing train in RGS4(-/-) mice. RGS4 deletion results in a predisposition to atrial fibrillation from enhanced activity in the Gαq/11-IP3 pathway, resulting in abnormal Ca(2+) release and corresponding electrical events.


Subject(s)
Atrial Fibrillation/genetics , Calcium/metabolism , RGS Proteins/genetics , Action Potentials , Animals , Atrial Fibrillation/metabolism , Calcium Signaling , Electric Stimulation , Genetic Predisposition to Disease , Heart Atria/metabolism , Heart Atria/pathology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/physiology , RGS Proteins/metabolism
16.
Eur Heart J ; 33(15): 1942-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22240500

ABSTRACT

AIMS: Anecdotal observations suggest that sub-clinical electrophysiological manifestations of arrhythmogenic right ventricular cardiomyopathy (ARVC) develop before detectable structural changes ensue on cardiac imaging. To test this hypothesis, we investigated a murine model with conditional cardiac genetic deletion of one desmoplakin allele (DSP ±) and compared the findings to patients with non-diagnostic features of ARVC who carried mutations in desmoplakin. METHODS AND RESULTS: Murine: the DSP (±) mice underwent electrophysiological, echocardiographic, and immunohistochemical studies. They had normal echocardiograms but delayed conduction and inducible ventricular tachycardia associated with mislocalization and reduced intercalated disc expression of Cx43. Sodium current density and myocardial histology were normal at 2 months of age. Human: ten patients with heterozygous mutations in DSP without overt structural heart disease (DSP+) and 12 controls with supraventricular tachycardia were studied by high-density electrophysiological mapping of the right ventricle. Using a standard S(1)-S(2) protocol, restitution curves of local conduction and repolarization parameters were constructed. Significantly greater mean increases in delay were identified particularly in the outflow tract vs. controls (P< 0.01) coupled with more uniform wavefront progression. The odds of a segment with a maximal activation-repolarization interval restitution slope >1 was 99% higher (95% CI: 13%; 351%, P = 0.017) in DSP+ vs. controls. Immunostaining revealed Cx43 mislocalization and variable Na channel distribution. CONCLUSION: Desmoplakin disease causes connexin mislocalization in the mouse and man preceding any overt histological abnormalities resulting in significant alterations in conduction-repolarization kinetics prior to morphological changes detectable on conventional cardiac imaging. Haploinsufficiency of desmoplakin is sufficient to cause significant Cx43 mislocalization. Changes in sodium current density and histological abnormalities may contribute to a worsening phenotype or disease but are not necessary to generate an arrhythmogenic substrate. This has important implications for the earlier diagnosis of ARVC and risk stratification.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Desmoplakins/genetics , Mutation/genetics , Adult , Aged , Animals , Case-Control Studies , Desmoplakins/deficiency , Electrocardiography , Female , Gene Deletion , Heart Conduction System/physiology , Heterozygote , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/physiology , Young Adult
17.
Bioconjug Chem ; 22(2): 132-6, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21271715

ABSTRACT

The introduction of non-natural entities into proteins by chemical modification has numerous applications in fundamental biological science and for the development and manipulation of peptide and protein therapeutics. The reduction of native disulfide bonds provides a convenient method to access two nucleophilic cysteine residues that can serve as ideal attachment points for such chemical modification. The optimum bioconjugation strategy utilizing these cysteine residues should include the reconstruction of a bridge to mimic the role of the disulfide bond, maintaining structure and stability of the protein. Furthermore, the bridging chemical modification should be as rapid as possible to prevent problems associated with protein unfolding, aggregation, or disulfide scrambling. This study reports on an in situ disulfide reduction-bridging strategy that ensures rapid sequestration of the free cysteine residues in a bridge, using dithiomaleimides. This approach is then used to PEGylate the peptide hormone somatostatin and retention of biological activity is demonstrated.


Subject(s)
Disulfides/chemistry , Maleimides/chemistry , Polyethylene Glycols/chemistry , Somatostatin/chemistry , Cell Line , Humans , Molecular Structure , Polyethylene Glycols/chemical synthesis , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/chemical synthesis
18.
Stem Cell Rev Rep ; 7(2): 364-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21120638

ABSTRACT

Cell therapy has developed as a complementary treatment for myocardial regeneration. While both autologous and allogeneic uses have been advocated, the ideal candidate has not been identified yet. Amniotic fluid-derived stem (AFS) cells are potentially a promising resource for cell therapy and tissue engineering of myocardial injuries. However, no information is available regarding their use in an allogeneic context. c-kit-sorted, GFP-positive rat AFS (GFP-rAFS) cells and neonatal rat cardiomyocytes (rCMs) were characterized by cytocentrifugation and flow cytometry for the expression of mesenchymal, embryonic and cell lineage-specific antigens. The activation of the myocardial gene program in GFP-rAFS cells was induced by co-culture with rCMs. The stem cell differentiation was evaluated using immunofluorescence, RT-PCR and single cell electrophysiology. The in vivo potential of Endorem-labeled GFP-rAFS cells for myocardial repair was studied by transplantation in the heart of animals with ischemia/reperfusion injury (I/R), monitored by magnetic resonance imaging (MRI). Three weeks after injection a small number of GFP-rAFS cells acquired an endothelial or smooth muscle phenotype and to a lesser extent CMs. Despite the low GFP-rAFS cells count in the heart, there was still an improvement of ejection fraction as measured by MRI. rAFS cells have the in vitro propensity to acquire a cardiomyogenic phenotype and to preserve cardiac function, even if their potential may be limited by poor survival in an allogeneic setting.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Myocytes, Cardiac/cytology , Stem Cells/cytology , Animals , Antigens, Differentiation/metabolism , Cell Separation/methods , Cell Transdifferentiation , Coculture Techniques , Disease Models, Animal , Female , Flow Cytometry , Graft Rejection , Green Fluorescent Proteins/metabolism , Myocardial Reperfusion Injury/therapy , Myocardium/pathology , Myocytes, Cardiac/physiology , Myosin Heavy Chains/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis , Stem Cell Transplantation , Stem Cells/physiology , Transplantation, Homologous , Troponin I/metabolism
19.
J Biol Chem ; 285(36): 27817-26, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20562107

ABSTRACT

In atrial and nodal cardiac myocytes, M2 muscarinic receptors activate inhibitory G-proteins (G(i/o)), which in turn stimulate G-protein-gated inwardly rectifying K(+) channels through direct binding of the Gbetagamma subunit. Despite also releasing Gbetagamma, G(s)-coupled receptors such as the beta-adrenergic receptor are not able to prominently activate this current. An appealing hypothesis would be if components were sequestered in membrane domains such as caveolae/rafts. Using biochemical fractionation followed by Western blotting and/or radioligand binding experiments, we examined the distribution of the components in stable HEK293 and HL-1 cells, which natively express the transduction cascade. The channel, M2 muscarinic, and A1 adenosine receptors were located in noncaveolar/nonraft fractions. G(i)alpha(1/2) was enriched in both caveolar/raft and noncaveolar/nonraft fractions. In contrast, G(s)alpha was only enriched in caveolar/raft fractions. We constructed YFP-tagged caveolin-2 (YFP-Cav2) and chimeras with the M2 (M2-YFP-Cav2) and A1 (A1-YFP-Cav2) receptors. Analysis of gradient fractions showed that these receptor chimeras were now localized to caveolae-enriched fractions. Microscopy showed that M2-YFP and A1-YFP had a diffuse homogenous membrane signal. YFP-Cav2, M2-YFP-Cav2, and A1-YFP-Cav2 revealed a more punctuate pattern. Finally, we looked at the consequences for signaling. Activation via M2-YFP-Cav2 or A1-YFP-Cav2 revealed substantially slower kinetics compared with M2-YFP or A1-YFP and was reversed by the addition of methyl-beta-cyclodextrin. Thus the localization of the channel signal transduction cascade in non-cholesterol rich domains substantially enhances the speed of signaling. The presence of G(s)alpha solely in caveolae may account for signaling selectivity between G(i/o) and G(s)-coupled receptors.


Subject(s)
Caveolae/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Caveolin 2/metabolism , Cell Line , Cholesterol/metabolism , Humans , Membrane Microdomains/metabolism , Mice , Protein Subunits/metabolism , Signal Transduction , Solubility , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism
20.
Circ Arrhythm Electrophysiol ; 3(4): 391-400, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20495013

ABSTRACT

BACKGROUND: We explored the role that inhibitory heterotrimeric G-proteins play in ventricular arrhythmia. METHODS AND RESULTS: Mice with global genetic deletion of Galpha(i2) [Galpha(i2) (-/-)] were studied and found, based on telemetry, to have a prolonged QT interval on surface ECG when awake. In vivo electrophysiology studies revealed that the Galpha(i2) (-/-) mice have a reduced ventricular effective refractory period and a predisposition to ventricular tachycardia when challenged with programmed electrical stimulation. Neither control nor combined global deletion of Galpha(i1) and Galpha(i3) mice showed these abnormalities. There was no evidence for structural heart disease at this time point in the Galpha(i2) (-/-) mice as assessed by cardiac histology and echocardiography. The absence of Galpha(i2) thus leads to a primary electrical abnormality, and we explored the basis for this finding. With patch clamping, single isolated ventricular cells showed that Galpha(i2) (-/-) mice had a prolonged ventricular action potential duration (APD) but steeper action potential shortening as the diastolic interval was reduced in restitution studies. Gene expression studies showed increased expression of L-type Ca(2+) channel subunits, and patch clamping revealed an increase in these currents in Galpha(i2) (-/-) mice. There were no changes in K(+) currents. CONCLUSIONS: The absence of inhibitory G-protein signaling mediated through Galpha(i2) is a substrate for ventricular arrhythmias.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/deficiency , Heart Conduction System/metabolism , Tachycardia, Ventricular/metabolism , Action Potentials , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Cardiac Pacing, Artificial , Disease Models, Animal , Echocardiography, Doppler, Pulsed , Electrocardiography, Ambulatory , GTP-Binding Protein alpha Subunit, Gi2/genetics , Heart Conduction System/physiopathology , Mice , Mice, Knockout , Patch-Clamp Techniques , Potassium/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Telemetry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...