Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Mol Cell Cardiol ; 168: 3-12, 2022 07.
Article in English | MEDLINE | ID: mdl-35390437

ABSTRACT

Engineered heart muscle (EHM) can be implanted epicardially to remuscularize the failing heart. In case of a severely scarred ventricle, excision of scar followed by transmural heart wall replacement may be a more desirable application. Accordingly, we tested the hypothesis that allograft (rat) and xenograft (human) EHM can also be administered as transmural heart wall replacement in a heterotopic, volume-loaded heart transplantation model. We first established a novel rat model model to test surgical transmural left heart wall repair. Subsequently and in continuation of our previous allograft studies, we tested outcome after implantation of contractile engineered heart muscle (EHM) and non-contractile engineered connective tissue (ECT) as well as engineered mesenchymal tissue (EMT) allografts as transmural heart wall replacement. Finally, proof-of-concept for the application of human EHM was obtained in an athymic nude rat model. Only in case of EHM implantation, remuscularization of the surgically created transmural defect was observed with palpable graft vascularization. Taken together, feasibility of transmural heart repair using bioengineered myocardial grafts could be demonstrated in a novel rat model of heterotopic heart transplantation.


Subject(s)
Heart Transplantation , Myocytes, Cardiac , Animals , Humans , Myocardium , Myocytes, Cardiac/physiology , Rats , Rats, Nude , Tissue Engineering
2.
Basic Res Cardiol ; 117(1): 13, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260914

ABSTRACT

Cancer therapies with anthracyclines have been shown to induce cardiovascular complications. The aims of this study were to establish an in vitro induced pluripotent stem cell model (iPSC) of anthracycline-induced cardiotoxicity (ACT) from patients with an aggressive form of B-cell lymphoma and to examine whether doxorubicin (DOX)-treated ACT-iPSC cardiomyocytes (CM) can recapitulate the clinical features exhibited by patients, and thus help uncover a DOX-dependent pathomechanism. ACT-iPSC CM generated from individuals with CD20+ B-cell lymphoma who had received high doses of DOX and suffered cardiac dysfunction were studied and compared to control-iPSC CM from cancer survivors without cardiac symptoms. In cellular studies, ACT-iPSC CM were persistently more susceptible to DOX toxicity including augmented disorganized myofilament structure, changed mitochondrial shape, and increased apoptotic events. Consistently, ACT-iPSC CM and cardiac fibroblasts isolated from fibrotic human ACT myocardium exhibited higher DOX-dependent reactive oxygen species. In functional studies, Ca2+ transient amplitude of ACT-iPSC CM was reduced compared to control cells, and diastolic sarcoplasmic reticulum Ca2+ leak was DOX-dependently increased. This could be explained by overactive CaMKIIδ in ACT CM. Together with DOX-dependent augmented proarrhythmic cellular triggers and prolonged action potentials in ACT CM, this suggests a cellular link to arrhythmogenic events and contractile dysfunction especially found in ACT engineered human myocardium. CamKIIδ inhibition prevented proarrhythmic triggers in ACT. In contrast, control CM upregulated SERCA2a expression in a DOX-dependent manner, possibly to avoid heart failure conditions. In conclusion, we developed the first human patient-specific stem cell model of DOX-induced cardiac dysfunction from patients with B-cell lymphoma. Our results suggest that DOX-induced stress resulted in arrhythmogenic events associated with contractile dysfunction and finally in heart failure after persistent stress activation in ACT patients.


Subject(s)
Heart Diseases , Heart Failure , Induced Pluripotent Stem Cells , Lymphoma, B-Cell , Neoplasms , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Doxorubicin/metabolism , Doxorubicin/toxicity , Heart Diseases/metabolism , Heart Failure/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Myocytes, Cardiac/metabolism , Neoplasms/metabolism
3.
Circulation ; 142(11): 1059-1076, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32623905

ABSTRACT

BACKGROUND: Noonan syndrome (NS) is a multisystemic developmental disorder characterized by common, clinically variable symptoms, such as typical facial dysmorphisms, short stature, developmental delay, intellectual disability as well as cardiac hypertrophy. The underlying mechanism is a gain-of-function of the RAS-mitogen-activated protein kinase signaling pathway. However, our understanding of the pathophysiological alterations and mechanisms, especially of the associated cardiomyopathy, remains limited and effective therapeutic options are lacking. METHODS: Here, we present a family with two siblings displaying an autosomal recessive form of NS with massive hypertrophic cardiomyopathy as clinically the most prevalent symptom caused by biallelic mutations within the leucine zipper-like transcription regulator 1 (LZTR1). We generated induced pluripotent stem cell-derived cardiomyocytes of the affected siblings and investigated the patient-specific cardiomyocytes on the molecular and functional level. RESULTS: Patients' induced pluripotent stem cell-derived cardiomyocytes recapitulated the hypertrophic phenotype and uncovered a so-far-not-described causal link between LZTR1 dysfunction, RAS-mitogen-activated protein kinase signaling hyperactivity, hypertrophic gene response and cellular hypertrophy. Calcium channel blockade and MEK inhibition could prevent some of the disease characteristics, providing a molecular underpinning for the clinical use of these drugs in patients with NS, but might not be a sustainable therapeutic option. In a proof-of-concept approach, we explored a clinically translatable intronic CRISPR (clustered regularly interspaced short palindromic repeats) repair and demonstrated a rescue of the hypertrophic phenotype. CONCLUSIONS: Our study revealed the human cardiac pathogenesis in patient-specific induced pluripotent stem cell-derived cardiomyocytes from NS patients carrying biallelic variants in LZTR1 and identified a unique disease-specific proteome signature. In addition, we identified the intronic CRISPR repair as a personalized and in our view clinically translatable therapeutic strategy to treat NS-associated hypertrophic cardiomyopathy.


Subject(s)
CRISPR-Cas Systems , Cardiomyopathies , Genetic Therapy , Induced Pluripotent Stem Cells/metabolism , Models, Cardiovascular , Mutation , Myocytes, Cardiac/metabolism , Noonan Syndrome , Transcription Factors , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/therapy , Humans , Introns , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Noonan Syndrome/therapy , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Basic Res Cardiol ; 115(3): 27, 2020 03 07.
Article in English | MEDLINE | ID: mdl-32146539

ABSTRACT

Heart failure is a major health problem worldwide with a significant morbidity and mortality rate. Although studied extensively in animal models, data from patients at the compensated disease stage are lacking. We sampled myocardium biopsies from aortic stenosis patients with compensated hypertrophy and moderate heart failure and used transcriptomics to study the transition to failure. Sequencing and comparative analysis of analogous samples of mice with transverse aortic constriction identified 25 candidate genes with similar regulation in response to pressure overload, reflecting highly conserved molecular processes. The gene cysteine-rich secretory protein LCCL domain containing 1 (CRISPLD1) is upregulated in the transition to failure in human and mouse and its function is unknown. Homology to ion channel regulatory toxins suggests a role in Ca2+ cycling. CRISPR/Cas9-mediated loss-of-function leads to dysregulated Ca2+ handling in human-induced pluripotent stem cell-derived cardiomyocytes. The downregulation of prohypertrophic, proapoptotic and Ca2+-signaling pathways upon CRISPLD1-KO and its upregulation in the transition to failure implicates a contribution to adverse remodeling. These findings provide new pathophysiological data on Ca2+ regulation in the transition to failure and novel candidate genes with promising potential for therapeutic interventions.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Adhesion Molecules/metabolism , Evolution, Molecular , Heart Failure/metabolism , Amino Acid Sequence , Animals , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Apoptosis , Biopsy , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Conserved Sequence , Down-Regulation , Female , Heart Failure/complications , Heart Failure/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Male , Mice , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
5.
Stem Cells ; 36(2): 265-277, 2018 02.
Article in English | MEDLINE | ID: mdl-29086457

ABSTRACT

The ability to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes (CMs) makes them an attractive source for repairing injured myocardium, disease modeling, and drug testing. Although current differentiation protocols yield hPSC-CMs to >90% efficiency, hPSC-CMs exhibit immature characteristics. With the goal of overcoming this limitation, we tested the effects of varying passive stretch on engineered heart muscle (EHM) structural and functional maturation, guided by computational modeling. Human embryonic stem cells (hESCs, H7 line) or human induced pluripotent stem cells (IMR-90 line) were differentiated to hPSC-derived cardiomyocytes (hPSC-CMs) in vitro using a small molecule based protocol. hPSC-CMs were characterized by troponin+ flow cytometry as well as electrophysiological measurements. Afterwards, 1.2 × 106 hPSC-CMs were mixed with 0.4 × 106 human fibroblasts (IMR-90 line) (3:1 ratio) and type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane reservoirs to vary nominal passive stretch of EHMs to 5, 7, or 9 mm. EHM characteristics were monitored for up to 50 days, with EHMs having a passive stretch of 7 mm giving the most consistent formation. Based on our initial macroscopic observations of EHM formation, we created a computational model that predicts the stress distribution throughout EHMs, which is a function of cellular composition, cellular ratio, and geometry. Based on this predictive modeling, we show cell alignment by immunohistochemistry and coordinated calcium waves by calcium imaging. Furthermore, coordinated calcium waves and mechanical contractions were apparent throughout entire EHMs. The stiffness and active forces of hPSC-derived EHMs are comparable with rat neonatal cardiomyocyte-derived EHMs. Three-dimensional EHMs display increased expression of mature cardiomyocyte genes including sarcomeric protein troponin-T, calcium and potassium ion channels, ß-adrenergic receptors, and t-tubule protein caveolin-3. Passive stretch affects the structural and functional maturation of EHMs. Based on our predictive computational modeling, we show how to optimize cell alignment and calcium dynamics within EHMs. These findings provide a basis for the rational design of EHMs, which enables future scale-up productions for clinical use in cardiovascular tissue engineering. Stem Cells 2018;36:265-277.


Subject(s)
Computational Biology/methods , Myocardium/cytology , Cell Line , Flow Cytometry , Humans , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Tissue Engineering/methods
6.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28167635

ABSTRACT

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Subject(s)
Embryonic Stem Cells/transplantation , Heart Failure/therapy , Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/transplantation , Tissue Engineering/methods , Ventricular Remodeling/physiology , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Heart Failure/pathology , Humans , Induced Pluripotent Stem Cells/physiology , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/physiology , Printing, Three-Dimensional , Rats , Rats, Nude
7.
Europace ; 20(FI1): f46-f56, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29566126

ABSTRACT

Aims: Our aim is to investigate the arrhythmogenic mechanism in arrhythmogenic right ventricular cardiomyopathy (ARVC)-patients by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods and results: Human-induced pluripotent stem cell-derived cardiomyocytes were generated from human skin fibroblasts of two healthy donors and an ARVC-patient with a desmoglein-2 (DSG2) mutation. Patch clamp, quantitative polymerase chain reaction, and calcium imaging techniques were employed for the study. The amplitude and maximal upstroke velocity (Vmax) of action potential (AP) in ARVC-cells were smaller than that in healthy donor cells, whereas the resting potential and AP duration (APD) was not changed. The reduced Vmax resulted from decreased peak sodium current. The reason for undetected changes in APD may be the counter-action of reduced transient outward, small conductance Ca2+-activated, adenosine triphosphate-sensitive, Na/Ca exchanger (INCX) currents, and enhanced rapidly delayed rectifier currents. Isoprenaline (Iso) reduced INCX and shortened APD in both donor and ARVC-hiPSC-CMs. However, the effects of Iso in ARVC-cells are significantly larger than that in donor cells. In addition, ARVC-hiPSC-CMs showed more frequently than donor cells arrhythmogenic events induced by adrenergic stimulation. Conclusion: Cardiomyocytes derived from the ARVC patient with a DSG2 mutation displayed multiple ion channel dysfunctions and abnormal cellular electrophysiology as well as enhanced sensitivity to adrenergic stimulation. These may underlie the arrhythmogenesis in ARVC patients.


Subject(s)
Action Potentials , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Adrenergic beta-Agonists/pharmacology , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Calcium Signaling , Case-Control Studies , Cells, Cultured , Delayed Rectifier Potassium Channels/metabolism , Desmoglein 2/genetics , Desmoglein 2/metabolism , Genetic Predisposition to Disease , Heart Rate , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Isoproterenol/pharmacology , Kinetics , Male , Middle Aged , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenotype , Sodium-Calcium Exchanger/metabolism
8.
J Mol Cell Cardiol ; 113: 9-21, 2017 12.
Article in English | MEDLINE | ID: mdl-28941705

ABSTRACT

The ability to generate patient-specific induced pluripotent stem cells (iPSCs) provides a unique opportunity for modeling heart disease in vitro. In this study, we generated iPSCs from a patient with dilated cardiomyopathy (DCM) caused by a missense mutation S635A in RNA-binding motif protein 20 (RBM20) and investigated the functionality and cell biology of cardiomyocytes (CMs) derived from patient-specific iPSCs (RBM20-iPSCs). The RBM20-iPSC-CMs showed abnormal distribution of sarcomeric α-actinin and defective calcium handling compared to control-iPSC-CMs, suggesting disorganized myofilament structure and altered calcium machinery in CMs of the RBM20 patient. Engineered heart muscles (EHMs) from RBM20-iPSC-CMs showed that not only active force generation was impaired in RBM20-EHMs but also passive stress of the tissue was decreased, suggesting a higher visco-elasticity of RBM20-EHMs. Furthermore, we observed a reduced titin (TTN) N2B-isoform expression in RBM20-iPSC-CMs by demonstrating a reduction of exon skipping in the PEVK region of TTN and an inhibition of TTN isoform switch. In contrast, in control-iPSC-CMs both TTN isoforms N2B and N2BA were expressed, indicating that the TTN isoform switch occurs already during early cardiogenesis. Using next generation RNA sequencing, we mapped transcriptome and splicing target profiles of RBM20-iPSC-CMs and identified different cardiac gene networks in response to the analyzed RBM20 mutation in cardiac-specific processes. These findings shed the first light on molecular mechanisms of RBM20-dependent pathological cardiac remodeling leading to DCM. Our data demonstrate that iPSC-CMs coupled with EHMs provide a powerful tool for evaluating disease-relevant functional defects and for a deeper mechanistic understanding of alternative splicing-related cardiac diseases.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Adult , Animals , Calcium/metabolism , Cells, Cultured , Connectin/metabolism , Female , Humans , Mice , Mutation , Phenotype , RNA Splicing/genetics , Sarcomeres/metabolism , Transcriptome/genetics
9.
Circ Res ; 117(8): 720-30, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26291556

ABSTRACT

RATIONALE: Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE: Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS: We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS: EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.


Subject(s)
Embryonic Stem Cells/transplantation , Graft Survival , Heart Transplantation/methods , Myocardial Infarction/surgery , Myocytes, Cardiac/transplantation , Papillary Muscles/transplantation , Tissue Engineering/methods , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Survival , Connexin 43/metabolism , Disease Models, Animal , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Heart Transplantation/adverse effects , Heterografts , Humans , Immunosuppressive Agents/pharmacology , Male , Myocardial Contraction , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Papillary Muscles/immunology , Papillary Muscles/metabolism , Papillary Muscles/pathology , Papillary Muscles/physiopathology , Rats, Nude , Rats, Sprague-Dawley , Stroke Volume , Time Factors , Transfection
11.
J Mol Cell Cardiol ; 88: 39-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26392029

ABSTRACT

Cardiac remodeling, a hallmark of heart disease, is associated with intense auto- and paracrine signaling leading to cardiac fibrosis. We hypothesized that the specific mediator of Gq/11-dependent RhoA activation p63RhoGEF, which is expressed in cardiac fibroblasts, plays a role in the underlying processes. We could show that p63RhoGEF is up-regulated in mouse hearts subjected to transverse aortic constriction (TAC). In an engineered heart muscle model (EHM), p63RhoGEF expression in cardiac fibroblasts increased resting and twitch tensions, and the dominant negative p63ΔN decreased both. In an engineered connective tissue model (ECT), p63RhoGEF increased tissue stiffness and its knockdown as well as p63ΔN reduced stiffness. In 2D cultures of neonatal rat cardiac fibroblasts, p63RhoGEF regulated the angiotensin II (Ang II)-dependent RhoA activation, the activation of the serum response factor, and the expression and secretion of the connective tissue growth factor (CTGF). All these processes were inhibited by the knockdown of p63RhoGEF or by p63ΔN likely based on their negative influence on the actin cytoskeleton. Moreover, we show that p63RhoGEF also regulates CTGF in engineered tissues and correlates with it in the TAC model. Finally, confocal studies revealed a closely related localization of p63RhoGEF and CTGF in the trans-Golgi network.


Subject(s)
Connective Tissue Growth Factor/genetics , Fibroblasts/metabolism , Myocardium/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Serum Response Factor/genetics , rhoA GTP-Binding Protein/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Animals, Newborn , Aorta/surgery , Autocrine Communication/genetics , Connective Tissue Growth Factor/metabolism , Constriction , Female , Fibroblasts/pathology , Fibroblasts/ultrastructure , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Models, Cardiovascular , Myocardium/pathology , Paracrine Communication/genetics , Rats , Rats, Wistar , Rho Guanine Nucleotide Exchange Factors/metabolism , Serum Response Factor/metabolism , Signal Transduction , Ventricular Remodeling , rhoA GTP-Binding Protein/metabolism , trans-Golgi Network/metabolism , trans-Golgi Network/ultrastructure
12.
Pflugers Arch ; 467(10): 2141-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25578858

ABSTRACT

Several genetically modified mouse models implicated that prolyl-4-hydroxylase domain (PHD) enzymes are critical mediators for protecting tissues from an ischemic insult including myocardial infarction by affecting the stability and activation of hypoxia-inducible factor (HIF)-1 and HIF-2. Thus, the current efforts to develop small-molecule PHD inhibitors open a new therapeutic option for myocardial tissue protection during ischemia. Therefore, we aimed to investigate the applicability and efficacy of pharmacological HIFα stabilization by a small-molecule PHD inhibitor in the heart. We tested for protective effects in the acute phase of myocardial infarction after pre- or post-conditional application of the inhibitor. Application of the specific PHD inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate (ICA) resulted in HIF-1α and HIF-2α accumulation in heart muscle cells in vitro and in vivo. The rapid and robust responsiveness of cardiac tissue towards ICA was further confirmed by induction of the known HIF target genes heme oxygenase-1 and PHD3. Pre- and post-conditional treatment of mice undergoing myocardial infarction resulted in a significantly smaller infarct size. Tissue protection from ischemia after pre- or post-conditional ICA treatment demonstrates that there is a therapeutic time window for the application of the PHD inhibitor (PHI) post-myocardial infarction, which might be exploited for acute medical interventions.


Subject(s)
Cardiotonic Agents/therapeutic use , Glycine/analogs & derivatives , Isoquinolines/pharmacology , Myocardial Infarction/drug therapy , Prolyl-Hydroxylase Inhibitors/therapeutic use , Animals , Glycine/pharmacology , Glycine/therapeutic use , Hypoxia-Inducible Factor 1/metabolism , Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Isoquinolines/therapeutic use , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Transcription Factors/metabolism
13.
Cell Physiol Biochem ; 34(2): 455-62, 2014.
Article in English | MEDLINE | ID: mdl-25095893

ABSTRACT

BACKGROUND/AIMS: The hypoxia inducible factor-1 (HIF-1) is a suitable marker for tissue oxygenation. We intended to develop cardiomyocytes (CMs) expressing the oxygen-dependent degradation domain of HIF-1α fused to the firefly luciferase (ODD-Luc) followed by proof-of-concept for its applicability in the assessment of heart muscle oxygenation. METHODS AND RESULTS: We first generated embryonic stem cell (ESC) lines (ODD-Luc ESCs) from a Tg ROSA26 ODD-Luc/+ mouse. Subsequent CMs selection was facilitated by stable integration of an antibiotic resistance expressed under the control of the αMHC promoter. ODD-Luc ESCs showed a strong Luc-signal within 1 h of hypoxia (1% oxygen), which coincided with endogenous HIF-1α. Engineered heart muscle (EHM) constructed with ODD-Luc CMs confirmed the utility of the model to sense hypoxia, and monitor reoxygenation also in a multicellular heart muscle model. Pharmacologically induced inotropy/chronotropy under isoprenaline resulted in enhanced Luc-signal suggesting enhanced oxygen consumption, leading to notable myocardial hypoxia. CONCLUSIONS: ODD-Luc-CMs can be used to monitor dynamic changes of cardiomyocyte oxygenation in living heart muscle samples. We provide proof-of-concept for pharmacologically induced myocardial interventions and envision applications of the developed model in drug screens and fundamental studies of ischemia/reperfusion injury.


Subject(s)
Genetic Engineering , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocytes, Cardiac/metabolism , Animals , Mice
14.
Eur Heart J ; 34(33): 2618-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22798560

ABSTRACT

AIMS: Induced pluripotent stem cells (iPSCs) provide a unique opportunity for the generation of patient-specific cells for use in disease modelling, drug screening, and regenerative medicine. The aim of this study was to compare human-induced pluripotent stem cells (hiPSCs) derived from different somatic cell sources regarding their generation efficiency and cardiac differentiation potential, and functionalities of cardiomyocytes. METHODS AND RESULTS: We generated hiPSCs from hair keratinocytes, bone marrow mesenchymal stem cells (MSCs), and skin fibroblasts by using two different virus systems. We show that MSCs and fibroblasts are more easily reprogrammed than keratinocytes. This corresponds to higher methylation levels of minimal promoter regions of the OCT4 and NANOG genes in keratinocytes than in MSCs and fibroblasts. The success rate and reprogramming efficiency was significantly higher by using the STEMCCA system than the OSNL system. All analysed hiPSCs are pluripotent and show phenotypical characteristics similar to human embryonic stem cells. We studied the cardiac differentiation efficiency of generated hiPSC lines (n = 24) and found that MSC-derived hiPSCs exhibited a significantly higher efficiency to spontaneously differentiate into beating cardiomyocytes when compared with keratinocyte-, and fibroblast-derived hiPSCs. There was no significant difference in the functionalities of the cardiomyocytes derived from hiPSCs with different origins, showing the presence of pacemaker-, atrial-, ventricular- and Purkinje-like cardiomyocytes, and exhibiting rhythmic Ca2+ transients and Ca2+ sparks in hiPSC-derived cardiomyocytes. Furthermore, spontaneously and synchronously beating and force-developing engineered heart tissues were generated. CONCLUSIONS: Human-induced pluripotent stem cells can be reprogrammed from all three somatic cell types, but with different efficiency. All analysed iPSCs can differentiate into cardiomyocytes, and the functionalities of cardiomyocytes derived from different cell origins are similar. However, MSC-derived hiPSCs revealed a higher cardiac differentiation efficiency than keratinocyte- and fibroblast-derived hiPSCs.


Subject(s)
Bone Marrow Cells/cytology , Fibroblasts/cytology , Hair/cytology , Induced Pluripotent Stem Cells/cytology , Keratinocytes/cytology , Skin/cytology , Action Potentials/physiology , Biomarkers/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cellular Reprogramming/physiology , DNA Methylation/physiology , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Tissue Engineering
15.
STAR Protoc ; 5(1): 102794, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38133957

ABSTRACT

Force generation is an essential property of skeletal muscle models in vitro. We describe a versatile 1-step procedure to direct undifferentiated human pluripotent stem cells (PSCs) into contractile skeletal muscle organoids (SMOs). Our protocol provides detailed steps for 3D casting of PSCs using either collagen-I/Matrigel- or fibrin/Geltrex-based hydrogels, SMO differentiation, and application of different culture platforms for mechanical loading and contractility analysis. The SMO model may be particularly useful to study human muscle development and developmental skeletal muscle disorders in vitro. For complete details on the use and execution of this protocol, please refer to Shahriyari et al.1.


Subject(s)
Organoids , Pluripotent Stem Cells , Humans , Muscle, Skeletal , Cell Differentiation
16.
Circ Res ; 108(2): 176-83, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21127295

ABSTRACT

RATIONALE: An increase in cardiac afterload typically produces concentric hypertrophy characterized by an increase in cardiomyocyte width, whereas volume overload or exercise results in eccentric growth characterized by cellular elongation and addition of sarcomeres in series. The signaling pathways that control eccentric versus concentric heart growth are not well understood. OBJECTIVE: To determine the role of extracellular signal-regulated kinase 1 and 2 (ERK1/2) in regulating the cardiac hypertrophic response. METHODS AND RESULTS: Here, we used mice lacking all ERK1/2 protein in the heart (Erk1(-/-) Erk2(fl/fl-Cre)) and mice expressing activated mitogen-activated protein kinase kinase (Mek)1 in the heart to induce ERK1/2 signaling, as well as mechanistic experiments in cultured myocytes to assess cellular growth characteristics associated with this signaling pathway. Although genetic deletion of all ERK1/2 from the mouse heart did not block the cardiac hypertrophic response per se, meaning that the heart still increased in weight with both aging and pathological stress stimulation, it did dramatically alter how the heart grew. For example, adult myocytes from hearts of Erk1(-/-) Erk2(fl/fl-Cre) mice showed preferential eccentric growth (lengthening), whereas myocytes from Mek1 transgenic hearts showed concentric growth (width increase). Isolated adult myocytes acutely inhibited for ERK1/2 signaling by adenoviral gene transfer showed spontaneous lengthening, whereas infection with an activated Mek1 adenovirus promoted constitutive ERK1/2 signaling and increased myocyte thickness. A similar effect was observed in engineered heart tissue under cyclic stretching, where ERK1/2 inhibition led to preferential lengthening. CONCLUSIONS: Taken together, these data demonstrate that the ERK1/2 signaling pathway uniquely regulates the balance between eccentric and concentric growth of the heart.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Cells, Cultured , Hypertrophy , MAP Kinase Kinase 1/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Models, Animal , Phosphorylation , Signal Transduction/physiology
17.
Circ Res ; 109(10): 1105-14, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21921264

ABSTRACT

RATIONALE: Cardiac tissue engineering should provide "realistic" in vitro heart muscle models and surrogate tissue for myocardial repair. For either application, engineered myocardium should display features of native myocardium, including terminal differentiation, organotypic maturation, and hypertrophic growth. OBJECTIVE: To test the hypothesis that 3D-engineered heart tissue (EHT) culture supports (1) terminal differentiation as well as (2) organotypic assembly and maturation of immature cardiomyocytes, and (3) constitutes a methodological platform to investigate mechanisms underlying hypertrophic growth. METHODS AND RESULTS: We generated EHTs from neonatal rat cardiomyocytes and compared morphological and molecular properties of EHT and native myocardium from fetal, neonatal, and adult rats. We made the following key observations: cardiomyocytes in EHT (1) gained a high level of binucleation in the absence of notable cytokinesis, (2) regained a rod-shape and anisotropic sarcomere organization, (3) demonstrated a fetal-to-adult gene expression pattern, and (4) responded to distinct hypertrophic stimuli with concentric or eccentric hypertrophy and reexpression of fetal genes. The process of terminal differentiation and maturation (culture days 7-12) was preceded by a tissue consolidation phase (culture days 0-7) with substantial cardiomyocyte apoptosis and dynamic extracellular matrix restructuring. CONCLUSIONS: This study documents the propensity of immature cardiomyocytes to terminally differentiate and mature in EHT in a remarkably organotypic manner. It moreover provides the rationale for the utility of the EHT technology as a methodological bridge between 2D cell culture and animal models.


Subject(s)
Cardiomegaly/pathology , Cell Differentiation , Cell Proliferation , Myocardium/pathology , Myocytes, Cardiac/pathology , Regeneration , Tissue Engineering , Age Factors , Aging , Animals , Animals, Newborn , Apoptosis , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cell Culture Techniques , Cell Differentiation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Myocardial Contraction , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Organogenesis , Proteomics/methods , Rats , Rats, Wistar , Regeneration/genetics , Sarcomeres/metabolism , Sarcomeres/pathology
18.
Nat Cardiovasc Res ; 2(12): 1262-1276, 2023.
Article in English | MEDLINE | ID: mdl-38665939

ABSTRACT

Arrhythmogenic cardiomyopathy is a severe cardiac disorder characterized by lethal arrhythmias and sudden cardiac death, with currently no effective treatment. Plakophilin 2 (PKP2) is the most frequently affected gene. Here we show that adeno-associated virus (AAV)-mediated delivery of PKP2 in PKP2c.2013delC/WT induced pluripotent stem cell-derived cardiomyocytes restored not only cardiac PKP2 levels but also the levels of other junctional proteins, found to be decreased in response to the mutation. PKP2 restoration improved sodium conduction, indicating rescue of the arrhythmic substrate in PKP2 mutant induced pluripotent stem cell-derived cardiomyocytes. Additionally, it enhanced contractile function and normalized contraction kinetics in PKP2 mutant engineered human myocardium. Recovery of desmosomal integrity and cardiac function was corroborated in vivo, by treating heterozygous Pkp2c.1755delA knock-in mice. Long-term treatment with AAV9-PKP2 prevented cardiac dysfunction in 12-month-old Pkp2c.1755delA/WT mice, without affecting wild-type mice. These findings encourage clinical exploration of PKP2 gene therapy for patients with PKP2 haploinsufficiency.

19.
Cardiovasc Res ; 119(16): 2623-2637, 2023 12 19.
Article in English | MEDLINE | ID: mdl-37677054

ABSTRACT

AIMS: Atrial fibrillation (AF) is associated with tachycardia-induced cellular electrophysiology alterations which promote AF chronification and treatment resistance. Development of novel antiarrhythmic therapies is hampered by the absence of scalable experimental human models that reflect AF-associated electrical remodelling. Therefore, we aimed to assess if AF-associated remodelling of cellular electrophysiology can be simulated in human atrial-like cardiomyocytes derived from induced pluripotent stem cells in the presence of retinoic acid (iPSC-aCM), and atrial-engineered human myocardium (aEHM) under short term (24 h) and chronic (7 days) tachypacing (TP). METHODS AND RESULTS: First, 24-h electrical pacing at 3 Hz was used to investigate whether AF-associated remodelling in iPSC-aCM and aEHM would ensue. Compared to controls (24 h, 1 Hz pacing) TP-stimulated iPSC-aCM presented classical hallmarks of AF-associated remodelling: (i) decreased L-type Ca2+ current (ICa,L) and (ii) impaired activation of acetylcholine-activated inward-rectifier K+ current (IK,ACh). This resulted in action potential shortening and an absent response to the M-receptor agonist carbachol in both iPSC-aCM and aEHM subjected to TP. Accordingly, mRNA expression of the channel-subunit Kir3.4 was reduced. Selective IK,ACh blockade with tertiapin reduced basal inward-rectifier K+ current only in iPSC-aCM subjected to TP, thereby unmasking an agonist-independent constitutively active IK,ACh. To allow for long-term TP, we developed iPSC-aCM and aEHM expressing the light-gated ion-channel f-Chrimson. The same hallmarks of AF-associated remodelling were observed after optical-TP. In addition, continuous TP (7 days) led to (i) increased amplitude of inward-rectifier K+ current (IK1), (ii) hyperpolarization of the resting membrane potential, (iii) increased action potential-amplitude and upstroke velocity as well as (iv) reversibly impaired contractile function in aEHM. CONCLUSIONS: Classical hallmarks of AF-associated remodelling were mimicked through TP of iPSC-aCM and aEHM. The use of the ultrafast f-Chrimson depolarizing ion channel allowed us to model the time-dependence of AF-associated remodelling in vitro for the first time. The observation of electrical remodelling with associated reversible contractile dysfunction offers a novel platform for human-centric discovery of antiarrhythmic therapies.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Heart Atria , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Action Potentials , Acetylcholine/pharmacology
20.
Methods Mol Biol ; 2485: 213-225, 2022.
Article in English | MEDLINE | ID: mdl-35618908

ABSTRACT

Different engineered heart muscle formats have been developed for applications in disease modeling, drug screening, and heart repair. The advantage of 3D engineered versus 2D monolayer and 3D aggregate cardiomyocyte cultures is a clearly advanced degree of maturation, which in many aspects resembles the postnatal rather than the embryonic or fetal heart, in the most advanced 3D culture formats. According to the desired in vitro (disease modeling or drug screening) and in vivo (heart repair) application, scale and geometry of tissue engineered heart muscle must be adapted. In this updated methods paper, we report a simple and scalable (up and down) collagen-based protocol for the construction of Engineered Human Myocardium (EHM) under defined, serum-free conditions.


Subject(s)
Cardiac Surgical Procedures , Myocardium , Drug Evaluation, Preclinical , Humans , Myocytes, Cardiac/physiology , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL