Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
ESC Heart Fail ; 9(1): 224-235, 2022 02.
Article in English | MEDLINE | ID: mdl-34931757

ABSTRACT

AIMS: Hippo signalling is an evolutionarily conserved pathway that controls organ size by regulating apoptosis, cell proliferation, and stem cell self-renewal. Recently, the pathway has been shown to exert powerful growth regulatory activity in cardiomyocytes. However, the functional role of this stress-related and cell death-related pathway in the human heart and cardiomyocytes is not known. In this study, we investigated the role of the transcriptional co-activators of Hippo signalling, YAP and TAZ, in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in response to cardiotoxic agents and investigated the effects of modulating the pathway on cardiomyocyte function and survival. METHODS AND RESULTS: RNA-sequencing analysis of human heart samples with doxorubicin-induced end-stage heart failure and healthy controls showed that YAP and ERBB2 (HER2) as upstream regulators of differentially expressed genes correlated with doxorubicin treatment. Thus, we tested the effects of doxorubicin on hiPSC-CMs in vitro. Using an automated high-content screen of 96 clinically relevant antineoplastic and cardiotherapeutic drugs, we showed that doxorubicin induced the highest activation of YAP/TAZ nuclear translocation in both hiPSC-CMs and control MCF7 breast cancer cells. The overexpression of YAP rescued doxorubicin-induced cell loss in hiPSC-CMs by inhibiting apoptosis and inducing proliferation. In contrast, silencing of YAP and TAZ by siRNAs resulted in elevated mitochondrial membrane potential loss in response to doxorubicin. hiPSC-CM calcium transients did not change in response to YAP/TAZ silencing. CONCLUSIONS: Our results suggest that Hippo signalling is involved in clinical anthracycline-induced cardiomyopathy. Modelling with hiPSC-CMs in vitro showed similar responses to doxorubicin as adult cardiomyocytes and revealed a potential cardioprotective effect of YAP in doxorubicin-induced cardiotoxicity.


Subject(s)
Cardiomyopathies , Transcription Factors , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Doxorubicin/metabolism , Humans , Myocytes, Cardiac/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , YAP-Signaling Proteins
2.
J Clin Microbiol ; 58(5)2020 04 23.
Article in English | MEDLINE | ID: mdl-32132188

ABSTRACT

Q fever, caused by Coxiella burnetii, is a worldwide zoonotic disease that may cause severe forms in humans and requires a specific and prolonged antibiotic treatment. Although current serological and molecular detection tools allow a reliable diagnosis of the disease, culture of C. burnetii strains is mandatory to assess their susceptibility to antibiotics and sequence their genome in order to optimize patient management and epidemiological studies. However, cultivating this fastidious microorganism is difficult and restricted to reference centers, as it requires biosafety level 3 laboratories and relies on cell culture performed by experienced technicians. In addition, the culture yield is low, which results in a small number of isolates being available. In this work, we developed a novel high-content screening (HCS) isolation strategy based on optimized high-throughput cell culture and automated microscopic detection of infected cells with specifically designed algorithms targeting cytopathic effects. This method was more efficient than the shell vial assay, at the level of time dependency, when applied to both frozen specimens (7 isolates recovered by HCS only, sensitivity 91% versus 78% for shell vial) and fresh samples (1 additional isolate using HCS, sensitivity 7% versus 5% for shell vial), for which most strains were recovered more rapidly with the new technique. In addition, detecting positive cultures by an automated microscope reduced the need for expertise and saved 24% of technician working time. Application of HCS to antibiotic susceptibility testing of 12 strains demonstrated that it was as efficient as the standard procedure that combines shell vial culture and quantitative PCR.


Subject(s)
Coxiella burnetii , Q Fever , Anti-Bacterial Agents/pharmacology , Coxiella burnetii/genetics , Humans , Q Fever/diagnosis , Real-Time Polymerase Chain Reaction
3.
Methods Mol Biol ; 2150: 57-77, 2020.
Article in English | MEDLINE | ID: mdl-30941720

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) and their differentiated derivatives became a new, promising source for in vitro screening techniques. Cell lines derived from healthy individuals can be applied for drug safety testing, while patient-derived cells provide a platform to model diseases in vitro and can be used as a tool for personalized medicine including specific drug efficacy testing and identification of new pharmacological targets as well as for tailoring pharmacological therapies. Efficient differentiation protocols yielding cardiomyocytes or endothelial cells derived from iPSCs have been developed recently. Phenotypic characterization and gene expression profiling of these derivatives can reveal clues for developmental and pathological questions. Moreover, functional analysis and cell-based assays using automated fluorescence imaging platform and high content analysis characterize cell type-specific profiles of hiPSC-derived cardiomyocytes (hiPSC-CM) and endothelial cells (hiPSC-EC) at the cellular and subcellular levels. This can be utilized in a platform which can provide multiple endpoint profiles of candidate compounds.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/cytology , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Biomarkers/metabolism , Cell Death , Cells, Cultured , Embryo, Mammalian/cytology , Feeder Cells/cytology , Fibroblasts/cytology , Freezing , Humans , Mice , Multivariate Analysis , Neovascularization, Physiologic
4.
PLoS One ; 10(5): e0126860, 2015.
Article in English | MEDLINE | ID: mdl-25993131

ABSTRACT

Huntington disease (HD; OMIM 143100), a progressive neurodegenerative disorder, is caused by an expanded trinucleotide CAG (polyQ) motif in the HTT gene. Cardiovascular symptoms, often present in early stage HD patients, are, in general, ascribed to dysautonomia. However, cardio-specific expression of polyQ peptides caused pathological response in murine models, suggesting the presence of a nervous system-independent heart phenotype in HD patients. A positive correlation between the CAG repeat size and severity of symptoms observed in HD patients has also been observed in in vitro HD cellular models. Here, we test the suitability of human embryonic stem cell (hESC) lines carrying HD-specific mutation as in vitro models for understanding molecular mechanisms of cardiac pathology seen in HD patients. We have differentiated three HD-hESC lines into cardiomyocytes and investigated CAG stability up to 60 days after starting differentiation. To assess CAG stability in other tissues, the lines were also subjected to in vivo differentiation into teratomas for 10 weeks. Neither directed differentiation into cardiomyocytes in vitro nor in vivo differentiation into teratomas, rich in immature neuronal tissue, led to an increase in the number of CAG repeats. Although the CAG stability might be cell line-dependent, induced pluripotent stem cells generated from patients with larger numbers of CAG repeats could have an advantage as a research tool for understanding cardiac symptoms of HD patients.


Subject(s)
Cell Differentiation/genetics , Human Embryonic Stem Cells/metabolism , Huntington Disease/pathology , Mutation/genetics , Myocytes, Cardiac/cytology , Trinucleotide Repeat Expansion/genetics , Cell Line , Humans , Pluripotent Stem Cells/cytology
5.
Cardiovasc Res ; 105(1): 31-43, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25341891

ABSTRACT

AIMS: During cardiac hypertrophy, cardiomyocytes (CMs) increase in the size and expression of cytoskeletal proteins while reactivating a foetal gene programme. The process is proposed to be dependent on increased nuclear export and, since nuclear pore trafficking has limited capacity, a linked decrease in import. Our objective was to investigate the role of nuclear import and export in control of hypertrophy in rat and human heart failure (HF). METHODS AND RESULTS: In myocardial tissue and isolated CMs from patients with dilated cardiomyopathy, nuclear size was increased; Nucleoporin p62, cytoplasmic RanBP1, and nuclear translocation of importins (α and ß) were decreased while Exportin-1 was increased. CM from a rat HF model 16 weeks after myocardial infarction (MI) reproduced these nuclear changes. Nuclear import, determined by the rate of uptake of nuclear localization sequence (NLS)-tagged fluorescent substrate, was also decreased and this change was observed from 4 weeks after MI, before HF has developed. Treatment of isolated rat CMs with phenylephrine (PE) for 48 h produced similar cell and nuclear size increases, nuclear import and export protein rearrangement, and NLS substrate uptake decrease through p38 MAPK and HDAC-dependent pathways. The change in NLS substrate uptake occurred within 15 min of PE exposure. Inhibition of nuclear export with leptomycin B reversed established nuclear changes in PE-treated rat CMs and decreased NLS substrate uptake and cell/nuclear size in human CMs. CONCLUSIONS: Nuclear transport changes related to increased export and decreased import are an early event in hypertrophic development. Hypertrophy can be prevented, or even reversed, by targeting import/export, which may open new therapeutic opportunities.


Subject(s)
Heart Failure/metabolism , Heart Failure/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Pore/pathology , Active Transport, Cell Nucleus , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Histone Deacetylases/metabolism , Humans , Male , Models, Cardiovascular , Nuclear Localization Signals/metabolism , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Stem Cells Dev ; 24(7): 869-78, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25387407

ABSTRACT

Vascular derivatives of human embryonic stem cells (hESC) are being developed as sources of tissue-specific cells for organ regeneration. However, identity of developmental pathways that modulate the specification of endothelial cells is not known yet. We studied phosphatidylinositol 3-kinase (PI3K)-Forkhead box O transcription factor 1A (FOXO1A) pathways during differentiation of hESC toward endothelial lineage and on proliferation, maturation, and cell death of hESC-derived endothelial cells (hESC-EC). During differentiation of hESC, expression of FOXO1A transcription factor was linked to the expression of a cluster of angiogenesis- and vascular remodeling-related genes. PI3K inhibitor LY294002 activated FOXO1A and induced formation of CD31(+) hESC-EC. In contrast, differentiating hESC with silenced FOXO1A by small interfering RNA (siRNA) showed lower mRNA levels of CD31 and angiopoietin2. LY294002 decreased proliferative activity of purified hESC-EC, while FOXO1A siRNA increased their proliferation. LY294002 inhibits migration and tube formation of hESC-EC; in contrast, FOXO1A siRNA increased in vitro tube formation activity of hESC-EC. After in vivo conditioning of cells in athymic nude rats, cells retain their low FOXO1A expression levels. PI3K/FOXO1A pathway is important for function and survival of hESC-EC and in the regulation of endothelial cell fate. Understanding these properties of hESC-EC may help in future applications for treatment of injured organs.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Human Umbilical Vein Endothelial Cells/cytology , Humans , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/genetics , Rats
7.
Stem Cell Reports ; 3(5): 905-14, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418732

ABSTRACT

Cardiomyocytes from human embryonic stem cells (hESC-CMs) and induced pluripotent stem cells (hiPSC-CMs) represent new models for drug discovery. Although hypertrophy is a high-priority target, we found that hiPSC-CMs were systematically unresponsive to hypertrophic signals such as the α-adrenoceptor (αAR) agonist phenylephrine (PE) compared to hESC-CMs. We investigated signaling at multiple levels to understand the underlying mechanism of this differential responsiveness. The expression of the normal α1AR gene, ADRA1A, was reversibly silenced during differentiation, accompanied by ADRA1B upregulation in either cell type. ADRA1B signaling was intact in hESC-CMs, but not in hiPSC-CMs. We observed an increased tonic activity of inhibitory kinase pathways in hiPSC-CMs, and inhibition of antihypertrophic kinases revealed hypertrophic increases. There is tonic suppression of cell growth in hiPSC-CMs, but not hESC-CMs, limiting their use in investigation of hypertrophic signaling. These data raise questions regarding the hiPSC-CM as a valid model for certain aspects of cardiac disease.


Subject(s)
Adrenergic Agents/pharmacology , Cell Size/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression/drug effects , Humans , Hypertrophy , Imidazoles/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Isoproterenol/pharmacology , Microscopy, Confocal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Stem Cells Dev ; 23(5): 467-76, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24192059

ABSTRACT

Cardiac cell replacement therapy by using human embryonic stem cell (hESC) derivatives remains a potential approach to regenerate myocardium. The major hurdles to clinical application of this technology are immunogenicity and post-transplantation cell death. Here we examined the effects of calcineurin-targeting immunosuppressants cyclosporine A (CsA) and FK506, as well as rapamycin and a selective inhibitor of calcineurin-binding downstream nuclear factor of activated T-cell (NFAT) transcription factor VIVIT on the proliferative activity, function, and survival of hESC-derived cardiomyocytes (hESC-CM) and endothelial cells (hESC-EC) in culture. As shown by automated microscopy, treatments with CsA, FK506, and rapamycin all decreased proliferation, reducing the percentage of hESC-CM and hESC-EC with the mitotic marker Ki67(+) by as much as 60% and 74%, respectively. Administration of the cell permeable analogue 11R-VIVIT protein did not modulate their proliferative activity. All immunosuppressants reversed the proapoptotic effect of chelerythrine in hESC-CM demonstrating an inhibitory role of calcineurin/NFAT and mammalian target of rapamycin (mTOR) pathways in hESC-CM survival (using apoptotic marker caspase-3), whereas the protection was less obvious in hESC-EC exposed to H2O2. Immunosuppressants did not affect cell viability in hESC-EC. Our results show that immunosuppressants reduce proliferation, while offsetting cell loss to a smaller extent by reduction in apoptosis of hESC-CM. Immunosuppressant therapy would be compatible with stem cell transplantation, but the resulting reduction in graft expansion capabilities would potentially necessitate implantation of increased cell numbers when immunosuppressants are given. The effects of NFAT-binding immunosuppressant molecules, which do not affect hESC-CM proliferation, may point the way forward for new classes of compounds better suited to cell implantation.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Embryonic Stem Cells/drug effects , Apoptosis/drug effects , Calcineurin/metabolism , Cells, Cultured , Cyclosporine/administration & dosage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oligopeptides/metabolism , Sirolimus/administration & dosage , Stem Cell Transplantation , Tacrolimus/administration & dosage
9.
Methods Mol Biol ; 1052: 29-39, 2013.
Article in English | MEDLINE | ID: mdl-23640255

ABSTRACT

Human pluripotent stem cells (hPSC) are investigated as a source of authentic human cardiac cells for drug discovery and toxicological tests. Cell-based assays using automated fluorescence imaging platform and high-content analysis characterize hypertrophic and toxicity profiles of compounds in hPSC-derived cardiomyocytes (hPSC-CM) at the cellular and subcellular levels. In purified population of hPSC-CM loaded with cell tracer probe and cell death markers, both hypertrophic and toxicity profiles can be assessed in live cardiomyocyte cultures. Alternatively, in non-purified cultures of hPSC-CM, hypertrophy, proliferation, and cell death assays can be performed specifically in the cardiomyocyte subpopulation using antibodies directed against cardiac proteins and a combination of cell death- and proliferation-specific fluorescent probes.


Subject(s)
Diagnostic Imaging/methods , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Apoptosis , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Hypertrophy , Pluripotent Stem Cells/cytology
10.
J Cardiovasc Transl Res ; 5(5): 593-604, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22896035

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CM) are being investigated as a new source of cardiac cells for drug safety assessment. We developed a novel scalable high content microscopy-based method for the detection of cell death in hPSC-CM that can serve for future predictive in vitro cardio-toxicological screens. Using rat neonatal ventricular cardiomyocytes (RVNC) or hPSC-CM, assays for nuclear remodelling, mitochondrial status, apoptosis and necrosis were designed using a combination of fluorescent dyes and antibodies on an automated microscopy platform. This allowed the observation of a chelerythrine-induced concentration-dependent apoptosis to necrosis switch and time-dependent progression of early apoptotic cells towards a necrotic-like phenotype. Susceptibility of hPSC-CM to chelerythrine-stimulated apoptosis varied with time after differentiation, but at most time points, hPSC-CM were more resistant than RVNC. This simple and scalable humanized high-content assay generates accurate cardiotoxicity profiles that can serve as a base for further assessment of cardioprotective strategies and drug safety.


Subject(s)
Apoptosis/drug effects , Benzophenanthridines/toxicity , Cell Tracking/methods , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/drug effects , Microscopy, Fluorescence , Myocytes, Cardiac/drug effects , Animals , Animals, Newborn , Automation, Laboratory , Caspases, Effector/metabolism , Cell Line , Cell Membrane Permeability/drug effects , Cell Nucleus Shape/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Image Processing, Computer-Assisted , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Phenotype , Rats , Time Factors
11.
J Mol Cell Cardiol ; 50(2): 367-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21047517

ABSTRACT

Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are being developed for tissue repair and as a model system for cardiac physiology and pathophysiology. However, the signaling requirements of their growth have not yet been fully characterized. We showed that hESC-CM retain their capacity for increase in size in long-term culture. Exposing hESC-CM to hypertrophic stimuli such as equiaxial cyclic stretch, angiotensin II, and phenylephrine (PE) increased cell size and volume, percentage of hESC-CM with organized sarcomeres, levels of ANF, and cytoskeletal assembly. PE effects on cell size were separable from those on cell cycle. Changes in cell size by PE were completely inhibited by p38-MAPK, calcineurin/FKBP, and mTOR blockers. p38-MAPK and calcineurin were also implicated in basal cell growth. Inhibitors of ERK, JNK, and CaMK II partially reduced PE effects; PKG or GSK3ß inhibitors had no effect. The role of p38-MAPK was confirmed by an additional pharmacological inhibitor and adenoviral infection of hESC-CM with a dominant-inhibitory form of p38-MAPK. Infection of hESC-CM with constitutively active upstream MAP2K3b resulted in an increased cell size, sarcomere and cytoskeletal assembly, elongation of the cells, and induction of ANF mRNA levels. siRNA knockdown of p38-MAPK inhibited PE-induced effects on cell size. These results reveal an important role for active protein kinase signaling in hESC-CM growth and hypertrophy, with potential implications for hESC-CM as a novel in vitro test system. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Angiotensin II/pharmacology , Cardiomegaly/chemically induced , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 3/metabolism , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Vasoconstrictor Agents/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
12.
J Cell Mol Med ; 13(9B): 3730-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19438812

ABSTRACT

Embryonic stem cell-derived cardiomyocytes (ESC-CM) have many of the phenotypic properties of authentic cardiomyocytes, and great interest has been shown in their possibilities for modelling human disease. Obstetric cholestasis affects 1 in 200 pregnant women in the United Kingdom. It is characterized by raised serum bile acids and complicated by premature delivery and unexplained fetal death at late gestation. It has been suggested that the fetal death is caused by the enhanced arrhythmogenic effect of bile acids in the fetal heart, and shown that neonatal susceptibility to bile acid-induced arrhythmia is lost in the adult rat cardiomyocyte. However, the mechanisms of the observed bile acid effects are not fully understood and their in vivo study in human beings is difficult. Here we use ESC-CM from both human and mouse ESCs to test our proposal that immature cardiomyocytes are more susceptible to the effect of raised bile acids than mature ones. We show that early ESC-CM exhibit bile acid-induced disruption of rhythm, depression of contraction and desynchronization of cell coupling. In both species the ESC-CM become resistant to these arrhythmias as the cells mature, and this develops in line with the respective gestational periods of mouse and human. This represents the first demonstration of the use of ESC-CM as a model system for human cardiac pathology, and opens the way for both investigation of mechanisms and a high throughput screen for drug discovery.


Subject(s)
Arrhythmias, Cardiac/pathology , Bile Acids and Salts/metabolism , Fetal Diseases/pathology , Myocytes, Cardiac/cytology , Animals , Calcium/metabolism , Cell Differentiation , Cholestasis/metabolism , Disease Models, Animal , Female , Humans , Mice , Myocytes, Cardiac/metabolism , Pregnancy , Pregnancy Complications, Cardiovascular , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...