Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell Rep ; 36(4): 109429, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320344

ABSTRACT

Patient-derived tumor organoids (TOs) are emerging as high-fidelity models to study cancer biology and develop novel precision medicine therapeutics. However, utilizing TOs for systems-biology-based approaches has been limited by a lack of scalable and reproducible methods to develop and profile these models. We describe a robust pan-cancer TO platform with chemically defined media optimized on cultures acquired from over 1,000 patients. Crucially, we demonstrate tumor genetic and transcriptomic concordance utilizing this approach and further optimize defined minimal media for organoid initiation and propagation. Additionally, we demonstrate a neural-network-based high-throughput approach for label-free, light-microscopy-based drug assays capable of predicting patient-specific heterogeneity in drug responses with applicability across solid cancers. The pan-cancer platform, molecular data, and neural-network-based drug assay serve as resources to accelerate the broad implementation of organoid models in precision medicine research and personalized therapeutic profiling programs.


Subject(s)
Neoplasms/pathology , Organoids/pathology , Precision Medicine , Cell Proliferation , Drug Screening Assays, Antitumor , Female , Fluorescence , Genomics , HLA Antigens/genetics , Humans , Loss of Heterozygosity , Male , Middle Aged , Models, Biological , Neoplasms/genetics , Neural Networks, Computer , Transcriptome/genetics
2.
Med Image Anal ; 56: 122-139, 2019 08.
Article in English | MEDLINE | ID: mdl-31226662

ABSTRACT

Breast cancer is the most common invasive cancer in women, affecting more than 10% of women worldwide. Microscopic analysis of a biopsy remains one of the most important methods to diagnose the type of breast cancer. This requires specialized analysis by pathologists, in a task that i) is highly time- and cost-consuming and ii) often leads to nonconsensual results. The relevance and potential of automatic classification algorithms using hematoxylin-eosin stained histopathological images has already been demonstrated, but the reported results are still sub-optimal for clinical use. With the goal of advancing the state-of-the-art in automatic classification, the Grand Challenge on BreAst Cancer Histology images (BACH) was organized in conjunction with the 15th International Conference on Image Analysis and Recognition (ICIAR 2018). BACH aimed at the classification and localization of clinically relevant histopathological classes in microscopy and whole-slide images from a large annotated dataset, specifically compiled and made publicly available for the challenge. Following a positive response from the scientific community, a total of 64 submissions, out of 677 registrations, effectively entered the competition. The submitted algorithms improved the state-of-the-art in automatic classification of breast cancer with microscopy images to an accuracy of 87%. Convolutional neuronal networks were the most successful methodology in the BACH challenge. Detailed analysis of the collective results allowed the identification of remaining challenges in the field and recommendations for future developments. The BACH dataset remains publicly available as to promote further improvements to the field of automatic classification in digital pathology.


Subject(s)
Breast Neoplasms/pathology , Neural Networks, Computer , Pattern Recognition, Automated , Algorithms , Female , Humans , Microscopy , Staining and Labeling
3.
Nat Biomed Eng ; 2(2): 104-113, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29721363

ABSTRACT

Stem-cell-based therapies hold considerable promise for regenerative medicine. However, acute donor-cell death within several weeks after cell delivery remains a critical hurdle for clinical translation. Co-transplantation of stem cells with pro-survival factors can improve cell engraftment, but this strategy has been hampered by the typically short half-lives of the factors and by the use of Matrigel and other scaffolds that are not chemically defined. Here, we report a collagen-dendrimer biomaterial crosslinked with pro-survival peptide analogues that adheres to the extracellular matrix and slowly releases the peptides, significantly prolonging stem cell survival in mouse models of ischaemic injury. The biomaterial can serve as a generic delivery system to improve functional outcomes in cell-replacement therapy.

4.
Cell Rep ; 20(8): 1978-1990, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834758

ABSTRACT

There is growing interest in using embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) derivatives for tissue regeneration. However, an increased understanding of human immune responses to stem cell-derived allografts is necessary for maintaining long-term graft persistence. To model this alloimmunity, humanized mice engrafted with human hematopoietic and immune cells could prove to be useful. In this study, an in-depth analysis of graft-infiltrating human lymphocytes and splenocytes revealed that humanized mice incompletely model human immune responses toward allogeneic stem cells and their derivatives. Furthermore, using an "allogenized" mouse model, we show the feasibility of reconstituting immunodeficient mice with a functional mouse immune system and describe a key role of innate immune cells in the rejection of mouse stem cell allografts.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunity, Innate/immunology , Pluripotent Stem Cells/metabolism , Transplantation Conditioning/methods , Animals , Disease Models, Animal , Graft Rejection , Humans , Mice
5.
Stem Cells ; 35(8): 1994-2000, 2017 08.
Article in English | MEDLINE | ID: mdl-28600830

ABSTRACT

Human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced PSCs (hiPSCs), have great potential as an unlimited donor source for cell-based therapeutics. The risk of teratoma formation from residual undifferentiated cells, however, remains a critical barrier to the clinical application of these cells. Herein, we describe external beam radiation therapy (EBRT) as an attractive option for the treatment of this iatrogenic growth. We present evidence that EBRT is effective in arresting growth of hESC-derived teratomas in vivo at day 28 post-implantation by using a microCT irradiator capable of targeted treatment in small animals. Within several days of irradiation, teratomas derived from injection of undifferentiated hESCs and hiPSCs demonstrated complete growth arrest lasting several months. In addition, EBRT reduced reseeding potential of teratoma cells during serial transplantation experiments, requiring irradiated teratomas to be seeded at 1 × 103 higher doses to form new teratomas. We demonstrate that irradiation induces teratoma cell apoptosis, senescence, and growth arrest, similar to established radiobiology mechanisms. Taken together, these results provide proof of concept for the use of EBRT in the treatment of existing teratomas and highlight a strategy to increase the safety of stem cell-based therapies. Stem Cells 2017;35:1994-2000.


Subject(s)
Pluripotent Stem Cells/pathology , Radiation, Ionizing , Teratoma/radiotherapy , Apoptosis/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Humans , Pluripotent Stem Cells/radiation effects , Teratoma/pathology
6.
Mol Ther ; 25(2): 427-442, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153093

ABSTRACT

Restoring pluripotency using chemical compounds alone would be a major step forward in developing clinical-grade pluripotent stem cells, but this has not yet been reported in human cells. We previously demonstrated that VPA_AFS cells, human amniocytes cultivated with valproic acid (VPA) acquired functional pluripotency while remaining distinct from human embryonic stem cells (hESCs), questioning the relationship between the modulation of cell fate and molecular regulation of the pluripotency network. Here, we used single-cell analysis and functional assays to reveal that VPA treatment resulted in a homogeneous population of self-renewing non-transformed cells that fulfill the hallmarks of pluripotency, i.e., a short G1 phase, a dependence on glycolytic metabolism, expression of epigenetic modifications on histones 3 and 4, and reactivation of endogenous OCT4 and downstream targets at a lower level than that observed in hESCs. Mechanistic insights into the process of VPA-induced reprogramming revealed that it was dependent on OCT4 promoter activation, which was achieved independently of the PI3K (phosphatidylinositol 3-kinase)/AKT/mTOR (mammalian target of rapamycin) pathway or GSK3ß inhibition but was concomitant with the presence of acetylated histones H3K9 and H3K56, which promote pluripotency. Our data identify, for the first time, the pluripotent transcriptional and molecular signature and metabolic status of human chemically induced pluripotent stem cells.


Subject(s)
Amnion/cytology , Cell Transdifferentiation/drug effects , Cellular Reprogramming/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Biomarkers , Cell Cycle/genetics , Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Energy Metabolism , Epigenesis, Genetic , Female , Gene Expression , Gene Expression Profiling , Genes, Reporter , Glycolysis , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins , TOR Serine-Threonine Kinases/metabolism , Transcriptional Activation
7.
J Am Coll Cardiol ; 68(19): 2086-2096, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27810048

ABSTRACT

BACKGROUND: Brugada syndrome (BrS), a disorder associated with characteristic electrocardiogram precordial ST-segment elevation, predisposes afflicted patients to ventricular fibrillation and sudden cardiac death. Despite marked achievements in outlining the organ level pathophysiology of the disorder, the understanding of human cellular phenotype has lagged due to a lack of adequate human cellular models of the disorder. OBJECTIVES: The objective of this study was to examine single cell mechanism of Brugada syndrome using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS: This study recruited 2 patients with type 1 BrS carrying 2 different sodium voltage-gated channel alpha subunit 5 variants as well as 2 healthy control subjects. We generated iPSCs from their skin fibroblasts by using integration-free Sendai virus. We used directed differentiation to create purified populations of iPSC-CMs. RESULTS: BrS iPSC-CMs showed reductions in inward sodium current density and reduced maximal upstroke velocity of action potential compared with healthy control iPSC-CMs. Furthermore, BrS iPSC-CMs demonstrated increased burden of triggered activity, abnormal calcium (Ca2+) transients, and beating interval variation. Correction of the causative variant by genome editing was performed, and resultant iPSC-CMs showed resolution of triggered activity and abnormal Ca2+ transients. Gene expression profiling of iPSC-CMs showed clustering of BrS compared with control subjects. Furthermore, BrS iPSC-CM gene expression correlated with gene expression from BrS human cardiac tissue gene expression. CONCLUSIONS: Patient-specific iPSC-CMs were able to recapitulate single-cell phenotype features of BrS, including blunted inward sodium current, increased triggered activity, and abnormal Ca2+ handling. This novel human cellular model creates future opportunities to further elucidate the cellular disease mechanism and identify novel therapeutic targets.


Subject(s)
Brugada Syndrome/genetics , Gene Expression Regulation , Heart Conduction System/physiopathology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , NAV1.5 Voltage-Gated Sodium Channel/genetics , RNA/genetics , Adolescent , Adult , Brugada Syndrome/metabolism , Brugada Syndrome/pathology , Cell Differentiation , Electrocardiography , Genotype , Heart Conduction System/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/biosynthesis , Pedigree , Phenotype , Polymerase Chain Reaction , Young Adult
9.
J Mol Cell Cardiol ; 85: 79-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25982839

ABSTRACT

Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSCs) are being increasingly used to model human heart diseases. hiPSC-CMs generated by earlier aggregation-based methods (i.e., embryoid body) often lack functional sarcoplasmic reticulum (SR) Ca stores characteristic of mature mammalian CMs. Newer monolayer-based cardiac differentiation methods (i.e., Matrigel sandwich or small molecule-based differentiation) produce hiPSC-CMs of high purity and yield, but their Ca handling has not been comprehensively investigated. Here, we studied Ca handling and cytosolic Ca buffering properties of hiPSC-CMs generated independently from multiple hiPSC lines at Stanford University, Vanderbilt University and University of Wisconsin-Madison. hiPSC-CMs were cryopreserved at each university. Frozen aliquots were shipped, recovered from cryopreservation, plated at low density and compared 3-5days after plating with acutely-isolated adult rabbit and mouse ventricular CMs. Although hiPSC-CM cell volume was significantly smaller, cell capacitance to cell volume ratio and cytoplasmic Ca buffering were not different from rabbit-CMs. hiPSC-CMs from all three laboratories exhibited robust L-type Ca currents, twitch Ca transients and caffeine-releasable SR Ca stores comparable to adult CMs. Ca transport by sarcoendoplasmic reticulum Ca ATPase (SERCA) and Na/Ca exchanger (NCX) was similar in all hiPSC-CM lines, but slower compared to rabbit-CMs. However, the relative contribution of SERCA and NCX to Ca transport of hiPSC-CMs was comparable to rabbit-CMs. Ca handling maturity of hiPSC-CMs increased from 15 to 21days post-induction. We conclude that hiPSC-CMs generated independently from multiple iPSC lines using monolayer-based methods can be reproducibly recovered from cryopreservation and exhibit comparable and functional SR Ca handling.


Subject(s)
Calcium/metabolism , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/metabolism , Animals , Calcium Signaling , Cell Differentiation , Cells, Cultured , Humans , Mice , Myocardial Contraction , Rabbits , Sarcoplasmic Reticulum/metabolism
11.
Circ Cardiovasc Imaging ; 8(2): e002851, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25609688

ABSTRACT

BACKGROUND: Although single-photon emission computed tomography myocardial perfusion imaging (SPECT MPI) has improved the diagnosis and risk stratification of patients with suspected coronary artery disease, it remains a primary source of low-dose radiation exposure for cardiac patients. To determine the biological effects of low-dose radiation from SPECT MPI, we measured the activation of the DNA damage response pathways using quantitative flow cytometry and single-cell gene expression profiling. METHODS AND RESULTS: Blood samples were collected from patients before and after SPECT MPI (n=63). Overall, analysis of all recruited patients showed no marked differences in the phosphorylation of proteins (H2AX, protein 53, and ataxia telangiectasia mutated) after SPECT. The majority of patients also had either downregulated or unchanged expression in DNA damage response genes at both 24 and 48 hours post-SPECT. Interestingly, a small subset of patients with increased phosphorylation had significant upregulation of genes associated with DNA damage, whereas those with no changes in phosphorylation had significant downregulation or no difference, suggesting that some patients may potentially be more sensitive to low-dose radiation exposure. CONCLUSIONS: Our findings showed that SPECT MPI resulted in a variable activation of the DNA damage response pathways. Although only a small subset of patients had increased protein phosphorylation and elevated gene expression postimaging, continued care should be taken to reduce radiation exposure to both the patients and operators.


Subject(s)
DNA Damage , Myocardial Perfusion Imaging/adverse effects , Radiation Dosage , Radiation Injuries/etiology , Tomography, Emission-Computed, Single-Photon/adverse effects , Adult , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins/blood , Biomarkers/blood , DNA Damage/genetics , Dose-Response Relationship, Radiation , Female , Flow Cytometry , Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Histones/blood , Humans , Male , Middle Aged , Myocardial Perfusion Imaging/methods , Phosphorylation , Predictive Value of Tests , RNA, Messenger/blood , Radiation Injuries/blood , Radiation Injuries/genetics , Radiopharmaceuticals/adverse effects , Risk Assessment , Risk Factors , Tumor Suppressor Protein p53/blood , Young Adult
12.
Circulation ; 130(11 Suppl 1): S60-9, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25200057

ABSTRACT

BACKGROUND: Despite the promise shown by stem cells for restoration of cardiac function after myocardial infarction, the poor survival of transplanted cells has been a major issue. Hypoxia-inducible factor-1 (HIF1) is a transcription factor that mediates adaptive responses to ischemia. Here, we hypothesize that codelivery of cardiac progenitor cells (CPCs) with a nonviral minicircle plasmid carrying HIF1 (MC-HIF1) into the ischemic myocardium can improve the survival of transplanted CPCs. METHODS AND RESULTS: After myocardial infarction, CPCs were codelivered intramyocardially into adult NOD/SCID mice with saline, MC-green fluorescent protein, or MC-HIF1 versus MC-HIF1 alone (n=10 per group). Bioluminescence imaging demonstrated better survival when CPCs were codelivered with MC-HIF1. Importantly, echocardiography showed mice injected with CPCs+MC-HIF1 had the highest ejection fraction 6 weeks after myocardial infarction (57.1±2.6%; P=0.002) followed by MC-HIF1 alone (48.5±2.6%; P=0.04), with no significant protection for CPCs+MC-green fluorescent protein (44.8±3.3%; P=NS) when compared with saline control (38.7±3.2%). In vitro mechanistic studies confirmed that cardiac endothelial cells produced exosomes that were actively internalized by recipient CPCs. Exosomes purified from endothelial cells overexpressing HIF1 had higher contents of miR-126 and miR-210. These microRNAs activated prosurvival kinases and induced a glycolytic switch in recipient CPCs, giving them increased tolerance when subjected to in vitro hypoxic stress. Inhibiting both of these miRs blocked the protective effects of the exosomes. CONCLUSIONS: In summary, HIF1 can be used to modulate the host microenvironment for improving survival of transplanted cells. The exosomal transfer of miRs from host cells to transplanted cells represents a unique mechanism that can be potentially targeted for improving survival of transplanted cells.


Subject(s)
Adult Stem Cells/transplantation , Exosomes , Genetic Therapy , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , MicroRNAs/therapeutic use , Multipotent Stem Cells/transplantation , Myocardial Infarction/therapy , Myocardial Ischemia/therapy , Animals , Cell Communication , Cellular Microenvironment , Combined Modality Therapy , Culture Media, Conditioned , DNA, Circular , Female , Genetic Vectors/therapeutic use , Graft Survival , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/surgery , Myocardial Ischemia/genetics , Myocardial Ischemia/surgery , Neovascularization, Physiologic , Plasmids , Random Allocation , Transfection
13.
J Am Coll Cardiol ; 64(5): 436-48, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25082575

ABSTRACT

BACKGROUND: Human-induced pluripotent stem cells (iPSCs) are a potentially unlimited source for generation of cardiomyocytes (iPSC-CMs). However, current protocols for iPSC-CM derivation face several challenges, including variability in somatic cell sources and inconsistencies in cardiac differentiation efficiency. OBJECTIVES: This study aimed to assess the effect of epigenetic memory on differentiation and function of iPSC-CMs generated from somatic cell sources of cardiac versus noncardiac origins. METHODS: Cardiac progenitor cells (CPCs) and skin fibroblasts from the same donors were reprogrammed into iPSCs and differentiated into iPSC-CMs via embryoid body and monolayer-based differentiation protocols. RESULTS: Differentiation efficiency was found to be higher in CPC-derived iPSC-CMs (CPC-iPSC-CMs) than in fibroblast-derived iPSC-CMs (Fib-iPSC-CMs). Gene expression analysis during cardiac differentiation demonstrated up-regulation of cardiac transcription factors in CPC-iPSC-CMs, including NKX2-5, MESP1, ISL1, HAND2, MYOCD, MEF2C, and GATA4. Epigenetic assessment revealed higher methylation in the promoter region of NKX2-5 in Fib-iPSC-CMs compared with CPC-iPSC-CMs. Epigenetic differences were found to dissipate with increased cell passaging, and a battery of in vitro assays revealed no significant differences in their morphological and electrophysiological properties at early passage. Finally, cell delivery into a small animal myocardial infarction model indicated that CPC-iPSC-CMs and Fib-iPSC-CMs possess comparable therapeutic capabilities in improving functional recovery in vivo. CONCLUSIONS: This is the first study to compare differentiation of iPSC-CMs from human CPCs versus human fibroblasts from the same donors. The authors demonstrate that although epigenetic memory improves differentiation efficiency of cardiac versus noncardiac somatic cell sources in vitro, it does not contribute to improved functional outcome in vivo.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/cytology , Stem Cell Transplantation , Tissue Donors , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/cytology
14.
Nat Commun ; 5: 3903, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24875164

ABSTRACT

The exact nature of the immune response elicited by autologous-induced pluripotent stem cell (iPSC) progeny is still not well understood. Here we show in murine models that autologous iPSC-derived endothelial cells (iECs) elicit an immune response that resembles the one against a comparable somatic cell, the aortic endothelial cell (AEC). These cells exhibit long-term survival in vivo and prompt a tolerogenic immune response characterized by elevated IL-10 expression. In contrast, undifferentiated iPSCs elicit a very different immune response with high lymphocytic infiltration and elevated IFN-γ, granzyme-B and perforin intragraft. Furthermore, the clonal structure of infiltrating T cells from iEC grafts is statistically indistinguishable from that of AECs, but is different from that of undifferentiated iPSC grafts. Taken together, our results indicate that the differentiation of iPSCs results in a loss of immunogenicity and leads to the induction of tolerance, despite expected antigen expression differences between iPSC-derived versus original somatic cells.


Subject(s)
Cell Differentiation/immunology , Endothelial Cells/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , Induced Pluripotent Stem Cells/transplantation , Self Tolerance/immunology , Animals , Aorta/cytology , Cells, Cultured , Endothelial Cells/cytology , Graft Survival , Granzymes/immunology , Induced Pluripotent Stem Cells/immunology , Interleukin-10/immunology , Mice , Perforin/immunology
15.
Stem Cells ; 31(11): 2354-63, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038578

ABSTRACT

RATIONALE: Human embryonic stem cell (hESC) derivatives are attractive candidates for therapeutic use. The engraftment and survival of hESC derivatives as xenografts or allografts require effective immunosuppression to prevent immune cell infiltration and graft destruction. OBJECTIVE: To test the hypothesis that a short-course, dual-agent regimen of two costimulation-adhesion blockade agents can induce better engraftment of hESC derivatives compared to current immunosuppressive agents. METHODS AND RESULTS: We transduced hESCs with a double fusion reporter gene construct expressing firefly luciferase (Fluc) and enhanced green fluorescent protein, and differentiated these cells to endothelial cells (hESC-ECs). Reporter gene expression enabled longitudinal assessment of cell engraftment by bioluminescence imaging. Costimulation-adhesion therapy resulted in superior hESC-EC and mouse EC engraftment compared to cyclosporine therapy in a hind limb model. Costimulation-adhesion therapy also promoted robust hESC-EC and hESC-derived cardiomyocyte survival in an ischemic myocardial injury model. Improved hESC-EC engraftment had a cardioprotective effect after myocardial injury, as assessed by magnetic resonance imaging. Mechanistically, costimulation-adhesion therapy is associated with systemic and intragraft upregulation of T-cell immunoglobulin and mucin domain 3 (TIM3) and a reduced proinflammatory cytokine profile. CONCLUSIONS: Costimulation-adhesion therapy is a superior alternative to current clinical immunosuppressive strategies for preventing the post-transplant rejection of hESC derivatives. By extending the window for cellular engraftment, costimulation-adhesion therapy enhances functional preservation following ischemic injury. This regimen may function through a TIM3-dependent mechanism.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cyclosporine/pharmacology , Embryonic Stem Cells/transplantation , Graft Rejection/prevention & control , Immunoconjugates/pharmacology , Immunosuppressive Agents/pharmacology , Prednisone/pharmacology , Abatacept , Animals , Cardiotonic Agents/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Endothelial Cells/immunology , Endothelial Cells/transplantation , Graft Rejection/immunology , Humans , Immune Tolerance , Immunosuppression Therapy/methods , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Random Allocation
16.
Circulation ; 128(11 Suppl 1): S3-13, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24030418

ABSTRACT

BACKGROUND: Drug-induced arrhythmia is one of the most common causes of drug development failure and withdrawal from market. This study tested whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with a low-impedance microelectrode array (MEA) system could improve on industry-standard preclinical cardiotoxicity screening methods, identify the effects of well-characterized drugs, and elucidate underlying risk factors for drug-induced arrhythmia. hiPSC-CMs may be advantageous over immortalized cell lines because they possess similar functional characteristics as primary human cardiomyocytes and can be generated in unlimited quantities. METHODS AND RESULTS: Pharmacological responses of beating embryoid bodies exposed to a comprehensive panel of drugs at 65 to 95 days postinduction were determined. Responses of hiPSC-CMs to drugs were qualitatively and quantitatively consistent with the reported drug effects in literature. Torsadogenic hERG blockers, such as sotalol and quinidine, produced statistically and physiologically significant effects, consistent with patch-clamp studies, on human embryonic stem cell-derived cardiomyocytes hESC-CMs. False-negative and false-positive hERG blockers were identified accurately. Consistent with published studies using animal models, early afterdepolarizations and ectopic beats were observed in 33% and 40% of embryoid bodies treated with sotalol and quinidine, respectively, compared with negligible early afterdepolarizations and ectopic beats in untreated controls. CONCLUSIONS: We found that drug-induced arrhythmias can be recapitulated in hiPSC-CMs and documented with low impedance MEA. Our data indicate that the MEA/hiPSC-CM assay is a sensitive, robust, and efficient platform for testing drug effectiveness and for arrhythmia screening. This system may hold great potential for reducing drug development costs and may provide significant advantages over current industry standard assays that use immortalized cell lines or animal models.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiovascular Agents/adverse effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Tissue Array Analysis/methods , Action Potentials/drug effects , Action Potentials/physiology , Adolescent , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Drug Evaluation, Preclinical/methods , Electric Impedance , Humans , Induced Pluripotent Stem Cells/physiology , Male , Microelectrodes , Myocytes, Cardiac/physiology
17.
Circulation ; 127(16): 1677-91, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23519760

ABSTRACT

BACKGROUND: Cardiotoxicity is a leading cause for drug attrition during pharmaceutical development and has resulted in numerous preventable patient deaths. Incidents of adverse cardiac drug reactions are more common in patients with preexisting heart disease than the general population. Here we generated a library of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients with various hereditary cardiac disorders to model differences in cardiac drug toxicity susceptibility for patients of different genetic backgrounds. METHODS AND RESULTS: Action potential duration and drug-induced arrhythmia were measured at the single cell level in hiPSC-CMs derived from healthy subjects and patients with hereditary long QT syndrome, familial hypertrophic cardiomyopathy, and familial dilated cardiomyopathy. Disease phenotypes were verified in long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy hiPSC-CMs by immunostaining and single cell patch clamp. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and the human ether-a-go-go-related gene expressing human embryonic kidney cells were used as controls. Single cell PCR confirmed expression of all cardiac ion channels in patient-specific hiPSC-CMs as well as hESC-CMs, but not in human embryonic kidney cells. Disease-specific hiPSC-CMs demonstrated increased susceptibility to known cardiotoxic drugs as measured by action potential duration and quantification of drug-induced arrhythmias such as early afterdepolarizations and delayed afterdepolarizations. CONCLUSIONS: We have recapitulated drug-induced cardiotoxicity profiles for healthy subjects, long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy patients at the single cell level for the first time. Our data indicate that healthy and diseased individuals exhibit different susceptibilities to cardiotoxic drugs and that use of disease-specific hiPSC-CMs may predict adverse drug responses more accurately than the standard human ether-a-go-go-related gene test or healthy control hiPSC-CM/hESC-CM screening assays.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic, Familial/genetics , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/genetics , Genetic Predisposition to Disease , Induced Pluripotent Stem Cells/cytology , Long QT Syndrome/genetics , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic, Familial/pathology , Cell Differentiation , Cell Line/drug effects , Cell Line/physiology , Cell Size , Cisapride/toxicity , Embryoid Bodies/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Profiling , HEK293 Cells/drug effects , HEK293 Cells/physiology , Humans , In Vitro Techniques , Ion Channels/biosynthesis , Ion Channels/genetics , Kidney/cytology , Kidney/embryology , Long QT Syndrome/pathology , Myocytes, Cardiac/physiology , Myofibrils/ultrastructure , Nicorandil/toxicity , Patch-Clamp Techniques , Quinazolines/toxicity , Verapamil/toxicity
18.
Cell Stem Cell ; 12(1): 101-13, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23290139

ABSTRACT

Familial hypertrophic cardiomyopathy (HCM) is a prevalent hereditary cardiac disorder linked to arrhythmia and sudden cardiac death. While the causes of HCM have been identified as genetic mutations in the cardiac sarcomere, the pathways by which sarcomeric mutations engender myocyte hypertrophy and electrophysiological abnormalities are not understood. To elucidate the mechanisms underlying HCM development, we generated patient-specific induced pluripotent stem cell cardiomyocytes (iPSC-CMs) from a ten-member family cohort carrying a hereditary HCM missense mutation (Arg663His) in the MYH7 gene. Diseased iPSC-CMs recapitulated numerous aspects of the HCM phenotype including cellular enlargement and contractile arrhythmia at the single-cell level. Calcium (Ca(2+)) imaging indicated dysregulation of Ca(2+) cycling and elevation in intracellular Ca(2+) ([Ca(2+)](i)) are central mechanisms for disease pathogenesis. Pharmacological restoration of Ca(2+) homeostasis prevented development of hypertrophy and electrophysiological irregularities. We anticipate that these findings will help elucidate the mechanisms underlying HCM development and identify novel therapies for the disease.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Cardiac Myosins/genetics , Cells, Cultured , Humans , Mutation, Missense , Myosin Heavy Chains/genetics
19.
Am J Pathol ; 182(2): 431-48, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23201090

ABSTRACT

Defects in urothelial integrity resulting in leakage and activation of underlying sensory nerves are potential causative factors of bladder pain syndrome, a clinical syndrome of pelvic pain and urinary urgency/frequency in the absence of a specific cause. Herein, we identified the microRNA miR-199a-5p as an important regulator of intercellular junctions. On overexpression in urothelial cells, it impairs correct tight junction formation and leads to increased permeability. miR-199a-5p directly targets mRNAs encoding LIN7C, ARHGAP12, PALS1, RND1, and PVRL1 and attenuates their expression levels to a similar extent. Using laser microdissection, we showed that miR-199a-5p is predominantly expressed in bladder smooth muscle but that it is also detected in mature bladder urothelium and primary urothelial cultures. In the urothelium, its expression can be up-regulated after activation of cAMP signaling pathways. While validating miR-199a-5p targets, we delineated novel functions of LIN7C and ARHGAP12 in urothelial integrity and confirmed the essential role of PALS1 in establishing and maintaining urothelial polarity and junction assembly. The present results point to a possible link between miR-199a-5p expression and the control of urothelial permeability in bladder pain syndrome. Up-regulation of miR-199a-5p and concomitant down-regulation of its multiple targets might be detrimental to the establishment of a tight urothelial barrier, leading to chronic pain.


Subject(s)
Cystitis, Interstitial/genetics , Cystitis, Interstitial/pathology , Urothelium/metabolism , Urothelium/pathology , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites/genetics , Cell Differentiation/genetics , Cell Line , Cyclic AMP/metabolism , Down-Regulation/genetics , Electric Impedance , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Luciferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Permeability , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tight Junction Proteins/metabolism , Up-Regulation/genetics , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder/ultrastructure , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
20.
Nat Protoc ; 7(5): 829-38, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22481529

ABSTRACT

Single-cell quantitative real-time PCR (qRT-PCR) combined with high-throughput arrays allows the analysis of gene expression profiles at a molecular level in approximately 11 h after cell sample collection. We present here a high-content microfluidic real-time platform as a powerful tool for comparatively investigating the regulation of developmental processes in single cells. This approach overcomes the limitations involving heterogeneous cell populations and sample amounts, and may shed light on differential regulation of gene expression in normal versus disease-related contexts. Furthermore, high-throughput single-cell qRT-PCR provides a standardized, comparative assay for in-depth analysis of the mechanisms underlying human pluripotent stem cell self-renewal and differentiation.


Subject(s)
Gene Expression Profiling/methods , Microfluidics/methods , Cells, Cultured , Embryonic Stem Cells/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Real-Time Polymerase Chain Reaction , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...