Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Syst ; 12(6): 670-687.e10, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34043964

ABSTRACT

Although some cell types may be defined anatomically or by physiological function, a rigorous definition of cell state remains elusive. Here, we develop a quantitative, imaging-based platform for the systematic and automated classification of subcellular organization in single cells. We use this platform to quantify subcellular organization and gene expression in >30,000 individual human induced pluripotent stem cell-derived cardiomyocytes, producing a publicly available dataset that describes the population distributions of local and global sarcomere organization, mRNA abundance, and correlations between these traits. While the mRNA abundance of some phenotypically important genes correlates with subcellular organization (e.g., the beta-myosin heavy chain, MYH7), these two cellular metrics are heterogeneous and often uncorrelated, which suggests that gene expression alone is not sufficient to classify cell states. Instead, we posit that cell state should be defined by observing full distributions of quantitative, multidimensional traits in single cells that also account for space, time, and function.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation/genetics , Humans , Myocytes, Cardiac/metabolism , Transcriptome/genetics
2.
Circulation ; 140(20): 1647-1660, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31587567

ABSTRACT

BACKGROUND: The giant sarcomere protein titin is important in both heart health and disease. Mutations in the gene encoding for titin (TTN) are the leading known cause of familial dilated cardiomyopathy. The uneven distribution of these mutations within TTN motivated us to seek a more complete understanding of this gene and the isoforms it encodes in cardiomyocyte (CM) sarcomere formation and function. METHODS: To investigate the function of titin in human CMs, we used CRISPR/Cas9 to generate homozygous truncations in the Z disk (TTN-Z-/-) and A-band (TTN-A-/-) regions of the TTN gene in human induced pluripotent stem cells. The resulting CMs were characterized with immunostaining, engineered heart tissue mechanical measurements, and single-cell force and calcium measurements. RESULTS: After differentiation, we were surprised to find that despite the more upstream mutation, TTN-Z-/--CMs had sarcomeres and visibly contracted, whereas TTN-A-/--CMs did not. We hypothesized that sarcomere formation was caused by the expression of a recently discovered isoform of titin, Cronos, which initiates downstream of the truncation in TTN-Z-/--CMs. Using a custom Cronos antibody, we demonstrate that this isoform is expressed and integrated into myofibrils in human CMs. TTN-Z-/--CMs exclusively express Cronos titin, but these cells produce lower contractile force and have perturbed myofibril bundling compared with controls expressing both full-length and Cronos titin. Cronos titin is highly expressed in human fetal cardiac tissue, and when knocked out in human induced pluripotent stem cell derived CMs, these cells exhibit reduced contractile force and myofibrillar disarray despite the presence of full-length titin. CONCLUSIONS: We demonstrate that Cronos titin is expressed in developing human CMs and is able to support partial sarcomere formation in the absence of full-length titin. Furthermore, Cronos titin is necessary for proper sarcomere function in human induced pluripotent stem cell derived CMs. Additional investigation is necessary to understand the molecular mechanisms of this novel isoform and how it contributes to human cardiac disease.


Subject(s)
Connectin/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , CRISPR-Cas Systems , Calcium Signaling , Cells, Cultured , Connectin/genetics , Fetal Heart/metabolism , Gene Editing , Genotype , Humans , Mutation , Myocardial Contraction/genetics , Phenotype
3.
Cell Rep ; 20(7): 1597-1608, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813672

ABSTRACT

We analyzed chromatin dynamics and transcriptional activity of human embryonic stem cell (hESC)-derived cardiac progenitor cells (CPCs) and KDR+/CD34+ endothelial cells generated from different mesodermal origins. Using an unbiased algorithm to hierarchically rank genes modulated at the level of chromatin and transcription, we identified candidate regulators of mesodermal lineage determination. HOPX, a non-DNA-binding homeodomain protein, was identified as a candidate regulator of blood-forming endothelial cells. Using HOPX reporter and knockout hESCs, we show that HOPX regulates blood formation. Loss of HOPX does not impact endothelial fate specification but markedly reduces primitive hematopoiesis, acting at least in part through failure to suppress Wnt/ß-catenin signaling. Thus, chromatin state analysis permits identification of regulators of mesodermal specification, including a conserved role for HOPX in governing primitive hematopoiesis.


Subject(s)
Chromatin/metabolism , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/metabolism , Mesoderm/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Algorithms , CRISPR-Cas Systems , Cell Differentiation , Cell Lineage/genetics , Chromatin/chemistry , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Genes, Reporter , Human Embryonic Stem Cells/cytology , Humans , Mesoderm/cytology , Mesoderm/growth & development , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Tumor Suppressor Proteins/deficiency , beta Catenin/genetics , beta Catenin/metabolism
4.
Nat Protoc ; 12(1): 15-31, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27906170

ABSTRACT

Human pluripotent stem cells (hPSCs) provide a valuable model for the study of human development and a means to generate a scalable source of cells for therapeutic applications. This protocol specifies cell fate efficiently into cardiac and endothelial lineages from hPSCs. The protocol takes 2 weeks to complete and requires experience in hPSC culture and differentiation techniques. Building on lessons taken from early development, this monolayer-directed differentiation protocol uses different concentrations of activin A and bone morphogenetic protein 4 (BMP4) to polarize cells into mesodermal subtypes that reflect mid-primitive-streak cardiogenic mesoderm and posterior-primitive-streak hemogenic mesoderm. This differentiation platform provides a basis for generating distinct cardiovascular progenitor populations that enable the derivation of cardiomyocytes and functionally distinct endothelial cell (EC) subtypes from cardiogenic versus hemogenic mesoderm with high efficiency without cell sorting. ECs derived from cardiogenic and hemogenic mesoderm can be matured into >90% CD31+/VE-cadherin+ definitive ECs. To test the functionality of ECs at different stages of differentiation, we provide methods for assaying the blood-forming potential and de novo lumen-forming activity of ECs. To our knowledge, this is the first protocol that provides a common platform for directed differentiation of cardiomyocytes and endothelial subtypes from hPSCs. This protocol yields endothelial differentiation efficiencies exceeding those of previously published protocols. Derivation of these cell types is a critical step toward understanding the basis of disease and generating cells with therapeutic potential.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Endothelial Cells/cytology , Mesoderm/cytology , Myocardium/cytology , Pluripotent Stem Cells/cytology , Cell Line , Cell Lineage , Humans
5.
Front Physiol ; 7: 566, 2016.
Article in English | MEDLINE | ID: mdl-27920728

ABSTRACT

Recent technological and protocol developments have greatly increased the ability to utilize stem cells transformed into cardiomyocytes as models to study human heart muscle development and how this is affected by disease associated mutations in a variety of sarcomere proteins. In this perspective we provide an overview of these emerging technologies and how they are being used to create better models of "disease in a dish" for both research and screening assays. We also consider the value of these assays as models to explore the seminal processes in initiation of the disease development and the possibility of early interventions.

6.
Stem Cell Reports ; 6(6): 885-896, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27161364

ABSTRACT

Tension production and contractile properties are poorly characterized aspects of excitation-contraction coupling of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Previous approaches have been limited due to the small size and structural immaturity of early-stage hiPSC-CMs. We developed a substrate nanopatterning approach to produce hiPSC-CMs in culture with adult-like dimensions, T-tubule-like structures, and aligned myofibrils. We then isolated myofibrils from hiPSC-CMs and measured the tension and kinetics of activation and relaxation using a custom-built apparatus with fast solution switching. The contractile properties and ultrastructure of myofibrils more closely resembled human fetal myofibrils of similar gestational age than adult preparations. We also demonstrated the ability to study the development of contractile dysfunction of myofibrils from a patient-derived hiPSC-CM cell line carrying the familial cardiomyopathy MYH7 mutation (E848G). These methods can bring new insights to understanding cardiomyocyte maturation and developmental mechanical dysfunction of hiPSC-CMs with cardiomyopathic mutations.


Subject(s)
Excitation Contraction Coupling/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Myofibrils/physiology , Biomechanical Phenomena , Cardiac Myosins/genetics , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cell Differentiation , Cell Line , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Kinetics , Mutation , Myocytes, Cardiac/cytology , Myofibrils/ultrastructure , Myosin Heavy Chains/genetics , Nanostructures/chemistry , Primary Cell Culture
SELECTION OF CITATIONS
SEARCH DETAIL
...