Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
iScience ; 25(2): 103806, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35198871

ABSTRACT

Dynamic profiling of changes in gene expression in response to stressors in specific microenvironments without requiring cellular destruction remains challenging. Current methodologies that seek to interrogate gene expression at a molecular level require sampling of cellular transcriptome and therefore lysis of the cell, preventing serial analysis of cellular transcriptome. To address this area of unmet need, we have recently developed a technology allowing transcriptomic analysis over time without cellular destruction. Our method, TRACE-seq (TRanscriptomic Analysis Captured in Extracellular vesicles using sequencing), is characterized by a cell-type specific transgene expression. It provides data on the transcriptome inside extracellular vesicles that provides an accurate representation of stress-responsive cellular transcriptomic changes. Thus, the transcriptome of cells expressing TRACE can be followed over time without destroying the source cell, which is a powerful tool for many fields of fundamental and translational biology research.

2.
iScience ; 23(3): 100959, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32179481

ABSTRACT

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

3.
Elife ; 82019 10 08.
Article in English | MEDLINE | ID: mdl-31591966

ABSTRACT

A fundamental goal in the biological sciences is to determine how individual cells with varied gene expression profiles and diverse functional characteristics contribute to development, physiology, and disease. Here, we report a novel strategy to assess gene expression and cell physiology in single living cells. Our approach utilizes fluorescently labeled mRNA-specific anti-sense RNA probes and dsRNA-binding protein to identify the expression of specific genes in real-time at single-cell resolution via FRET. We use this technology to identify distinct myocardial subpopulations expressing the structural proteins myosin heavy chain α and myosin light chain 2a in real-time during early differentiation of human pluripotent stem cells. We combine this live-cell gene expression analysis with detailed physiologic phenotyping to capture the functional evolution of these early myocardial subpopulations during lineage specification and diversification. This live-cell mRNA imaging approach will have wide ranging application wherever heterogeneity plays an important biological role.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Gene Expression Profiling/methods , Intravital Microscopy/methods , Single-Cell Analysis/methods , Cell Differentiation , Humans , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Staining and Labeling/methods
4.
Biotechnol Bioeng ; 116(11): 3098-3111, 2019 11.
Article in English | MEDLINE | ID: mdl-31317531

ABSTRACT

Here we propose a bio-MEMS device designed to evaluate contractile force and conduction velocity of cell sheets in response to mechanical and electrical stimulation of the cell source as it grows to form a cellular sheet. Moreover, the design allows for the incorporation of patient-specific data and cell sources. An optimized device would allow cell sheets to be cultured, characterized, and conditioned to be compatible with a specific patient's cardiac environment in vitro, before implantation. This design draws upon existing methods in the literature but makes an important advance by combining the mechanical and electrical stimulation into a single system for optimized cell sheet growth. The device has been designed to achieve cellular alignment, electrical stimulation, mechanical stimulation, conduction velocity readout, contraction force readout, and eventually cell sheet release. The platform is a set of comb electrical contacts consisting of three-dimensional walls made of polydimethylsiloxane and coated with electrically conductive metals on the tops of the walls. Not only do the walls serve as a method for stimulating cells that are attached to the top, but their geometry is tailored such that they are flexible enough to be bent by the cells and used to measure force. The platform can be stretched via a linear actuator setup, allowing for simultaneous electrical and mechanical stimulation that can be derived from patient-specific clinical data.


Subject(s)
Micro-Electrical-Mechanical Systems , Myocardial Contraction , Myocardium/metabolism , Tissue Engineering/instrumentation , Animals , Electric Stimulation , Humans
5.
Acta Biomater ; 83: 211-220, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30352286

ABSTRACT

Post-cardiac surgical sternal and epicardial adhesions increase the risk and complexity of cardiac re-operative surgeries, which represent a significant challenge for patients with the congenital cardiac disease. Bioresorbable membranes can serve as barriers to prevent postoperative adhesions. Herein, we fabricated a bioresorbable gelatin/polycaprolactone (GT/PCL) composite membrane via electrospinning. The membrane was characterized in terms of morphology, mechanical properties, and biocompatibility. We then evaluated its efficacy as a physical barrier to prevent cardiac operative adhesions in a rabbit model. Our results showed that the membrane had a nanofibrous structure and was sturdy enough to be handled for the surgical procedures. In vitro studies with rabbit cardiac fibroblasts demonstrated that the membrane was biocompatible and inhibited cell infiltration. Further application of the membrane in a rabbit cardiac adhesion model revealed that the membrane was resorbed gradually and effectively resisted the sternal and epicardial adhesions. Interestingly, six months after the operation, the GT/PCL membrane was completely resorbed with simultaneous ingrowth of host cells to form a natural barrier. Collectively, these results indicated that the GT/PCL membrane might be a suitable barrier to prevent sternal and epicardial adhesions and might be utilized as a novel pericardial substitute for cardiac surgery. STATEMENT OF SIGNIFICANCE: Electrospinning is a versatile method to prepare nanofibrous membranes for tissue engineering and regenerative medicine applications. However, with the micro-/nano-scale structure and high porosity, the electrospun membrane might be an excellent candidate as a barrier to prevent postoperative adhesion. Here we prepared an electropun GT/PCL nanofibrous membrane and applied it as a barrier to prevent sternal and epicardial adhesions. Our results showed that the membrane had sufficient mechanical strength, good biocompatibility, and effectively resisted the sternal and epicardial adhesions. What's more, the membrane was bioresorbable and allowed simultaneous ingrowth of host cells to form a natural barrier. We believe that the current will inspire more research on nanomaterials to prevent postoperative adhesion applications.


Subject(s)
Biocompatible Materials , Gelatin , Membranes, Artificial , Nanofibers , Polyesters , Tissue Adhesions/prevention & control , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Gelatin/chemistry , Gelatin/pharmacology , Myocardium/metabolism , Myocardium/pathology , Nanofibers/chemistry , Nanofibers/therapeutic use , Polyesters/chemistry , Polyesters/pharmacology , Rabbits , Tissue Adhesions/metabolism , Tissue Adhesions/pathology
6.
PLoS One ; 13(10): e0205108, 2018.
Article in English | MEDLINE | ID: mdl-30281646

ABSTRACT

Understanding the molecular pathways regulating cardiogenesis is crucial for the early diagnosis of heart diseases and improvement of cardiovascular disease. During normal mammalian cardiac development, collagen and calcium-binding EGF domain-1 (Ccbe1) is expressed in the first and second heart field progenitors as well as in the proepicardium, but its role in early cardiac commitment remains unknown. Here we demonstrate that during mouse embryonic stem cell (ESC) differentiation Ccbe1 is upregulated upon emergence of Isl1- and Nkx2.5- positive cardiac progenitors. Ccbe1 is markedly enriched in Isl1-positive cardiac progenitors isolated from ESCs differentiating in vitro or embryonic hearts developing in vivo. Disruption of Ccbe1 activity by shRNA knockdown or blockade with a neutralizing antibody results in impaired differentiation of embryonic stem cells along the cardiac mesoderm lineage resulting in a decreased expression of mature cardiomyocyte markers. In addition, knockdown of Ccbe1 leads to smaller embryoid bodies. Collectively, our results show that CCBE1 is essential for the commitment of cardiac mesoderm and consequently, for the formation of cardiac myocytes in differentiating mouse ESCs.


Subject(s)
Calcium-Binding Proteins/deficiency , Cell Differentiation/physiology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Calcium-Binding Proteins/genetics , Cells, Cultured , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Heart/embryology , Homeobox Protein Nkx-2.5/metabolism , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , RNA, Small Interfering , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
7.
Circ Res ; 123(9): 1066-1079, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30355156

ABSTRACT

RATIONALE: Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are a readily available, robustly reproducible, and physiologically appropriate human cell source for cardiac disease modeling, drug discovery, and toxicity screenings in vitro. However, unlike adult myocardial cells in vivo, hPSC-CMs cultured in vitro maintain an immature metabolic phenotype, where majority of ATP is produced through aerobic glycolysis instead of oxidative phosphorylation in the mitochondria. Little is known about the underlying signaling pathways controlling hPSC-CMs' metabolic and functional maturation. OBJECTIVE: To define the molecular pathways controlling cardiomyocytes' metabolic pathway selections and improve cardiomyocyte metabolic and functional maturation. METHODS AND RESULTS: We cultured hPSC-CMs in different media compositions including glucose-containing media, glucose-containing media supplemented with fatty acids, and glucose-free media with fatty acids as the primary carbon source. We found that cardiomyocytes cultured in the presence of glucose used primarily aerobic glycolysis and aberrantly upregulated HIF1α (hypoxia-inducible factor 1α) and its downstream target lactate dehydrogenase A. Conversely, glucose deprivation promoted oxidative phosphorylation and repressed HIF1α. Small molecule inhibition of HIF1α or lactate dehydrogenase A resulted in a switch from aerobic glycolysis to oxidative phosphorylation. Likewise, siRNA inhibition of HIF1α stimulated oxidative phosphorylation while inhibiting aerobic glycolysis. This metabolic shift was accompanied by an increase in mitochondrial content and cellular ATP levels. Furthermore, functional gene expressions, sarcomere length, and contractility were improved by HIF1α/lactate dehydrogenase A inhibition. CONCLUSIONS: We show that under standard culture conditions, the HIF1α-lactate dehydrogenase A axis is aberrantly upregulated in hPSC-CMs, preventing their metabolic maturation. Chemical or siRNA inhibition of this pathway results in an appropriate metabolic shift from aerobic glycolysis to oxidative phosphorylation. This in turn improves metabolic and functional maturation of hPSC-CMs. These findings provide key insight into molecular control of hPSC-CMs' metabolism and may be used to generate more physiologically mature cardiomyocytes for drug screening, disease modeling, and therapeutic purposes.


Subject(s)
Aminoquinolines/pharmacology , Cell Differentiation/drug effects , Disulfides/pharmacology , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Indole Alkaloids/pharmacology , Induced Pluripotent Stem Cells/drug effects , L-Lactate Dehydrogenase/antagonists & inhibitors , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Sulfonamides/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Induced Pluripotent Stem Cells/enzymology , L-Lactate Dehydrogenase/metabolism , Male , Mice, Inbred C57BL , Mitochondria, Heart/enzymology , Mitochondria, Heart/genetics , Myocytes, Cardiac/enzymology , Oxidative Phosphorylation/drug effects , Phenotype , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
8.
PLoS One ; 13(4): e0196697, 2018.
Article in English | MEDLINE | ID: mdl-29698509

ABSTRACT

AIMS: Atrial natriuretic peptide (ANP), secreted primarily by atrial cardiomyocytes, decreases blood pressure by raising cyclic 3',5'-guanosine monophosphate (cGMP) levels and inducing vasorelaxation, natriuresis, and diuresis. Raising the level of ANP has been shown to be an effective treatment for hypertension. To advance the future development of an anti-microRNA (miR) approach to increasing expression of ANP, we investigated the regulation of NPPA expression by two miRs: miR-425 and miR-155. We examined whether miR-425 and miR-155 have an additive effect on the expression and function of ANP. METHODS AND RESULTS: Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were transfected with miR-425, miR-155, or a combination of the two miRs. Two days later, NPPA expression was measured using real time qPCR. Each of the miRs decreased NPPA expression over a wide range of concentrations, with a significant reduction at concentrations as low as 1 nM. The combination of miR-425 and miR-155 reduced NPPA expression to a greater extent than either miR-425 or miR-155 alone. An in vitro assay was developed to study the potential biological significance of the miR-induced decrease in NPPA expression. The cooperative effect of miR-425 and miR-155 on NPPA expression was associated with a significant decrease in cGMP levels. CONCLUSIONS: These data demonstrate that miR-425 and miR-155 regulate NPPA expression in a cooperative manner. Targeting both miRNAs with anti-miRs (possibly at submaximal concentrations) might prove to be a more effective strategy to modulate ANP levels, and thus blood pressure, than targeting either miRNA alone.


Subject(s)
Atrial Natriuretic Factor/metabolism , Cyclic GMP/metabolism , MicroRNAs/metabolism , Animals , Atrial Natriuretic Factor/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Human Embryonic Stem Cells/cytology , Humans , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Transfection
9.
Biotechnol Bioeng ; 115(3): 630-644, 2018 03.
Article in English | MEDLINE | ID: mdl-29178315

ABSTRACT

Three-dimensional (3D) cultures of human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) hold great promise for drug discovery, providing a better approximation to the in vivo physiology over standard two-dimensional (2D) monolayer cultures. However, the transition of CM differentiation protocols from 2D to 3D cultures is not straightforward. In this work, we relied on the aggregation of hPSC-derived cardiac progenitors and their culture under agitated conditions to generate highly pure cardiomyocyte aggregates. Whole-transcriptome analysis and 13 C-metabolic flux analysis allowed to demonstrate at both molecular and fluxome levels that such 3D culture environment enhances metabolic maturation of hiPSC-CMs. When compared to 2D, 3D cultures of hiPSC-CMs displayed down-regulation of genes involved in glycolysis and lipid biosynthesis and increased expression of genes involved in OXPHOS. Accordingly, 3D cultures of hiPSC-CMs had lower fluxes through glycolysis and fatty acid synthesis and increased TCA-cycle activity. Importantly, we demonstrated that the 3D culture environment reproducibly improved both CM purity and metabolic maturation across different hPSC lines, thereby providing a robust strategy to derive enriched hPSC-CMs with metabolic features closer to that of adult CMs.


Subject(s)
Cell Culture Techniques/methods , Glycolysis , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Lipid Metabolism , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Cell Line , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology
10.
Circ Cardiovasc Genet ; 10(6)2017 Dec.
Article in English | MEDLINE | ID: mdl-29212899

ABSTRACT

BACKGROUND: Restrictive cardiomyopathy (RCM) is a rare cardiomyopathy characterized by impaired diastolic ventricular function resulting in a poor clinical prognosis. Rarely, heritable forms of RCM have been reported, and mutations underlying RCM have been identified in genes that govern the contractile function of the cardiomyocytes. METHODS AND RESULTS: We evaluated 8 family members across 4 generations by history, physical examination, electrocardiography, and echocardiography. Affected individuals presented with a pleitropic syndrome of progressive RCM, atrioventricular septal defects, and a high prevalence of atrial fibrillation. Exome sequencing of 5 affected members identified a single novel missense variant in a highly conserved residue of FLNC (filamin C; p.V2297M). FLNC encodes filamin C-a protein that acts as both a scaffold for the assembly and organization of the central contractile unit of striated muscle and also as a mechanosensitive signaling molecule during cell migration and shear stress. Immunohistochemical analysis of FLNC localization in cardiac tissue from an affected family member revealed a diminished localization at the z disk, whereas traditional localization at the intercalated disk was preserved. Stem cell-derived cardiomyocytes mutated to carry the effect allele had diminished contractile activity when compared with controls. CONCLUSION: We have identified a novel variant in FLNC as pathogenic variant for familial RCM-a finding that further expands on the genetic basis of this rare and morbid cardiomyopathy.


Subject(s)
Cardiomyopathy, Restrictive/genetics , Filamins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Adult , Aged , Amino Acid Sequence , Base Sequence , Cardiomyopathy, Restrictive/pathology , Family , Female , Filamins/chemistry , Humans , Male , Middle Aged , Phenotype
11.
Curr Protoc Stem Cell Biol ; 43: 1F.20.1-1F.20.9, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29140569

ABSTRACT

Human pluripotent stem-cell derived cardiomyocytes (hPSC-CMs) hold great promise for applications in human disease modeling, drug discovery, cardiotoxicity screening, and, ultimately, regenerative medicine. The ability to study multiple parameters of hPSC-CM function, such as contractile and electrical activity, calcium cycling, and force generation, is therefore of paramount importance. hPSC-CMs cultured on stiff substrates like glass or polystyrene do not have the ability to shorten during contraction, making them less suitable for the study of hPSC-CM contractile function. Other approaches require highly specialized hardware and are difficult to reproduce. Here we describe a protocol for the preparation of hPSC-CMs on soft substrates that enable shortening, and subsequently the simultaneous quantitative analysis of their contractile and electrical activity, calcium cycling, and force generation at single-cell resolution. This protocol requires only affordable and readily available materials and works with standard imaging hardware. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Cell Culture Techniques/methods , Myocytes, Cardiac , Pluripotent Stem Cells , Glass/chemistry , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Polystyrenes/chemistry
12.
Sci Transl Med ; 9(415)2017 Nov 08.
Article in English | MEDLINE | ID: mdl-29118264

ABSTRACT

In response to heart failure (HF), the heart reacts by repressing adult genes and expressing fetal genes, thereby returning to a more fetal-like gene profile. To identify genes involved in this process, we carried out transcriptional analysis on murine hearts at different stages of development and on hearts from adult mice with HF. Our screen identified Oplah, encoding for 5-oxoprolinase, a member of the γ-glutamyl cycle that functions by scavenging 5-oxoproline. OPLAH depletion occurred as a result of cardiac injury, leading to elevated 5-oxoproline and oxidative stress, whereas OPLAH overexpression improved cardiac function after ischemic injury. In HF patients, we observed elevated plasma 5-oxoproline, which was associated with a worse clinical outcome. Understanding and modulating fetal-like genes in the failing heart may lead to potential diagnostic, prognostic, and therapeutic options in HF.


Subject(s)
Cardiotonic Agents/metabolism , Myocardium/metabolism , Myocardium/pathology , Pyroglutamate Hydrolase/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Animals , Fetus/metabolism , Heart Failure/blood , Heart Failure/pathology , Heart Failure/physiopathology , Heart Function Tests , Humans , Mice, Transgenic , Myocardial Infarction/blood , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Oxidative Stress , Pyrrolidonecarboxylic Acid/blood , Rats , Receptors, Estrogen/metabolism , Reperfusion Injury/blood , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Sequence Analysis, RNA , Stress, Mechanical , Transcription, Genetic , ERRalpha Estrogen-Related Receptor
13.
Sci Rep ; 7(1): 8590, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819274

ABSTRACT

The immature phenotype of human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) constrains their potential in cell therapy and drug testing. In this study, we report that shifting hPSC-CMs from glucose-containing to galactose- and fatty acid-containing medium promotes their fast maturation into adult-like CMs with higher oxidative metabolism, transcriptional signatures closer to those of adult ventricular tissue, higher myofibril density and alignment, improved calcium handling, enhanced contractility, and more physiological action potential kinetics. Integrated "-Omics" analyses showed that addition of galactose to culture medium improves total oxidative capacity of the cells and ameliorates fatty acid oxidation avoiding the lipotoxicity that results from cell exposure to high fatty acid levels. This study provides an important link between substrate utilization and functional maturation of hPSC-CMs facilitating the application of this promising cell type in clinical and preclinical applications.


Subject(s)
Carbon/pharmacology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Biomarkers/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Fatty Acids/toxicity , Galactose/pharmacology , Glucose/deficiency , Glycolysis/drug effects , Heart Ventricles/cytology , Humans , Kinetics , Lactose/pharmacology , Models, Biological , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/ultrastructure , Transcription, Genetic/drug effects , Transcriptome/genetics
14.
Trends Cell Biol ; 27(5): 352-364, 2017 05.
Article in English | MEDLINE | ID: mdl-28007424

ABSTRACT

The advent of human pluripotent stem cell (hPSC) biology has opened unprecedented opportunities for the use of tissue engineering to generate human cardiac tissue for in vitro study. Engineering cardiac constructs that recapitulate human development and disease requires faithful recreation of the cardiac niche in vitro. Here we discuss recent progress in translating the in vivo cardiac microenvironment into PSC models of the human heart. We review three key physiologic features required to recreate the cardiac niche and facilitate normal cardiac differentiation and maturation: the biochemical, biophysical, and bioelectrical signaling cues. Finally, we discuss key barriers that must be overcome to fulfill the promise of stem cell biology in preclinical applications and ultimately in clinical practice.


Subject(s)
Cellular Microenvironment , Disease , Models, Biological , Physiological Phenomena , Pluripotent Stem Cells/cytology , Humans
15.
Adv Healthc Mater ; 6(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-27774763

ABSTRACT

In this essay the authors argue that chamber pressure dominates the biomechanics of the contraction cycle of the heart, while tissue stiffness dominates the relaxation cycle. This appears to be an under-recognized challenge in cardiac tissue engineering. Optimal approaches will involve constructing chambers or modulating the stiffness of the scaffold/substrate in synchrony with the beating cycle.


Subject(s)
Myocardium , Tissue Engineering/methods , Tissue Scaffolds , Animals , Humans
16.
Stem Cell Reports ; 7(6): 1037-1049, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27818139

ABSTRACT

The transcriptional regulator CITED2 is essential for heart development. Here, we investigated the role of CITED2 in the specification of cardiac cell fate from mouse embryonic stem cells (ESC). The overexpression of CITED2 in undifferentiated ESC was sufficient to promote cardiac cell emergence upon differentiation. Conversely, the depletion of Cited2 at the onset of differentiation resulted in a decline of ESC ability to generate cardiac cells. Moreover, loss of Cited2 expression impairs the expression of early mesoderm markers and cardiogenic transcription factors (Isl1, Gata4, Tbx5). The cardiogenic defects in Cited2-depleted cells were rescued by treatment with recombinant CITED2 protein. We showed that Cited2 expression is enriched in cardiac progenitors either derived from ESC or mouse embryonic hearts. Finally, we demonstrated that CITED2 and ISL1 proteins interact physically and cooperate to promote ESC differentiation toward cardiomyocytes. Collectively, our results show that Cited2 plays a pivotal role in cardiac commitment of ESC.


Subject(s)
Cell Differentiation , LIM-Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Lineage , Gene Expression Regulation, Developmental , Humans , Mesoderm/metabolism , Mice , Protein Binding , Repressor Proteins/genetics , Trans-Activators/genetics
17.
Sci Transl Med ; 8(342): 342ps13, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27280684

ABSTRACT

The promise of cardiac tissue engineering is in the ability to recapitulate in vitro the functional aspects of a healthy heart and disease pathology as well as to design replacement muscle for clinical therapy. Parts of this promise have been realized; others have not. In a meeting of scientists in this field, five central challenges or "big questions" were articulated that, if addressed, could substantially advance the current state of the art in modeling heart disease and realizing heart repair.


Subject(s)
Myocardium/cytology , Tissue Engineering/methods , Animals , Heart Diseases/metabolism , Humans , Myocardium/metabolism , Myocytes, Cardiac/cytology
18.
Mol Cell Biol ; 36(14): 1977-87, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27185878

ABSTRACT

Atrial natriuretic peptide (ANP) has a central role in regulating blood pressure in humans. Recently, microRNA 425 (miR-425) was found to regulate ANP production by binding to the mRNA of NPPA, the gene encoding ANP. mRNAs typically contain multiple predicted microRNA (miRNA)-binding sites, and binding of different miRNAs may independently or coordinately regulate the expression of any given mRNA. We used a multifaceted screening strategy that integrates bioinformatics, next-generation sequencing data, human genetic association data, and cellular models to identify additional functional NPPA-targeting miRNAs. Two novel miRNAs, miR-155 and miR-105, were found to modulate ANP production in human cardiomyocytes and target genetic variants whose minor alleles are associated with higher human plasma ANP levels. Both miR-155 and miR-105 repressed NPPA mRNA in an allele-specific manner, with the minor allele of each respective variant conferring resistance to the miRNA either by disruption of miRNA base pairing or by creation of wobble base pairing. Moreover, miR-155 enhanced the repressive effects of miR-425 on ANP production in human cardiomyocytes. Our study combines computational, genomic, and cellular tools to identify novel miRNA regulators of ANP production that could be targeted to raise ANP levels, which may have applications for the treatment of hypertension or heart failure.


Subject(s)
Atrial Natriuretic Factor/genetics , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Alleles , Atrial Natriuretic Factor/metabolism , Blood Pressure , Cells, Cultured , Down-Regulation , Female , Genetic Variation , Humans , Placenta/metabolism , Pregnancy
19.
Cell Rep ; 14(7): 1662-1672, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876178

ABSTRACT

A hallmark of cardiac development is the formation of myocardial trabeculations exclusively from the luminal surface of the primitive heart tube. Although a number of genetic defects in the endocardium and cardiac jelly disrupt myocardial trabeculation, the role of cell polarization remains unclear. Here, we demonstrate that atypical protein kinase C iota (Prkci) and its interacting partners are localized primarily to the luminal side of myocardial cells of early murine embryonic hearts. A subset of these cells undergoes polarized cell division with the cell division plane perpendicular to the heart's lumen. Disruption of the cell polarity complex by targeted gene mutations results in aberrant mitotic spindle alignment, loss of polarized cardiomyocyte division, and loss of normal myocardial trabeculation. Collectively, these results suggest that, in response to inductive signals, Prkci and its downstream partners direct polarized cell division of luminal myocardial cells to drive trabeculation in the nascent heart.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Division/genetics , Isoenzymes/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Protein Kinase C/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens/genetics , Antigens/metabolism , Cell Polarity , Embryo, Mammalian , Endocardium/embryology , Endocardium/metabolism , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Isoenzymes/metabolism , Mice , Myocytes, Cardiac/ultrastructure , Protein Binding , Protein Kinase C/metabolism , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Troponin T/genetics , Troponin T/metabolism
20.
J Am Coll Cardiol ; 67(7): 804-812, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26892417

ABSTRACT

BACKGROUND: The cardiac natriuretic peptides (NPs), atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP), have central roles in sodium and blood pressure regulation. Extracardiac factors (e.g., obesity and diabetes) influence NP production, potentially altering cardiovascular responses to volume and pressure stress. OBJECTIVES: This study examined the effects of acute carbohydrate intake on the NP system in humans, and investigated underlying mechanisms. METHODS: Normotensive subjects (N = 33) were given a high-carbohydrate shake. Venous blood was sampled to measure N-terminal (NT)-proANP and NT-proBNP levels. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and HepG2 cells were treated with glucose, and expression levels of NPs and micro ribonucleic acid 425 (miR-425), a negative regulator of ANP, were examined. The role of nuclear factor kappa B (NF-κB) in the glucose-mediated effects was investigated using a NF-κB inhibitor and expression plasmids encoding NF-κB subunits. RESULTS: We observed a 27% reduction in the levels of circulating NT-proANP (p < 0.001, maximal at 6 h) after carbohydrate challenge, with no effect on NT-proBNP levels in our human subjects. Glucose treatment of hESC-CMs for 6 h and 24 h increased levels of the primary transcript of miR-425 (pri-miR-425) and mature miR-425. A corresponding decrease in NPPA messenger RNA levels was also observed at both time points. Overexpression of NF-κB subunits in H9c2 cardiomyocytes increased miR-425 levels, whereas inhibition of NF-κB abrogated the glucose-mediated increase in pri-miR-425 levels in HepG2 cells. CONCLUSIONS: Acute carbohydrate challenge is associated with a reduction in ANP production. The mechanism appears to involve a glucose-induced increase in the expression of miR-425, mediated by NF-κB signaling.


Subject(s)
Blood Pressure/physiology , Myocytes, Cardiac/metabolism , Natriuretic Peptides/genetics , Obesity/metabolism , Sodium/metabolism , Adult , Animals , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/genetics , Female , Gene Expression Regulation , Hep G2 Cells/metabolism , Humans , Male , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/biosynthesis , Natriuretic Peptide, Brain/genetics , Natriuretic Peptides/biosynthesis , Obesity/genetics , Obesity/pathology , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Protein Precursors , RNA, Messenger/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...