Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Sci ; 169(1): 280-292, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31059573

ABSTRACT

Alcohol use prior to and during pregnancy remains a significant societal problem and can lead to developmental fetal abnormalities including compromised myocardia function and increased risk for heart disease later in life. Alcohol-induced cardiac toxicity has traditionally been studied in animal-based models. These models have limitations due to physiological differences from human cardiomyocytes (CMs) and are also not suitable for high-throughput screening. We hypothesized that human-induced pluripotent stem cell-derived CMs (hiPSC-CMs) could serve as a useful tool to study alcohol-induced cardiac defects and/or toxicity. In this study, hiPSC-CMs were treated with ethanol at doses corresponding to the clinically relevant levels of alcohol intoxication. hiPSC-CMs exposed to ethanol showed a dose-dependent increase in cellular damage and decrease in cell viability, corresponding to increased production of reactive oxygen species. Furthermore, ethanol exposure also generated dose-dependent increased irregular Ca2+ transients and contractility in hiPSC-CMs. RNA-seq analysis showed significant alteration in genes belonging to the potassium voltage-gated channel family or solute carrier family, partially explaining the irregular Ca2+ transients and contractility in ethanol-treated hiPSC-CMs. RNA-seq also showed significant upregulation in the expression of genes associated with collagen and extracellular matrix modeling, and downregulation of genes involved in cardiovascular system development and actin filament-based process. These results suggest that hiPSC-CMs can be a novel and physiologically relevant system for the study of alcohol-induced cardiac toxicity.


Subject(s)
Ethanol/toxicity , Heart Diseases/chemically induced , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Calcium Signaling/drug effects , Cardiotoxicity , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Risk Assessment
2.
Acta Biomater ; 71: 235-246, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29505894

ABSTRACT

Despite recent advances in tissue engineered heart valves (TEHV), a major challenge is identifying a cell source for seeding TEHV scaffolds. Native heart valves are durable because valve interstitial cells (VICs) maintain tissue homeostasis by synthesizing and remodeling the extracellular matrix. This study demonstrates that induced pluripotent stem cells (iPSC)-derived mesenchymal stem cells (iMSCs) can be derived from iPSCs using a feeder-free protocol and then further matured into VICs by encapsulation within 3D hydrogels. The differentiation efficiency was characterized using flow cytometry, immunohistochemistry staining, and trilineage differentiation. Using our feeder-free differentiation protocol, iMSCs were differentiated from iPSCs and had CD90+, CD44+, CD71+, αSMA+, and CD45- expression. Furthermore, iMSCs underwent trilineage differentiation when cultured in induction media for 21 days. iMSCs were then encapsulated in poly(ethylene glycol)diacrylate (PEGDA) hydrogels grafted with adhesion peptide (RGDS) to promote remodeling and further maturation into VIC-like cells. VIC phenotype was assessed by the expression of alpha-smooth muscle actin (αSMA), vimentin, and collagen production after 28 days. When MSC-derived cells were encapsulated in PEGDA hydrogels that mimic the leaflet modulus, a decrease in αSMA expression and increase in vimentin was observed. In addition, iMSCs synthesized collagen type I after 28 days in 3D hydrogel culture. Thus, the results from this study suggest that iMSCs may be a promising cell source for TEHV. STATEMENT OF SIGNIFICANCE: Developing a suitable cell source is a critical component for the success and durability of tissue engineered heart valves. The significance of this study is the generation of iPSCs-derived mesenchymal stem cells (iMSCs) that have the capacity to mature into valve interstitial-like cells when introduced into a 3D cell culture designed to mimic the layers of the valve leaflet. iMSCs were generated using a feeder-free protocol, which is one major advantage over other methods, as it is more clinically relevant. In addition to generating a potential new cell source for heart valve tissue engineering, this study also highlights the importance of a 3D culture environment to influence cell phenotype and function.


Subject(s)
Cell Differentiation , Cells, Immobilized/metabolism , Heart Valves/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Polyethylene Glycols/chemistry , Antigens, Differentiation/biosynthesis , Cells, Immobilized/cytology , Heart Valves/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology
3.
Stem Cell Reports ; 9(2): 513-527, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28793247

ABSTRACT

Understanding molecules involved in differentiation of human pluripotent stem cells (hPSCs) into cardiomyocytes and endothelial cells is important in advancing hPSCs for cell therapy and drug testing. Here, we report that LGR5, a leucine-rich repeat-containing G-protein-coupled receptor, plays a critical role in hPSC differentiation into cardiomyocytes and endothelial cells. LGR5 expression was transiently upregulated during the early stage of cardiomyocyte differentiation, and knockdown of LGR5 resulted in reduced expression of cardiomyocyte-associated markers and poor cardiac differentiation. In contrast, knockdown of LGR5 promoted differentiation of endothelial-like cells with increased expression of endothelial cell markers and appropriate functional characteristics, including the ability to form tube-like structures and to take up acetylated low-density lipoproteins. Furthermore, knockdown of LGR5 significantly reduced the proliferation of differentiated cells and increased the nuclear translocation of ß-catenin and expression of Wnt signaling-related genes. Therefore, regulation of LGR5 may facilitate efficient generation of cardiomyocytes or endothelial cells from hPSCs.


Subject(s)
Cell Differentiation/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Body Patterning/genetics , Cell Proliferation , Gene Knockdown Techniques , Humans , Mesoderm/cytology , Mesoderm/embryology , Wnt Signaling Pathway
4.
Adv Exp Med Biol ; 951: 123-135, 2016.
Article in English | MEDLINE | ID: mdl-27837559

ABSTRACT

In recent years, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as a vital cell source for in vitro modeling of genetic cardiovascular disorders, drug screening, and in vivo cardiac regeneration research. Looking forward, the ability to efficiently cryopreserve hPSC-CMs without compromising their normal biochemical and physiologic functions will dramatically facilitate their various biomedical applications. Although working protocols for freezing, storing, and thawing hPSC-CMs have been established, the question remains as to whether they are optimal. In this chapter, we discuss our current understanding of cryopreservation appertaining to hPSC-CMs, and proffer key questions regarding the mechanical, contractile, and regenerative properties of cryopreserved hPSC-CMs.


Subject(s)
Cryopreservation/methods , Microvascular Angina/therapy , Myocardial Ischemia/therapy , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cryopreservation/instrumentation , Cryoprotective Agents/pharmacology , Dimethyl Sulfoxide/pharmacology , Disease Models, Animal , Drug Discovery , Drugs, Investigational/pharmacology , Humans , Mice , Microvascular Angina/pathology , Myocardial Ischemia/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Myocytes, Cardiac/transplantation , Pluripotent Stem Cells/physiology , Regeneration/physiology
5.
Sci Rep ; 6: 30956, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27492371

ABSTRACT

Efficient generation of cardiomyocytes from human pluripotent stem cells is critical for their regenerative applications. Microgravity and 3D culture can profoundly modulate cell proliferation and survival. Here, we engineered microscale progenitor cardiac spheres from human pluripotent stem cells and exposed the spheres to simulated microgravity using a random positioning machine for 3 days during their differentiation to cardiomyocytes. This process resulted in the production of highly enriched cardiomyocytes (99% purity) with high viability (90%) and expected functional properties, with a 1.5 to 4-fold higher yield of cardiomyocytes from each undifferentiated stem cell as compared with 3D-standard gravity culture. Increased induction, proliferation and viability of cardiac progenitors as well as up-regulation of genes associated with proliferation and survival at the early stage of differentiation were observed in the 3D culture under simulated microgravity. Therefore, a combination of 3D culture and simulated microgravity can be used to efficiently generate highly enriched cardiomyocytes.


Subject(s)
Computer Simulation , Myoblasts, Cardiac/physiology , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Organ Culture Techniques , Tissue Engineering , Weightlessness Simulation
6.
Dis Model Mech ; 9(9): 927-39, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27491078

ABSTRACT

Although ß-blockers can be used to eliminate stress-induced ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT), this treatment is unsuccessful in ∼25% of cases. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) generated from these patients have potential for use in investigating the phenomenon, but it remains unknown whether they can recapitulate patient-specific drug responses to ß-blockers. This study assessed whether the inadequacy of ß-blocker therapy in an individual can be observed in vitro using patient-derived CPVT iPSC-CMs. An individual with CPVT harboring a novel mutation in the type 2 cardiac ryanodine receptor (RyR2) was identified whose persistent ventricular arrhythmias during ß-blockade with nadolol were abolished during flecainide treatment. iPSC-CMs generated from this patient and two control individuals expressed comparable levels of excitation-contraction genes, but assessment of the sarcoplasmic reticulum Ca(2+) leak and load relationship revealed intracellular Ca(2+) homeostasis was altered in the CPVT iPSC-CMs. ß-adrenergic stimulation potentiated spontaneous Ca(2+) waves and unduly frequent, large and prolonged Ca(2+) sparks in CPVT compared with control iPSC-CMs, validating the disease phenotype. Pursuant to the patient's in vivo responses, nadolol treatment during ß-adrenergic stimulation achieved negligible reduction of Ca(2+) wave frequency and failed to rescue Ca(2+) spark defects in CPVT iPSC-CMs. In contrast, flecainide reduced both frequency and amplitude of Ca(2+) waves and restored the frequency, width and duration of Ca(2+) sparks to baseline levels. By recapitulating the improved response of an individual with CPVT to flecainide compared with ß-blocker therapy in vitro, these data provide new evidence that iPSC-CMs can capture basic components of patient-specific drug responses.


Subject(s)
Catecholamines/metabolism , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/pathology , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/physiopathology , Biomarkers/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Electrophysiological Phenomena/drug effects , Female , Flecainide/pharmacology , Flecainide/therapeutic use , Homeostasis/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Male , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pedigree , Receptors, Adrenergic, beta/metabolism , Tachycardia, Ventricular/physiopathology
7.
Inflamm Res ; 64(10): 809-815, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26275932

ABSTRACT

BACKGROUND AND OBJECTIVE: Omega-3 fatty acids, such as α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are polyunsaturated fatty acids (PUFA) that have long been associated with anti-inflammatory activity and general benefit toward human health. Over the last decade, the identification of a family of cell-surface G protein-coupled receptors that bind and are activated by free-fatty acids, including omega-3 fatty acids, suggest that many effects of PUFA are receptor-mediated. One such receptor, free-fatty acid receptor-4 (FFAR4), previously described as GPR120, has been shown to modulate anti-inflammatory and insulin-sensitizing effects in response to PUFA such as ALA and DHA. Additionally, FFAR4 stimulates secretion of the insulin secretagogue glucagon-like peptide-1 (GLP-1) from the GI tract and acts as a dietary sensor to regulate energy availability. The aim of the current study was to assess the effects of dietary omega-3 fatty acid supplementation on FFAR4 expression in the rat colon. METHODS: Sprague-Dawley rats were fed control soybean oil diets or alternatively, diets supplemented with either fish oil, which is enriched in DHA and EPA, or flaxseed oil, which is enriched in ALA, for 7 weeks. GLP-1 and blood glucose levels were monitored weekly and at the end of the study period, expression of FFAR4 and the inflammatory marker TNF-α was assessed. RESULTS: Our findings indicate that GLP-1 and blood glucose levels were unaffected by omega-3 fatty acid supplementation, however, animals that were fed fish or flaxseed oil-supplemented diets had significantly heightened colonic FFAR4 and actin expression, and reduced expression of the pro-inflammatory cytokine TNF-α compared to animals fed control diets. CONCLUSIONS: These results suggest that similar to ingestion of other fats, dietary-intake of omega-3 fatty acids can alter FFAR4 expression within the colon.


Subject(s)
Colon/metabolism , Fish Oils/pharmacology , Linseed Oil/pharmacology , Receptors, G-Protein-Coupled/biosynthesis , Animals , Blood Glucose/metabolism , Colon/drug effects , Diet , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Glucagon-Like Peptide 1/metabolism , Male , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
8.
Biochem Pharmacol ; 87(4): 650-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24412271

ABSTRACT

Free fatty acid receptor 4 (FFA4), previously known as GPR120, is a G protein-coupled receptor that promotes numerous anti-inflammatory and antidiabetic effects upon its agonism by long chained unsaturated fatty acids. We have previously demonstrated that agonism of FFA4 with docosahexaenoic acid (DHA) and alpha-linoleic acid (ALA) facilitates rapid and transient phosphorylation of FFA4 expressed ectopically on the surface of HEK293 cells. However, the precise mechanisms that promote FFA4 phosphorylation remain elusive. In the current study, we examined the mechanisms behind both heterologous and homologous phosphorylation of FFA4 and set out to identify the foci of FFA4 phosphorylation. Our results demonstrate that basal and heterologous phosphorylation of FFA4 are mediated by protein kinase C (PKC), while G protein-coupled receptor kinase 6 (GRK6) plays the predominant role in DHA-mediated phosphorylation of FFA4. Furthermore, we identify Thr(347), Ser(350), and Ser(357) in the C-terminal tail as major sites of FFA4 phosphorylation. Concurrent mutation of these three sites leads to a FFA4 receptor that seemingly affects Gαq/11 signaling in a positive manner as demonstrated by heightened intracellular Ca(2+) responses following agonism with DHA. Importantly, this phosphodefective FFA4 mutant lacked the ability to promote ß-arrestin-2 recruitment to the cell membrane. Since many of the functionally beneficial physiological effects of FFA4 are noted to be ß-arrestin mediated, these findings could provide insight into the structural requirements for FFA4 function.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Peptide Fragments/metabolism , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/metabolism , Serine/metabolism , Threonine/metabolism , Amino Acid Sequence , G-Protein-Coupled Receptor Kinases/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Peptide Fragments/genetics , Phosphorylation/physiology , Protein Kinase C/genetics , Receptors, G-Protein-Coupled/genetics , Sequence Homology, Amino Acid , Serine/genetics , Threonine/genetics
9.
Biochem Pharmacol ; 84(5): 661-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22728070

ABSTRACT

The ß2-adrenergic receptor (ß2AR) is the prototypical member of the heptahelical G protein-coupled receptor (GPCR) superfamily and is well-known to elicit biological effects through both G protein-dependent and G protein-independent signaling cascades. Agonism of ß2AR has been described to promote phosphorylation and activation of extracellular signal-regulated kinases (ERK1/2) via a G-protein/PKA pathway that transpires rapidly upon receptor agonism, as well as by a distinct ß-arrestin-mediated pathway that occurs at later time points. We have previously shown that ß2AR agonism promotes generation of intracellular reactive oxygen species (ROS) and that ß2AR-associated G protein signaling is dependent on ROS formation. It has also been suggested that ß2AR-mediated ROS generation occurs via recruitment of ß-arrestins. In this study, we confirm the effects of ß-arrestin on ß2AR-induced ROS generation, and investigate the ROS-dependency of ß-arrestin-linked ß2AR signaling. In HEK293 cells, both coimmunoprecipitation and BRET studies reveal that ROS are vital for the physical interaction of ß2AR with ß-arrestin partner proteins. Using phosphorylation of ERK1/2 as a functional endpoint to assess the role of ROS in ß2AR-ß-arrestin signaling, our results show that inhibition of intracellular ROS abrogates both the ß-arrestin and G protein-mediated phosphorylation of ERK1/2 upon agonism of ß2AR. Importantly, both the G protein and ß-arrestin components were reversed upon exogenous administration of ROS, suggesting a critical role for oxidants in stabilization of ß2AR. Taken together, our data signify that ROS serve purposeful roles in stabilizing both G protein- and ß-arrestin-mediated ß2AR signaling in HEK293 cells.


Subject(s)
Arrestins/metabolism , MAP Kinase Signaling System , Reactive Oxygen Species/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Blotting, Western , Cell Line , Energy Transfer , Flow Cytometry , Humans , Hydrogen Peroxide/pharmacology , Phosphorylation , Protein Binding , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...