Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Circ Res ; 127(10): 1221-1232, 2020 10 23.
Article in English | MEDLINE | ID: mdl-32791884

ABSTRACT

RATIONALE: Endothelial cells are thought to emerge de novo from the mesoderm to form the entire circulatory system. Recently, erythro-myeloid progenitors (EMPs) have been proposed to be another remarkable developmental origin for blood vessels in multiple organs, including the hindbrain, liver, lung, and heart, as demonstrated by lineage tracing studies using different genetic tools. These observations challenge the current consensus that intraembryonic vessels are thought to expand solely by the proliferation of preexisting endothelial cells. Resolution of this controversy over the developmental origin of endothelial cells is crucial for developing future therapeutics for vessel-dependent organ repair and regeneration. OBJECTIVE: To examine the contribution of EMPs to intraembryonic endothelial cells. METHODS AND RESULTS: We first used a transgenic mouse expressing a tamoxifen-inducible Mer-iCre fusion protein driven by the Csf1r (colony stimulating factor 1 receptor) promoter. Genetic lineage tracing based on Csf1r-Mer-iCre-Mer showed no contribution of EMPs to brain endothelial cells identified by several markers. We also generated a knock-in mouse line by inserting an internal ribosome entry site-iCre cassette into the 3' untranslated region of Csf1r gene to further investigate the cellular fates of EMPs. Similarly, we did not find any Csf1r-ires-iCre traced endothelial cells in brain, liver, lung, or heart in development either. Additionally, we found that Kit (KIT proto-oncogene receptor tyrosine kinase) was expressed not only in EMPs but also in embryonic hindbrain endothelial cells. Therefore, Kit promoter-driven recombinase, such as Kit-CreER, is a flawed tool for lineage tracing when examining the contribution of EMPs to hindbrain endothelial cells. We also traced CD45 (protein tyrosine phosphatase receptor type C; Ptprc)+ circulating EMPs and did not find any CD45 lineage-derived endothelial cells during development. CONCLUSIONS: Our study suggested that EMPs are not the origin of intraembryonic endothelial cells.


Subject(s)
Cell Lineage , Endothelial Cells/cytology , Erythroid Precursor Cells/cytology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Fetal Heart/cytology , Liver/cytology , Liver/embryology , Lung/cytology , Lung/embryology , Macrophages/cytology , Mesoderm/cytology , Mice , Rhombencephalon/cytology , Rhombencephalon/embryology
3.
Circ Res ; 125(3): 343-355, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31185811

ABSTRACT

RATIONALE: The developing heart is composed of cardiomyocytes and noncardiomyocytes since the early stage. It is generally believed that noncardiomyocytes including the cardiac progenitors contribute to new cardiomyocytes of the looping heart. However, it remains unclear what the cellular dynamics of nonmyocyte to cardiomyocyte conversion are and when the lineage segregation occurs during development. It also remains unknown whether nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. OBJECTIVE: We quantify the lineage conversion of noncardiomyocytes to cardiomyocytes in the embryonic and neonatal hearts and determine when the 2 cell lineages segregate during heart development. Moreover, we directly test if nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. METHODS AND RESULTS: We generated a dual genetic lineage tracing strategy in which cardiomyocytes and noncardiomyocytes of the developing heart could be simultaneously labeled by 2 orthogonal recombination systems. Genetic fate mapping showed that nonmyocyte to cardiomyocyte conversion peaks at E8.0 (embryonic day) to E8.5 and gradually declines at E9.5 and E10.5. Noncardiomyocytes do not generate any cardiomyocyte at and beyond E11.5 to E12.5. In the neonatal heart, noncardiomyocytes also do not contribute to any new cardiomyocyte in homeostasis or after injury. CONCLUSIONS: Noncardiomyocytes contribute to new cardiomyocytes of the developing heart at early embryonic stage before E11.5. The noncardiomyocyte and cardiomyocyte lineage segregation occurs between E10.5 and E11.5, which is maintained afterward even during neonatal heart regeneration.


Subject(s)
Cell Lineage , Fetal Heart/cytology , Genes, Reporter , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Tracking , Gene Expression Regulation, Developmental , Genetic Markers , Gestational Age , Heart/embryology , Heart/physiology , Mice , Mice, Transgenic , Regeneration , Stem Cells/classification , Stem Cells/cytology
4.
Circulation ; 139(20): 2342-2357, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30818997

ABSTRACT

BACKGROUND: The primary cilium is a singular cellular structure that extends from the surface of many cell types and plays crucial roles in vertebrate development, including that of the heart. Whereas ciliated cells have been described in developing heart, a role for primary cilia in adult heart has not been reported. This, coupled with the fact that mutations in genes coding for multiple ciliary proteins underlie polycystic kidney disease, a disorder with numerous cardiovascular manifestations, prompted us to identify cells in adult heart harboring a primary cilium and to determine whether primary cilia play a role in disease-related remodeling. METHODS: Histological analysis of cardiac tissues from C57BL/6 mouse embryos, neonatal mice, and adult mice was performed to evaluate for primary cilia. Three injury models (apical resection, ischemia/reperfusion, and myocardial infarction) were used to identify the location and cell type of ciliated cells with the use of antibodies specific for cilia (acetylated tubulin, γ-tubulin, polycystin [PC] 1, PC2, and KIF3A), fibroblasts (vimentin, α-smooth muscle actin, and fibroblast-specific protein-1), and cardiomyocytes (α-actinin and troponin I). A similar approach was used to assess for primary cilia in infarcted human myocardial tissue. We studied mice silenced exclusively in myofibroblasts for PC1 and evaluated the role of PC1 in fibrogenesis in adult rat fibroblasts and myofibroblasts. RESULTS: We identified primary cilia in mouse, rat, and human heart, specifically and exclusively in cardiac fibroblasts. Ciliated fibroblasts are enriched in areas of myocardial injury. Transforming growth factor ß-1 signaling and SMAD3 activation were impaired in fibroblasts depleted of the primary cilium. Extracellular matrix protein levels and contractile function were also impaired. In vivo, depletion of PC1 in activated fibroblasts after myocardial infarction impaired the remodeling response. CONCLUSIONS: Fibroblasts in the neonatal and adult heart harbor a primary cilium. This organelle and its requisite signaling protein, PC1, are required for critical elements of fibrogenesis, including transforming growth factor ß-1-SMAD3 activation, production of extracellular matrix proteins, and cell contractility. Together, these findings point to a pivotal role of this organelle, and PC1, in disease-related pathological cardiac remodeling and suggest that some of the cardiovascular manifestations of autosomal dominant polycystic kidney disease derive directly from myocardium-autonomous abnormalities.


Subject(s)
Fibroblasts/ultrastructure , Myocardium/pathology , Polycystic Kidney, Autosomal Dominant/pathology , 3T3 Cells/ultrastructure , Animals , Animals, Newborn , Atrial Remodeling , Cilia , Fetal Heart/cytology , Fibrosis , Heart Injuries/pathology , Humans , Kinesins/deficiency , Kinesins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Rats , Signal Transduction , Smad3 Protein/physiology , TRPP Cation Channels/deficiency , TRPP Cation Channels/physiology , Transforming Growth Factor beta1/physiology , Ventricular Remodeling
5.
FASEB J ; 33(6): 7417-7426, 2019 06.
Article in English | MEDLINE | ID: mdl-30884246

ABSTRACT

Fetal cardiomyocytes shift from glycolysis to oxidative phosphorylation around the time of birth. Myeloid ecotropic viral integration site 1 (MEIS1) is a transcription factor that promotes glycolysis in hematopoietic stem cells. We reasoned that MEIS1 could have a similar role in the developing heart. We hypothesized that suppression of MEIS1 expression in fetal sheep cardiomyocytes leads to a metabolic switch as found at birth. Expression of MEIS1 was assayed in left ventricular cardiac tissue and primary cultures of cardiomyocytes from fetal (100- and 135-d gestation, term = 145 d), neonatal, and adult sheep. Cultured cells were treated with short interfering RNA (siRNA) to suppress MEIS1. Oxygen consumption rate was assessed with the Seahorse metabolic flux analyzer, and mitochondrial activity was assessed by staining cells with MitoTracker Orange. Cardiomyocyte respiratory capacity increased with advancing age concurrently with decreased expression of MEIS1. MEIS1 suppression with siRNA increased maximal oxygen consumption in fetal cells but not in postnatal cells. Mitochondrial activity was increased and expression of glycolytic genes decreased when MEIS1 expression was suppressed. Thus, we conclude that MEIS1 is a key regulator of cardiomyocyte metabolism and that the normal down-regulation of MEIS1 with age underlies a gradual switch to oxidative metabolism.-Lindgren, I. M., Drake, R. R., Chattergoon, N. N., Thornburg, K. L. Down-regulation of MEIS1 promotes the maturation of oxidative phosphorylation in perinatal cardiomyocytes.


Subject(s)
Aging/metabolism , Fetal Heart/cytology , Gene Expression Regulation, Developmental , Mitochondria, Heart/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/physiology , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/biosynthesis , Aging/genetics , Animals , Cells, Cultured , Female , Fetal Heart/metabolism , Gestational Age , Glycolysis , Heart/growth & development , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/biosynthesis , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myocardium/cytology , Oxygen/blood , Oxygen Consumption , Partial Pressure , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Sheep
6.
Mol Ther ; 27(1): 29-45, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30528086

ABSTRACT

Reactivating post-natal myocardial regeneration potential may be a feasible strategy to regenerate the injured adult heart. Long non-coding RNAs (lncRNAs) have been implicated in regulating cellular differentiation, but whether they can elicit a regenerative response in the post-natal heart remains unknown. In this study, by characterizing the lncRNA transcriptome in human hearts during the fetal-to-adult transition, we found that 3,092 lncRNAs were differentially expressed, and we further identified a novel upregulated fetal lncRNA that we called endogenous cardiac regeneration-associated regulator (ECRAR), which promoted DNA synthesis, mitosis, and cytokinesis in post-natal day 7 and adult rat cardiomyocytes (CMs). Overexpression of ECRAR markedly stimulated myocardial regeneration and induced recovery of cardiac function after myocardial infarction (MI). Knockdown of ECRAR inhibited post-natal day 1 CM proliferation and prevented post-MI recovery. ECRAR was transcriptionally upregulated by E2F transcription factor 1 (E2F1). In addition, ECRAR directly bound to and promoted the phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), resulting in downstream targets of cyclin D1 and cyclin E1 activation, which, in turn, activated E2F1. The E2F1-ECRAR-ERK1/2 signaling formed a positive feedback loop to drive cell cycle progression, and, therefore, it promoted CM proliferation. These findings indicated that our newly discovered ECRAR may be a valuable therapeutic target for heart failure.


Subject(s)
MAP Kinase Signaling System/physiology , Myocardium/cytology , Myocardium/metabolism , RNA, Long Noncoding/metabolism , Regeneration/physiology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Heart/cytology , Fetal Heart/metabolism , Humans , MAP Kinase Signaling System/genetics , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , Rats , Rats, Wistar , Regeneration/genetics , Signal Transduction/genetics , Signal Transduction/physiology
7.
J Physiol ; 597(8): 2163-2176, 2019 04.
Article in English | MEDLINE | ID: mdl-30770568

ABSTRACT

KEY POINTS: Plasma thyroid hormone (tri-iodo-l-thyronine; T3 ) concentrations rise near the end of gestation and is known to inhibit proliferation and stimulate maturation of cardiomyocytes before birth. Thyroid hormone receptors are required for the action of thyroid hormone in fetal cardiomyocytes. Loss of thyroid hormone receptor (TR)α1 abolishes T3 signalling via extracellular signal-related kinase and Akt in fetal cardiomyocytes. The expression of TRα1 and TRß1 in ovine fetal myocardium increases with age, although TRα1 levels always remain higher than those of TRß1. Near term fetal cardiac myocytes are more sensitive than younger myocytes to thyroid receptor blockade by antagonist, NH3, and to the effects of TRα1/α2 short interfering RNA. Although T3 is known to abrogate ovine cardiomyocyte proliferation stimulated by insulin-like growth factor 1, this effect is mediated via the genomic action of thyroid hormone receptors, with little evidence for non-genomic mechanisms. ABSTRACT: We have previously shown that the late-term rise in tri-iodo-l-thyronine (T3 ) in fetal sheep leads to the inhibition of proliferation and promotion of maturation in cardiomyocytes. The present study was designed to determine whether these T3 -induced changes are mediated via thyroid hormone receptors (TRs) or by non-genomic mechanisms. Fetal cardiomyocytes were isolated from 102 ± 3 and 135 ± 1 days of gestational age (dGA) sheep (n = 7 per age; term ∼145 dGA). Cells were treated with T3 (1.5 nm), insulin-like growth factor (IGF)-1 (1 µg mL-1 ) or a combination in the presence of TR antagonist NH3 (100 nm) or following short interfering RNA (siRNA) knockdown of TRα1/α2. Proliferation was quantified by 5-bromo-2'-deoxyuridine (BrdU) uptake (10 µm). Western blots measured protein levels of extracellular signal-related kinase (ERK), Akt, TRα1/ß1 and p21. Age specific levels of TRα1/ß1 were measured in normal hearts from fetuses [95 dGA (n = 8), 135 dGA (n = 7)], neonates (n = 8) and adult ewes (n = 7). TRα1 protein levels were consistently >50% more than TRß1 at each gestational age (P < 0.05). T3 reduced IGF-1 stimulated proliferation by ∼50% in 100 dGA and by ∼75% in 135 dGA cardiomyocytes (P < 0.05). NH3 blocked the T3  + IGF-1 reduction of BrdU uptake without altering the phosphorylation of ERK or Akt at both ages. NH3 did not suppress T3 -induced p21 expression in 100 dGA cardiomyocytes in 135 dGA cardiomyocytes, NH3 alone reduced BrdU uptake (-28%, P < 0.05), as well as T3 -induced p21 (-75%, P < 0.05). In both ages, siRNA knockdown of TRα1/α2 blocked the T3  + IGF-1 reduction of BrdU uptake and dramatically reduced ERK and Akt signalling in 135 dGA cardiomyocytes. In conclusion, TRs are required for normal proliferation and T3 signalling in fetal ovine cardiomyocytes, with the sensitivity to TR blockade being age-dependent.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Cell Proliferation , Cells, Cultured , Fetal Heart/cytology , Fetal Heart/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sheep , Triiodothyronine/metabolism
8.
Part Fibre Toxicol ; 16(1): 24, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31215478

ABSTRACT

BACKGROUND: Nano-titanium dioxide (nano-TiO2) is amongst the most widely utilized engineered nanomaterials (ENMs). However, little is known regarding the consequences maternal ENM inhalation exposure has on growing progeny during gestation. ENM inhalation exposure has been reported to decrease mitochondrial bioenergetics and cardiac function, though the mechanisms responsible are poorly understood. Reactive oxygen species (ROS) are increased as a result of ENM inhalation exposure, but it is unclear whether they impact fetal reprogramming. The purpose of this study was to determine whether maternal ENM inhalation exposure influences progeny cardiac development and epigenomic remodeling. RESULTS: Pregnant FVB dams were exposed to nano-TiO2 aerosols with a mass concentration of 12.09 ± 0.26 mg/m3 starting at gestational day five (GD 5), for 6 h over 6 non-consecutive days. Aerosol size distribution measurements indicated an aerodynamic count median diameter (CMD) of 156 nm with a geometric standard deviation (GSD) of 1.70. Echocardiographic imaging was used to assess cardiac function in maternal, fetal (GD 15), and young adult (11 weeks) animals. Electron transport chain (ETC) complex activities, mitochondrial size, complexity, and respiration were evaluated, along with 5-methylcytosine, Dnmt1 protein expression, and Hif1α activity. Cardiac functional analyses revealed a 43% increase in left ventricular mass and 25% decrease in cardiac output (fetal), with an 18% decrease in fractional shortening (young adult). In fetal pups, hydrogen peroxide (H2O2) levels were significantly increased (~ 10 fold) with a subsequent decrease in expression of the antioxidant enzyme, phospholipid hydroperoxide glutathione peroxidase (GPx4). ETC complex activity IV was decreased by 68 and 46% in fetal and young adult cardiac mitochondria, respectively. DNA methylation was significantly increased in fetal pups following exposure, along with increased Hif1α activity and Dnmt1 protein expression. Mitochondrial ultrastructure, including increased size, was observed at both fetal and young adult stages following maternal exposure. CONCLUSIONS: Maternal inhalation exposure to nano-TiO2 results in adverse effects on cardiac function that are associated with increased H2O2 levels and dysregulation of the Hif1α/Dnmt1 regulatory axis in fetal offspring. Our findings suggest a distinct interplay between ROS and epigenetic remodeling that leads to sustained cardiac contractile dysfunction in growing and young adult offspring following maternal ENM inhalation exposure.


Subject(s)
Epigenesis, Genetic/drug effects , Heart Diseases/chemically induced , Maternal Exposure/adverse effects , Nanoparticles/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Reactive Oxygen Species/metabolism , Titanium/toxicity , Animals , Female , Fetal Heart/cytology , Fetal Heart/drug effects , Fetal Heart/metabolism , Heart Diseases/embryology , Heart Diseases/metabolism , Male , Mice , Mice, Inbred Strains , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nanoparticles/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Titanium/administration & dosage
9.
Circ Res ; 118(2): 254-65, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26659641

ABSTRACT

RATIONALE: Unraveling the developmental origin of cardiac fat could offer important implications for the treatment of cardiovascular disease. The recent identification of the mesothelial source of epicardial fat tissues reveals a heterogeneous origin of adipocytes in the adult heart. However, the developmental origin of adipocytes inside the heart, namely intramyocardial adipocytes, remains largely unknown. OBJECTIVE: To trace the developmental origin of intramyocardial adipocytes. METHODS AND RESULTS: In this study, we identified that the majority of intramyocardial adipocytes were restricted to myocardial regions in close proximity to the endocardium. Using a genetic lineage tracing model of endocardial cells, we found that Nfatc1(+) endocardial cells contributed to a substantial number of intramyocardial adipocytes. Despite the capability of the endocardium to generate coronary vascular endothelial cells surrounding the intramyocardial adipocytes, results from our lineage tracing analyses showed that intramyocardial adipocytes were not derived from coronary vessels. Nevertheless, the endocardium of the postnatal heart did not contribute to intramyocardial adipocytes during homeostasis or after myocardial infarction. CONCLUSIONS: Our in vivo fate-mapping studies demonstrated that the developing endocardium, but not the vascular endothelial cells, gives rise to intramyocardial adipocytes in the adult heart.


Subject(s)
Adipocytes/cytology , Adipogenesis , Cell Lineage , Endocardium/cytology , Fetal Heart/cytology , Adipocytes/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Endocardium/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Homeostasis , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phenotype
10.
Circ Res ; 117(1): 80-8, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26089365

ABSTRACT

Disease models are essential for understanding cardiovascular disease pathogenesis and developing new therapeutics. The human induced pluripotent stem cell (iPSC) technology has generated significant enthusiasm for its potential application in basic and translational cardiac research. Patient-specific iPSC-derived cardiomyocytes offer an attractive experimental platform to model cardiovascular diseases, study the earliest stages of human development, accelerate predictive drug toxicology tests, and advance potential regenerative therapies. Harnessing the power of iPSC-derived cardiomyocytes could eliminate confounding species-specific and interpersonal variations and ultimately pave the way for the development of personalized medicine for cardiovascular diseases. However, the predictive power of iPSC-derived cardiomyocytes as a valuable model is contingent on comprehensive and rigorous molecular and functional characterization.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Action Potentials , Bioengineering/methods , Calcium Signaling , Cardiovascular Diseases/pathology , Cations/metabolism , Cell Differentiation , Cell Lineage , Drug Evaluation, Preclinical/methods , Electrophysiology , Energy Metabolism , Excitation Contraction Coupling , Fetal Heart/cytology , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Ion Channels/metabolism , Myocardial Contraction , Myocytes, Cardiac/classification , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Phenotype
11.
Circ Res ; 116(5): 797-803, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25623957

ABSTRACT

RATIONALE: Treatment of sinus node disease with regenerative or cell-based therapies will require a detailed understanding of gene regulatory networks in cardiac pacemaker cells (PCs). OBJECTIVE: To characterize the transcriptome of PCs using RNA sequencing and to identify transcriptional networks responsible for PC gene expression. METHODS AND RESULTS: We used laser capture microdissection on a sinus node reporter mouse line to isolate RNA from PCs for RNA sequencing. Differential expression and network analysis identified novel sinoatrial node-enriched genes and predicted that the transcription factor Islet-1 is active in developing PCs. RNA sequencing on sinoatrial node tissue lacking Islet-1 established that Islet-1 is an important transcriptional regulator within the developing sinoatrial node. CONCLUSIONS: (1) The PC transcriptome diverges sharply from other cardiomyocytes; (2) Islet-1 is a positive transcriptional regulator of the PC gene expression program.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/physiology , Myocytes, Cardiac/metabolism , RNA, Messenger/biosynthesis , Sinoatrial Node/cytology , Transcription Factors/physiology , Animals , Female , Fetal Heart/cytology , Gene Expression Profiling , Gene Regulatory Networks , Genes, Reporter , Heart Atria/cytology , Heart Atria/embryology , Heart Atria/metabolism , High-Throughput Nucleotide Sequencing , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Laser Capture Microdissection , Male , Mice , Molecular Sequence Data , Myocardial Contraction , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Sinoatrial Node/embryology , Sinoatrial Node/metabolism , Subtraction Technique , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transcriptome
12.
J Theor Biol ; 419: 269-277, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28237394

ABSTRACT

The developmental program of the heart requires accurate regulation to ensure continuous circulation and simultaneous cardiac morphogenesis, because any functional abnormalities may progress to congenital heart malformation. Notably, energy metabolism in fetal ventricular cells is regulated in a manner that differs from adult ventricular cells: fetal cardiomyocytes generally have immature mitochondria and fetal ventricular cells show greater dependence on glycolytic ATP production. However, although various characteristics of energy metabolism in fetal ventricular cells have been reported, to our knowledge, a quantitative description of the contributions of these factors to fetal ventricular cell functions has not yet been established. Here, we constructed a mathematical model to integrate various characteristics of fetal ventricular cells and predicted the contribution of each characteristic to the maintenance of intracellular ATP concentration and sarcomere contraction under anoxic conditions. Our simulation results demonstrated that higher glycogen content, higher hexokinase activity, and lower creatine concentration helped prolong the time for which ventricular cell contraction was maintained under anoxic conditions. The integrated model also enabled us to quantitatively assess the contributions of factors related to energy metabolism in ventricular cells. Because fetal cardiomyocytes exhibit similar energy metabolic profiles to stem cell-derived cardiomyocytes and those in the failing heart, an improved understanding of these fetal ventricular cells will contribute to a better comprehension of the processes in stem cell-derived cardiomyocytes or under pathological conditions.


Subject(s)
Adenosine Triphosphate/biosynthesis , Glycolysis , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Action Potentials , Animals , Computer Simulation , Energy Metabolism , Fetal Heart/cytology , Fetal Heart/metabolism , Fetal Heart/physiology , Fetal Hypoxia , Glycogen/metabolism , Guinea Pigs , Heart Ventricles/cytology , Heart Ventricles/embryology , Heart Ventricles/metabolism , Hexokinase/metabolism , Models, Cardiovascular , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Sarcomeres/metabolism
13.
BMC Dev Biol ; 16(1): 41, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27863467

ABSTRACT

BACKGROUND: Long non-coding RNAs (LncRNAs) have been identified to play important roles in epigenetic processes that underpin organogenesis. However, the role of LncRNAs in the regulation of transition from fetal to adult life of human heart has not been evaluated. METHODS: Immunofiuorescent staining was used to determine the extent of cardiac cell proliferation. Human LncRNA microarrays were applied to define gene expression signatures of the fetal (13-17 weeks of gestation, n = 4) and adult hearts (30-40 years old, n = 4). Pathway analysis was performed to predict the function of differentially expressed mRNAs (DEM). DEM related to cell proliferation were selected to construct a lncRNA-mRNA co-expression network. Eight lncRNAs were confirmed by quantificational real-time polymerase chain reaction (n = 6). RESULTS: Cardiac cell proliferation was significant in the fetal heart. Two thousand six hundred six lncRNAs and 3079 mRNAs were found to be differentially expressed. Cell cycle was the most enriched pathway in down-regulated genes in the adult heart. Eight lncRNAs (RP11-119 F7.5, AX747860, HBBP1, LINC00304, TPTE2P6, AC034193.5, XLOC_006934 and AL833346) were predicted to play a central role in cardiac cell proliferation. CONCLUSIONS: We discovered a profile of lncRNAs differentially expressed between the human fetal and adult heart. Several meaningful lncRNAs involved in cardiac cell proliferation were disclosed.


Subject(s)
Fetal Heart/cytology , Gene Expression Profiling/methods , Gene Regulatory Networks , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis/methods , RNA, Long Noncoding/genetics , Adult , Cell Cycle , Cell Proliferation , Cells, Cultured , Female , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Humans , Pregnancy , Pregnancy Trimester, First
14.
Circulation ; 132(2): 109-21, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-25995316

ABSTRACT

BACKGROUND: Heart development is tightly regulated by signaling events acting on a defined number of progenitor and differentiated cardiac cells. Although loss of function of these signaling pathways leads to congenital malformation, the consequences of cardiac progenitor cell or embryonic cardiomyocyte loss are less clear. In this study, we tested the hypothesis that embryonic mouse hearts exhibit a robust mechanism for regeneration after extensive cell loss. METHODS AND RESULTS: By combining a conditional cell ablation approach with a novel blastocyst complementation strategy, we generated murine embryos that exhibit a full spectrum of cardiac progenitor cell or cardiomyocyte ablation. Remarkably, ablation of up to 60% of cardiac progenitor cells at embryonic day 7.5 was well tolerated and permitted embryo survival. Ablation of embryonic cardiomyocytes to a similar degree (50% to 60%) at embryonic day 9.0 could be fully rescued by residual myocytes with no obvious adult cardiac functional deficit. In both ablation models, an increase in cardiomyocyte proliferation rate was detected and accounted for at least some of the rapid recovery of myocardial cellularity and heart size. CONCLUSION: Our study defines the threshold for cell loss in the embryonic mammalian heart and reveals a robust cardiomyocyte compensatory response that sustains normal fetal development.


Subject(s)
Cell Proliferation/physiology , Embryonic Stem Cells/physiology , Fetal Heart/cytology , Myocytes, Cardiac/physiology , Animals , Cell Count/methods , Fetal Heart/growth & development , Gene Knock-In Techniques , Mice , Mice, Transgenic
15.
Biochem Biophys Res Commun ; 464(4): 1000-1007, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26168730

ABSTRACT

The precise assemblage of several types of cardiac precursors controls heart organogenesis. The cardiac precursors show dynamic movement during early development and then form the complicated heart structure. However, cardiomyocyte movements inside the newly organized mammalian heart remain unclear. We previously established the method of ex vivo time-lapse imaging of the murine heart to study cardiomyocyte behavior by using the Fucci (fluorescent ubiquitination-based cell cycle indicator) system, which can effectively label individual G1, S/G2/M, and G1/S-transition phase nuclei in living cardiomyocytes as red, green, and yellow, respectively. Global analysis of gene expression in Fucci green positive ventricular cardiomyocytes confirmed that cell cycle regulatory genes expressed in G1/S, S, G2/M, and M phase transitions were upregulated. Interestingly, pathway analysis revealed that many genes related to the cell cycle were significantly upregulated in the Fucci green positive ventricular cardiomyocytes, while only a small number of genes related to cell motility were upregulated. Time-lapse imaging showed that murine proliferating cardiomyocytes did not exhibit dynamic movement inside the heart, but stayed on site after entering the cell cycle.


Subject(s)
Fetal Heart/cytology , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Animals , Cell Cycle Checkpoints/genetics , Cell Movement , Cell Proliferation , Female , Fetal Heart/embryology , Gene Expression Regulation, Developmental , Genes, Reporter , Heart/growth & development , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Oligonucleotide Array Sequence Analysis , Pregnancy
16.
Am J Physiol Heart Circ Physiol ; 307(8): H1216-25, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128174

ABSTRACT

In the first two-thirds of gestation, ovine fetal cardiomyocytes undergo mitosis to increase cardiac mass and accommodate fetal growth. Thereafter, some myocytes continue to proliferate while others mature and terminally differentiate into binucleated cells. At term (145 days gestational age; dGA) about 60% of cardiomyocytes become binucleated and exit the cell cycle under hormonal control. Rising thyroid hormone (T3) levels near term (135 dGA) inhibit proliferation and stimulate maturation. However, the degree to which intracellular signaling patterns change with age in response to T3 is unknown. We hypothesized that in vitro activation of ERK, Akt, and p70(S6K) by two regulators of cardiomyocyte cell cycle activity, T3 and insulin like growth factor-1 (IGF-1), would be similar in cardiomyocytes at gestational ages 100 and 135 dGA. IGF-1 and T3 each independently stimulated phosphorylation of ERK, Akt, and p70(S6K) in cells at both ages. In the younger mononucleated myocytes, the phosphorylation of ERK and Akt was reduced in the presence of IGF-1 and T3. However, the same hormone combination led to a dramatic twofold increase in the phosphorylation of these signaling proteins in the 135 dGA cardiomyocytes-even in cells that were not proliferating. In the older cells, both mono- and binucleated cells were affected. In conclusion, fetal ovine cardiomyocytes undergo profound maturation-related changes in signaling in response to T3 and IGF-1, but not to either factor alone. Differences in age-related response are likely to be related to milestones in fetal cardiac development as the myocardium prepares for ex utero life.


Subject(s)
Fetal Heart/metabolism , MAP Kinase Signaling System , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Heart/cytology , Fetal Heart/drug effects , Insulin-Like Growth Factor I/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sheep , Thyroid Hormones/pharmacology
17.
Development ; 138(2): 303-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21177343

ABSTRACT

Dishevelled-associated activator of morphogenesis 1 (Daam1), a member of the formin protein family, plays an important role in regulating the actin cytoskeleton via mediation of linear actin assembly. Previous functional studies of Daam1 in lower species suggest its essential role in Drosophila trachea formation and Xenopus gastrulation. However, its in vivo physiological function in mammalian systems is largely unknown. We have generated Daam1-deficient mice via gene-trap technology and found that Daam1 is highly expressed in developing murine organs, including the heart. Daam1-deficient mice exhibit embryonic and neonatal lethality and suffer multiple cardiac defects, including ventricular noncompaction, double outlet right ventricles and ventricular septal defects. In vivo genetic rescue experiments further confirm that the lethality of Daam1-deficient mice results from the inherent cardiac abnormalities. In-depth analyses have revealed that Daam1 is important for regulating filamentous actin assembly and organization, and consequently for cytoskeletal function in cardiomyocytes, which contributes to proper heart morphogenesis. Daam1 is also found to be important for proper cytoskeletal architecture and functionalities in embryonic fibroblasts. Biochemical analyses indicate that Daam1 does not regulate cytoskeletal organization through RhoA, Rac1 or Cdc42. Our study highlights a crucial role for Daam1 in regulating the actin cytoskeleton and tissue morphogenesis.


Subject(s)
Fetal Heart/embryology , Microfilament Proteins/physiology , rho GTP-Binding Proteins/physiology , Actins/metabolism , Animals , Apoptosis , Base Sequence , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Primers/genetics , Female , Fetal Heart/abnormalities , Fetal Heart/cytology , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , Pregnancy , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics
18.
Circulation ; 126(11 Suppl 1): S20-8, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22965984

ABSTRACT

BACKGROUND: Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. METHODS AND RESULTS: We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. CONCLUSIONS: The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Subject(s)
Fetal Stem Cells/transplantation , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/surgery , Mutagenesis, Insertional/methods , Myocardial Infarction/surgery , Animals , Cell Differentiation , Cell Division , Chromosomes, Human, Pair 19/genetics , Endothelial Cells/cytology , Female , Fetal Heart/cytology , Fetal Stem Cells/cytology , Fetal Stem Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Integrases , Intracellular Signaling Peptides and Proteins , Ischemia/physiopathology , Luciferases, Firefly/genetics , Luminescent Proteins/genetics , Magnetic Resonance Imaging , Mice , Mice, SCID , Proteins/genetics , Random Allocation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Thymidine Kinase/genetics , Transgenes , Vesicular Transport Proteins , Viral Proteins/genetics , Virus Integration , Red Fluorescent Protein
19.
Pflugers Arch ; 465(7): 955-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23338940

ABSTRACT

Cardiac CaV1.2 channels play a critical role in cardiac function. It has been proposed that the carboxyl-terminal intracellular tail of the CaV1.2 channel is the target of Ca(2+)-dependent and Ca(2+)-independent regulation of the channel. Recent studies on C-terminal truncated forms of the CaV1.2 channel reported neonatal death, reduced CaV1.2 current, and failure of ß-adrenergic stimulation of these channels in ventricular cardiomyocytes (CMs). Here, we used atrial CMs at embryonic day 18.5 that expressed a C-terminal truncated form of the CaV1.2 channel (Stop/Stop). Surprisingly, the atrial CMs showed robust L-type Ca(2+) currents which could be stimulated by forskolin, an activator of adenylyl cyclase. These currents exhibited a left-ward shift in the voltage-dependent activation curve and a reduced sensitivity to the Ca(2+) channel blocker isradipine as compared to currents in wild-type atrial CMs. RT-PCR analysis revealed normal levels of mRNA for the CaV1.2 channel but a twofold increase in the level of mRNA for the CaV1.3 channel in the Stop/Stop atrium as compared to wild-type atrium. A Western blot analysis indicated an increase of CaV1.3 protein in the Stop/Stop atrium. We suggest that, in contrast to Stop/Stop ventricular CMs, Stop/Stop atrial CMs can compensate the functional loss of the truncated CaV1.2 channel with an upregulation of the CaV1.3 channel.


Subject(s)
Action Potentials , Calcium Channels, L-Type/metabolism , Gene Deletion , Myocytes, Cardiac/physiology , Transcription, Genetic , Animals , Aspartic Acid/genetics , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Colforsin/pharmacology , Fetal Heart/cytology , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Structure, Tertiary
20.
Am J Physiol Heart Circ Physiol ; 304(2): H246-52, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23161881

ABSTRACT

Cardiomyocyte organization is a critical determinant of coordinated cardiac contractile function. Because of the acute opening of the pulmonary circulation, the relative workload of the left ventricle (LV) and right ventricle (RV) changes substantially immediately after birth. We hypothesized that three-dimensional cardiomyocyte architecture might be required to adapt rapidly to accommodate programmed perinatal changes of cardiac function. Isolated fixed hearts from pig fetuses or pigs at midgestation, preborn, postnatal day 1 (P1), postnatal day 5, postnatal day 14 (P14), and adulthood (n = 5 for each group) were acquired for diffusion-weighted magnetic resonance imaging. Cardiomyocyte architecture was visualized by three-dimensional fiber tracking and was quantitatively evaluated by the measured helix angle (α(h)). Upon the completion of MRI, hearts were sectioned and stained with hematoxylin/eosin (H&E) to evaluate cardiomyocyte alignment, with picrosirius red to evaluate collagen content, and with anti-Ki67 to evaluate postnatal cell proliferation. The helical architecture of cardiomyocyte was observed as early as the midgestational period. Postnatal changes of cardiomyocyte architecture were observed from P1 to P14, which primary occurred in the septum and RV free wall (RVFW). In the septum, the volume ratio of LV- vs. RV-associated cardiomyocytes rapidly changed from RV-LV balanced pattern at birth to LV dominant pattern by P14. In the RVFW, subendocardial α(h) decreased by ~30° from P1 to P14. These findings indicate that the helical architecture of cardiomyocyte is developed as early as the midgestation period. Substantial and rapid adaptive changes in cardiac microarchitecture suggested considerable developmental plasticity of cardiomyocyte form and function in the postnatal period in response to altered cardiac mechanical function.


Subject(s)
Cell Shape , Diffusion Tensor Imaging , Fetal Heart/physiology , Myocytes, Cardiac/physiology , Adaptation, Physiological , Age Factors , Aging , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Proliferation , Collagen/metabolism , Fetal Heart/cytology , Fetal Heart/metabolism , Gestational Age , Heart Ventricles/cytology , Heart Ventricles/embryology , Imaging, Three-Dimensional , Immunohistochemistry , Ki-67 Antigen/metabolism , Morphogenesis , Myocytes, Cardiac/metabolism , Swine , Ventricular Function, Left , Ventricular Function, Right , Ventricular Remodeling , Ventricular Septum/cytology , Ventricular Septum/embryology , Ventricular Septum/physiology
SELECTION OF CITATIONS
SEARCH DETAIL