Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
2.
J Am Heart Assoc ; 10(2): e017839, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33399005

ABSTRACT

The failure of adult cardiomyocytes to reproduce themselves to repair an injury results in the development of severe cardiac disability leading to death in many cases. The quest for an understanding of the inability of cardiac myocytes to repair an injury has been ongoing for decades with the identification of various factors which have a temporary effect on cell-cycle activity. Fetal cardiac myocytes are continuously replicating until the time that the developing fetus reaches a stage of maturity sufficient for postnatal life around the time of birth. Recent reports of the ability for early neonatal mice and pigs to completely repair after the severe injury has stimulated further study of the regulators of the cardiomyocyte cell cycle to promote replication for the remuscularization of injured heart. In all mammals just before or after birth, single-nucleated hyperplastically growing cardiomyocytes, 1X2N, undergo ≥1 additional DNA replications not followed by cytokinesis, resulting in cells with ≥2 nuclei or as in primates, multiple DNA replications (polyploidy) of 1 nucleus, 2X2(+)N or 1X4(+)N. All further growth of the heart is attributable to hypertrophy of cardiomyocytes. Animal studies ranging from zebrafish with 100% 1X2N cells in the adult to some strains of mice with up to 98% 2X2N cells in the adult and other species with variable ratios of 1X2N and 2X2N cells are reviewed relative to the time of conversion. Various structural, physiologic, metabolic, genetic, hormonal, oxygenation, and other factors that play a key role in the inability of post-neonatal and adult myocytes to undergo additional cytokinesis are also reviewed.


Subject(s)
Cell Cycle/physiology , Fetal Heart , Myocytes, Cardiac , Animals , Animals, Newborn/growth & development , Animals, Newborn/physiology , Developmental Biology/methods , Developmental Biology/trends , Fetal Heart/cytology , Fetal Heart/growth & development , Fetal Heart/metabolism , Mammals , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
PLoS One ; 13(11): e0207248, 2018.
Article in English | MEDLINE | ID: mdl-30412598

ABSTRACT

BACKGROUND: The objective of this study was to investigate the expression and localisation of thymosin ß4 (Tß4) in the developing human heart. Tß4 is a cardioprotective protein which may have therapeutic potential. While Tß4 is an endogenously produced protein with known importance during development, its role within the developing human heart is not fully understood. Elucidating the localisation of Tß4 within the developing heart will help in understanding its role during cardiac development and is crucial for understanding its potential for cardioprotection and repair in the adult heart. METHODS: Expression of Tß4 mRNA in the early fetal human heart was assessed by PCR using both ventricular and atrial tissue. Fluorescence immunohistochemistry was used to assess the localisation of Tß4 in sections of early fetal human heart. Co-staining with CD31, an endothelial cell marker, and with myosin heavy chain, a cardiomyocyte marker, was used to determine whether Tß4 is localised to these cell types within the early fetal human heart. RESULTS: Tß4 mRNA was found to be expressed in both the atria and the ventricles of the early fetal human heart. Tß4 protein was found to be primarily localised to CD31-expressing endothelial cells and the endocardium as well as being present in the epicardium. Tß4-associated fluorescence was greater in the compact layer of the myocardial wall and the interventricular septum than in the trabecular layer of the myocardium. CONCLUSIONS: The data presented illustrates expression of Tß4 in the developing human heart and demonstrates for the first time that Tß4 in the human heart is primarily localised to endothelial cells of the cardiac microvasculature and coronary vessels as-well as to the endothelial-like cells of the endocardium and to the epicardium.


Subject(s)
Fetal Heart/metabolism , Thymosin/genetics , Thymosin/metabolism , Coronary Vessels/cytology , Coronary Vessels/embryology , Coronary Vessels/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fetal Heart/cytology , Fetal Heart/embryology , Gene Expression Regulation, Developmental , Heart Ventricles/cytology , Heart Ventricles/embryology , Heart Ventricles/metabolism , Humans , Immunohistochemistry , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Nat Commun ; 9(1): 754, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467410

ABSTRACT

The cellular mechanisms driving cardiac tissue formation remain poorly understood, largely due to the structural and functional complexity of the heart. It is unclear whether newly generated myocytes originate from cardiac stem/progenitor cells or from pre-existing cardiomyocytes that re-enter the cell cycle. Here, we identify the source of new cardiomyocytes during mouse development and after injury. Our findings suggest that cardiac progenitors maintain proliferative potential and are the main source of cardiomyocytes during development; however, the onset of αMHC expression leads to reduced cycling capacity. Single-cell RNA sequencing reveals a proliferative, "progenitor-like" population abundant in early embryonic stages that decreases to minimal levels postnatally. Furthermore, cardiac injury by ligation of the left anterior descending artery was found to activate cardiomyocyte proliferation in neonatal but not adult mice. Our data suggest that clonal dominance of differentiating progenitors mediates cardiac development, while a distinct subpopulation of cardiomyocytes may have the potential for limited proliferation during late embryonic development and shortly after birth.


Subject(s)
Heart Injuries/pathology , Heart/growth & development , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryonic Stem Cells/cytology , Female , Fetal Heart/cytology , Fetal Heart/growth & development , Heart Injuries/genetics , Male , Mice , Mice, Transgenic , Myoblasts, Cardiac/cytology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Pericardium/cytology , Pericardium/embryology , Pericardium/growth & development , Pregnancy , Sequence Analysis, RNA
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(11): 1534-1538, 2017 Nov.
Article in Chinese | MEDLINE | ID: mdl-29268859

ABSTRACT

Objective To explore the optimal method for in vitro cultivation of SD fetal rat cardiomyocytes. Methods Cardiomyocytes of SD fetal rats from pregnant rats on gestational day 19 were obtained by digesting cardiac tissues with 0.8 g/L trypsin and 0.4 g/L collagenase II. With differential centrifugation and different animal serums (bovine fetal serum or horse serum), we tried to look for the optimal culture conditions. The morphology and beat frequency of cardiomyocytes were observed under an inverted phase-contrast microscope. In addition, the expressions of alpha-sarcomeric actinin (α-SA) and cardiac troponin I (cTnI) in the cultured cardiomyocytes were detected by immunofluorescence staining to identify cardiomyocytes and the purity of these cells after 24, 48, 72 and 96 hours of cultivation. Results After 24 hours of cultivation, we had seen a majority of adherent cells and a few of suspension cells, and also seen some pulsating cardiomyocytes. The positive rates of α-SA and cTnI in the cardiomyocytes of the horse serum-cultured group were obviously higher than those in the bovine fetal serum-cultured group after 48, 72 and 96 hours of cultivation. Conclusion Co-digestion with trypsin and collagenaseII in combination with horse serum cultivation are easier to cultivate fetal rat cardiomyocytes.


Subject(s)
Cell Culture Techniques/methods , Fetal Heart/cytology , Myocytes, Cardiac/cytology , Actinin/metabolism , Animals , Cells, Cultured , Female , Fetal Heart/metabolism , Fluorescent Antibody Technique , Male , Myocytes, Cardiac/metabolism , Pregnancy , Rats, Sprague-Dawley , Time Factors , Troponin I/metabolism
13.
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
14.
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
15.
Stem Cell Reports ; 6(4): 607-617, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27052314

ABSTRACT

The intrinsic regenerative capacity of human fetal cardiac mesenchymal stromal cells (MSCs) has not been fully characterized. Here we demonstrate that we can expand cells with characteristics of cardiovascular progenitor cells from the MSC population of human fetal hearts. Cells cultured on cardiac muscle laminin (LN)-based substrata in combination with stimulation of the canonical Wnt/ß-catenin pathway showed increased gene expression of ISL1, OCT4, KDR, and NKX2.5. The majority of cells stained positive for PDGFR-α, ISL1, and NKX2.5, and subpopulations also expressed the progenitor markers TBX18, KDR, c-KIT, and SSEA-1. Upon culture of the cardiac MSCs in differentiation media and on relevant LNs, portions of the cells differentiated into spontaneously beating cardiomyocytes, and endothelial and smooth muscle-like cells. Our protocol for large-scale culture of human fetal cardiac MSCs enables future exploration of the regenerative functions of these cells in the context of myocardial injury in vitro and in vivo.


Subject(s)
Cell Proliferation/genetics , Mesenchymal Stem Cells/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Cardiovascular System/cytology , Cell Differentiation/genetics , Cells, Cultured , Fetal Heart/cytology , Gene Expression Profiling/methods , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Laminin/metabolism , Mesenchymal Stem Cells/cytology , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , beta Catenin/metabolism
16.
Prog Biophys Mol Biol ; 120(1-3): 149-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26777584

ABSTRACT

Bile acids are elevated in the blood of women with intrahepatic cholestasis of pregnancy (ICP) and this may lead to fetal arrhythmia, fetal hypoxia and potentially fetal death in utero. The bile acid taurocholic acid (TC) causes abnormal calcium dynamics and contraction in neonatal rat cardiomyocytes. Ursodeoxycholic acid (UDCA), a drug clinically used to treat ICP, prevents adverse effects of TC. During development, the fetus is in a state of relative hypoxia. Although this is essential for the development of the heart and vasculature, resident fibroblasts can transiently differentiate into myofibroblasts and form gap junctions with cardiomyocytes in vitro, resulting in cardiomyocyte depolarization. We expanded on previously published work using an in vitro hypoxia model to investigate the differentiation of human fetal fibroblasts into myofibroblasts. Recent evidence shows that potassium channels are involved in maintaining the membrane potential of ventricular fibroblasts and that ATP-dependent potassium (KATP) channel subunits are expressed in cultured fibroblasts. KATP channels are a valuable target as they are thought to have a cardioprotective role during ischaemic and hypoxic conditions. We investigated whether UDCA could modulate fibroblast membrane potential. We established the isolation and culture of human fetal cardiomyocytes and fibroblasts to investigate the effect of hypoxia, TC and UDCA on human fetal cardiac cells. UDCA hyperpolarized myofibroblasts and prevented TC-induced depolarisation, possibly through the activation of KATP channels that are expressed in cultured fibroblasts. Also, similar to the rat model, UDCA can counteract TC-induced calcium abnormalities in human fetal cultures of cardiomyocytes and myofibroblasts. Under normoxic conditions, we found a higher number of myofibroblasts in cultures derived from human fetal hearts compared to cells isolated from neonatal rat hearts, indicating a possible increased number of myofibroblasts in human fetal hearts. Hypoxia further increased the number of human fetal and rat neonatal myofibroblasts. However, chronically administered UDCA reduced the number of myofibroblasts and prevented hypoxia-induced depolarisation. In conclusion, our results show that the protective effect of UDCA involves both the reduction of fibroblast differentiation into myofibroblasts, and hyperpolarisation of myofibroblasts, most likely through the stimulation of potassium channels, i.e. KATP channels. This could be important in validating UDCA as an antifibrotic and antiarrhythmic drug for treatment of failing hearts and fetal arrhythmia.


Subject(s)
Fetal Heart/cytology , Fibroblasts/drug effects , Myocytes, Cardiac/drug effects , Ursodeoxycholic Acid/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cell Hypoxia/drug effects , Cell Separation , Cytoprotection/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Membrane Potentials/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Taurocholic Acid/pharmacology
17.
Article in English | MEDLINE | ID: mdl-26421632

ABSTRACT

The human fetal heart is formed early during embryogenesis as a result of cell migrations, differentiation, and formative blood flow. It begins to beat around gestation day 22. Progenitor cells are derived from mesoderm (endocardium and myocardium), proepicardium (epicardium and coronary vessels), and neural crest (heart valves, outflow tract septation, and parasympathetic innervation). A variety of molecular disturbances in the factors regulating the specification and differentiation of these cells can cause congenital heart disease. This review explores the contribution of different cardiac progenitors to the embryonic heart development; the pathways and transcription factors guiding their expansion, migration, and functional differentiation; and the endogenous regenerative capacity of the adult heart including the plasticity of cardiomyocytes. Unfolding these mechanisms will become the basis for understanding the dynamics of specific congenital heart disease as well as a means to develop therapy for fetal as well as postnatal cardiac defects and heart failure.


Subject(s)
Fetal Heart/embryology , Fetal Stem Cells/cytology , Heart Defects, Congenital/embryology , Mesoderm/cytology , Neural Crest/cytology , Cell Differentiation , Cell Movement , Coronary Vessels/cytology , Coronary Vessels/embryology , Endocardium/cytology , Endocardium/embryology , Fetal Heart/cytology , Humans , Myocardium/cytology , Pericardium/cytology , Pericardium/embryology
18.
J Endocrinol ; 228(1): R1-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26432905

ABSTRACT

Immature contractile cardiomyocytes proliferate to rapidly increase cell number, establishing cardiomyocyte endowment in the perinatal period. Developmental changes in cellular maturation, size and attrition further contribute to cardiac anatomy. These physiological processes occur concomitant with a changing hormonal environment as the fetus prepares itself for the transition to extrauterine life. There are complex interactions between endocrine, hemodynamic and nutritional regulators of cardiac development. Birth has been long assumed to be the trigger for major differences between the fetal and postnatal cardiomyocyte growth patterns, but investigations in normally growing sheep and rodents suggest this may not be entirely true; in sheep, these differences are initiated before birth, while in rodents they occur after birth. The aim of this review is to draw together our understanding of the temporal regulation of these signals and cardiomyocyte responses relative to birth. Further, we consider how these dynamics are altered in stressed and suboptimal intrauterine environments.


Subject(s)
Endocrine Glands/physiology , Myocytes, Cardiac/physiology , Sheep/growth & development , Angiotensin II , Animals , Animals, Newborn/growth & development , Apoptosis , Cell Count , Cell Differentiation , Cell Proliferation , Cell Size , Female , Fetal Heart/cytology , Fetal Heart/growth & development , Heart/growth & development , Hydrocortisone , Models, Animal , Pregnancy , Somatomedins , Thyroid Hormones
19.
Stem Cells Dev ; 25(3): 277-84, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26573225

ABSTRACT

ATP-binding cassette transporter subfamily G member 2 (Abcg2)-expressing cardiac-side population cells have been identified in the developing and adult heart, although the role they play in mammalian heart growth and regeneration remains unclear. In this study, we use genetic lineage tracing to follow the cell fate of Abcg2-expressing cells in the embryonic and adult heart. During cardiac embryogenesis, the Abcg2 lineage gives rise to multiple cardiovascular cell types, including cardiomyocytes, endothelial cells, and vascular smooth muscle cells. This capacity for Abcg2-expressing cells to contribute to cardiomyocytes decreases rapidly during the postnatal period. We further tested the role of the Abcg2 lineage following myocardial injury. One month following ischemia reperfusion injury, Abcg2-expressing cells contributed significantly to the endothelial cell lineage, however, there was no contribution to regenerated cardiomyocytes. Furthermore, consistent with previous results showing that Abcg2 plays an important cytoprotective role during oxidative stress, we show an increase in Abcg2 labeling of the vasculature, a decrease in the scar area, and a moderate improvement in cardiac function following myocardial injury. We have uncovered a difference in the capacity of Abcg2-expressing cells to generate the cardiovascular lineages during embryogenesis, postnatal growth, and cardiac regeneration.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Lineage , Fetal Heart/cytology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/cytology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Fetal Heart/metabolism , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Oxidative Stress
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...