Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Elife ; 122023 10 20.
Article in English | MEDLINE | ID: mdl-37861292

ABSTRACT

Millions suffer from incurable lung diseases, and the donor lung shortage hampers organ transplants. Generating the whole organ in conjunction with the thymus is a significant milestone for organ transplantation because the thymus is the central organ to educate immune cells. Using lineage-tracing mice and human pluripotent stem cell (PSC)-derived lung-directed differentiation, we revealed that gastrulating Foxa2 lineage contributed to both lung mesenchyme and epithelium formation. Interestingly, Foxa2 lineage-derived cells in the lung mesenchyme progressively increased and occupied more than half of the mesenchyme niche, including endothelial cells, during lung development. Foxa2 promoter-driven, conditional Fgfr2 gene depletion caused the lung and thymus agenesis phenotype in mice. Wild-type donor mouse PSCs injected into their blastocysts rescued this phenotype by complementing the Fgfr2-defective niche in the lung epithelium and mesenchyme and thymic epithelium. Donor cell is shown to replace the entire lung epithelial and robust mesenchymal niche during lung development, efficiently complementing the nearly entire lung niche. Importantly, those mice survived until adulthood with normal lung function. These results suggest that our Foxa2 lineage-based model is unique for the progressive mobilization of donor cells into both epithelial and mesenchymal lung niches and thymus generation, which can provide critical insights into studying lung transplantation post-transplantation shortly.


Subject(s)
Endothelial Cells , Pluripotent Stem Cells , Mice , Humans , Animals , Adult , Pluripotent Stem Cells/metabolism , Cell Differentiation , Lung , Blastocyst/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism
2.
Front Pharmacol ; 14: 1158222, 2023.
Article in English | MEDLINE | ID: mdl-37101545

ABSTRACT

Introduction: Tyrosine kinase inhibitor drugs (TKIs) are highly effective cancer drugs, yet many TKIs are associated with various forms of cardiotoxicity. The mechanisms underlying these drug-induced adverse events remain poorly understood. We studied mechanisms of TKI-induced cardiotoxicity by integrating several complementary approaches, including comprehensive transcriptomics, mechanistic mathematical modeling, and physiological assays in cultured human cardiac myocytes. Methods: Induced pluripotent stem cells (iPSCs) from two healthy donors were differentiated into cardiac myocytes (iPSC-CMs), and cells were treated with a panel of 26 FDA-approved TKIs. Drug-induced changes in gene expression were quantified using mRNA-seq, changes in gene expression were integrated into a mechanistic mathematical model of electrophysiology and contraction, and simulation results were used to predict physiological outcomes. Results: Experimental recordings of action potentials, intracellular calcium, and contraction in iPSC-CMs demonstrated that modeling predictions were accurate, with 81% of modeling predictions across the two cell lines confirmed experimentally. Surprisingly, simulations of how TKI-treated iPSC-CMs would respond to an additional arrhythmogenic insult, namely, hypokalemia, predicted dramatic differences between cell lines in how drugs affected arrhythmia susceptibility, and these predictions were confirmed experimentally. Computational analysis revealed that differences between cell lines in the upregulation or downregulation of particular ion channels could explain how TKI-treated cells responded differently to hypokalemia. Discussion: Overall, the study identifies transcriptional mechanisms underlying cardiotoxicity caused by TKIs, and illustrates a novel approach for integrating transcriptomics with mechanistic mathematical models to generate experimentally testable, individual-specific predictions of adverse event risk.

3.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35686629

ABSTRACT

The specification of distinct cardiac lineages occurs before chamber formation and acquisition of bona fide atrial or ventricular identity. However, the mechanisms underlying these early specification events remain poorly understood. Here, we performed single cell analysis at the murine cardiac crescent, primitive heart tube and heart tube stages to uncover the transcriptional mechanisms underlying formation of atrial and ventricular cells. We find that progression towards differentiated cardiomyocytes occurs primarily based on heart field progenitor identity, and that progenitors contribute to ventricular or atrial identity through distinct differentiation mechanisms. We identify new candidate markers that define such differentiation processes and examine their expression dynamics using computational lineage trajectory methods. We further show that exposure to exogenous retinoic acid causes defects in ventricular chamber size, dysregulation in FGF signaling and a shunt in differentiation towards orthogonal lineages. Retinoic acid also causes defects in cell-cycle exit resulting in formation of hypomorphic ventricles. Collectively, our data identify, at a single cell level, distinct lineage trajectories during cardiac specification and differentiation, and the precise effects of manipulating cardiac progenitor patterning via retinoic acid signaling.


Subject(s)
Heart , Tretinoin , Animals , Cell Differentiation , Heart Atria , Heart Ventricles/metabolism , Mice , Myocytes, Cardiac/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
4.
Cell Stem Cell ; 29(4): 559-576.e7, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35325615

ABSTRACT

Pluripotent stem-cell-derived cardiomyocytes (PSC-CMs) provide an unprecedented opportunity to study human heart development and disease, but they are functionally and structurally immature. Here, we induce efficient human PSC-CM (hPSC-CM) maturation through metabolic-pathway modulations. Specifically, we find that peroxisome-proliferator-associated receptor (PPAR) signaling regulates glycolysis and fatty acid oxidation (FAO) in an isoform-specific manner. While PPARalpha (PPARa) is the most active isoform in hPSC-CMs, PPARdelta (PPARd) activation efficiently upregulates the gene regulatory networks underlying FAO, increases mitochondrial and peroxisome content, enhances mitochondrial cristae formation, and augments FAO flux. PPARd activation further increases binucleation, enhances myofibril organization, and improves contractility. Transient lactate exposure, which is frequently used for hPSC-CM purification, induces an independent cardiac maturation program but, when combined with PPARd activation, still enhances oxidative metabolism. In summary, we investigate multiple metabolic modifications in hPSC-CMs and identify a role for PPARd signaling in inducing the metabolic switch from glycolysis to FAO in hPSC-CMs.


Subject(s)
Induced Pluripotent Stem Cells , PPAR delta , Pluripotent Stem Cells , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , PPAR delta/metabolism
5.
Stem Cell Reports ; 16(12): 3036-3049, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34739849

ABSTRACT

A library of well-characterized human induced pluripotent stem cell (hiPSC) lines from clinically healthy human subjects could serve as a useful resource of normal controls for in vitro human development, disease modeling, genotype-phenotype association studies, and drug response evaluation. We report generation and extensive characterization of a gender-balanced, racially/ethnically diverse library of hiPSC lines from 40 clinically healthy human individuals who range in age from 22 to 61 years. The hiPSCs match the karyotype and short tandem repeat identities of their parental fibroblasts, and have a transcription profile characteristic of pluripotent stem cells. We provide whole-genome sequencing data for one hiPSC clone from each individual, genomic ancestry determination, and analysis of mendelian disease genes and risks. We document similar transcriptomic profiles, single-cell RNA-sequencing-derived cell clusters, and physiology of cardiomyocytes differentiated from multiple independent hiPSC lines. This extensive characterization makes this hiPSC library a valuable resource for many studies on human biology.


Subject(s)
Health , Induced Pluripotent Stem Cells/cytology , Adult , Calcium Signaling , Cell Differentiation , Cell Line , Clone Cells , Ethnicity , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Variation , Heart Atria/cytology , Heart Ventricles/cytology , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Risk Factors , Young Adult
6.
Circulation ; 141(11): 916-930, 2020 03 17.
Article in English | MEDLINE | ID: mdl-31992066

ABSTRACT

BACKGROUND: Sphingolipids have recently emerged as a biomarker of recurrence and mortality after myocardial infarction (MI). The increased ceramide levels in mammalian heart tissues during acute MI, as demonstrated by several groups, is associated with higher cell death rates in the left ventricle and deteriorated cardiac function. Ceramidase, the only enzyme known to hydrolyze proapoptotic ceramide, generates sphingosine, which is then phosphorylated by sphingosine kinase to produce the prosurvival molecule sphingosine-1-phosphate. We hypothesized that Acid Ceramidase (AC) overexpression would counteract the negative effects of elevated ceramide and promote cell survival, thereby providing cardioprotection after MI. METHODS: We performed transcriptomic, sphingolipid, and protein analyses to evaluate sphingolipid metabolism and signaling post-MI. We investigated the effect of altering ceramide metabolism through a loss (chemical inhibitors) or gain (modified mRNA [modRNA]) of AC function post hypoxia or MI. RESULTS: We found that several genes involved in de novo ceramide synthesis were upregulated and that ceramide (C16, C20, C20:1, and C24) levels had significantly increased 24 hours after MI. AC inhibition after hypoxia or MI resulted in reduced AC activity and increased cell death. By contrast, enhancing AC activity via AC modRNA treatment increased cell survival after hypoxia or MI. AC modRNA-treated mice had significantly better heart function, longer survival, and smaller scar size than control mice 28 days post-MI. We attributed the improvement in heart function post-MI after AC modRNA delivery to decreased ceramide levels, lower cell death rates, and changes in the composition of the immune cell population in the left ventricle manifested by lowered abundance of proinflammatory detrimental neutrophils. CONCLUSIONS: Our findings suggest that transiently altering sphingolipid metabolism through AC overexpression is sufficient and necessary to induce cardioprotection post-MI, thereby highlighting the therapeutic potential of AC modRNA in ischemic heart disease.


Subject(s)
Acid Ceramidase/physiology , Genetic Therapy , Hypoxia/metabolism , Myocardial Infarction/metabolism , RNA, Messenger/therapeutic use , Sphingolipids/metabolism , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/genetics , Animals , Animals, Newborn , Apoptosis , Ceramides/metabolism , Cicatrix/pathology , Embryoid Bodies , Enzyme Induction , Female , Humans , Hypoxia/etiology , Hypoxia/pathology , Induced Pluripotent Stem Cells/metabolism , Inflammation , Male , Mice , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Transfection , Up-Regulation
7.
Cell ; 176(5): 947-949, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30794778

ABSTRACT

The adult mammalian heart is minimally regenerative after injury, whereas neonatal hearts fully recover even after major damage. New work from the Red-Horse and Woo labs (Das et al., 2019) shows that collateral artery formation is a key mechanism contributing to successful regeneration in newborn mice and provides insights into how collateral arteries form.


Subject(s)
Myocytes, Cardiac , Regeneration , Animals , Mice , Animals, Newborn , Arteries , Heart , Horses
8.
Cell ; 171(3): 573-587.e14, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033129

ABSTRACT

Progenitor cells differentiate into specialized cell types through coordinated expression of lineage-specific genes and modification of complex chromatin configurations. We demonstrate that a histone deacetylase (Hdac3) organizes heterochromatin at the nuclear lamina during cardiac progenitor lineage restriction. Specification of cardiomyocytes is associated with reorganization of peripheral heterochromatin, and independent of deacetylase activity, Hdac3 tethers peripheral heterochromatin containing lineage-relevant genes to the nuclear lamina. Deletion of Hdac3 in cardiac progenitor cells releases genomic regions from the nuclear periphery, leading to precocious cardiac gene expression and differentiation into cardiomyocytes; in contrast, restricting Hdac3 to the nuclear periphery rescues myogenesis in progenitors otherwise lacking Hdac3. Our results suggest that availability of genomic regions for activation by lineage-specific factors is regulated in part through dynamic chromatin-nuclear lamina interactions and that competence of a progenitor cell to respond to differentiation signals may depend upon coordinated movement of responding gene loci away from the nuclear periphery.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Nuclear Lamina/metabolism , Stem Cells/cytology , Animals , Genome , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cells/metabolism
9.
J Vis Exp ; (128)2017 10 12.
Article in English | MEDLINE | ID: mdl-29053691

ABSTRACT

The use of ever-advancing imaging techniques has contributed broadly to our increased understanding of embryonic development. Pre-implantation development and organogenesis are two areas of research that have benefitted greatly from these advances, due to the high quality of data that can be obtained directly from imaging pre-implantation embryos or ex vivo organs. While pre-implantation embryos have yielded data with especially high spatial resolution, later stages have been less amenable to three-dimensional reconstruction. Obtaining high-quality 3D or volumetric data for known embryonic structures in combination with fate mapping or genetic lineage tracing will allow for a more comprehensive analysis of the morphogenetic events taking place during embryogenesis. This protocol describes a whole-mount immunofluorescence approach that allows for the labeling, visualization, and quantification of progenitor cell populations within the developing cardiac crescent, a key structure formed during heart development. The approach is designed in such a way that both cell- and tissue-level information can be obtained. Using confocal microscopy and image processing, this protocol allows for three-dimensional spatial reconstruction of the cardiac crescent, thereby providing the ability to analyze the localization and organization of specific progenitor populations during this critical phase of heart development. Importantly, the use of reference antibodies allows for successive masking of the cardiac crescent and subsequent quantitative measurements of areas within the crescent. This protocol will not only enable a detailed examination of early heart development, but with adaptations should be applicable to most organ systems in the gastrula to early somite stage mouse embryo.


Subject(s)
Embryonic Development/physiology , Fluorescent Antibody Technique/methods , Heart/embryology , Microscopy, Confocal/methods , Animals , Female , Mice , Pregnancy
10.
Nat Commun ; 8: 14428, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195173

ABSTRACT

The recent identification of progenitor populations that contribute to the developing heart in a distinct spatial and temporal manner has fundamentally improved our understanding of cardiac development. However, the mechanisms that direct atrial versus ventricular specification remain largely unknown. Here we report the identification of a progenitor population that gives rise primarily to cardiovascular cells of the ventricles and only to few atrial cells (<5%) of the differentiated heart. These progenitors are specified during gastrulation, when they transiently express Foxa2, a gene not previously implicated in cardiac development. Importantly, Foxa2+ cells contribute to previously identified progenitor populations in a defined pattern and ratio. Lastly, we describe an analogous Foxa2+ population during differentiation of embryonic stem cells. Together, these findings provide insight into the developmental origin of ventricular and atrial cells, and may lead to the establishment of new strategies for generating chamber-specific cell types from pluripotent stem cells.


Subject(s)
Cell Differentiation/physiology , Heart Ventricles/cytology , Heart Ventricles/growth & development , Hepatocyte Nuclear Factor 3-beta/metabolism , Animals , Cell Line , Embryonic Development/physiology , Female , Gastrulation/physiology , Gene Expression Regulation, Developmental , Heart Atria/cytology , Heart Atria/diagnostic imaging , Heart Atria/growth & development , Heart Atria/metabolism , Heart Ventricles/diagnostic imaging , Hepatocyte Nuclear Factor 3-beta/genetics , Mesoderm/cytology , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
11.
Nat Med ; 22(12): 1421-1427, 2016 12.
Article in English | MEDLINE | ID: mdl-27841875

ABSTRACT

Familial dysautonomia (FD) is a debilitating disorder that affects derivatives of the neural crest (NC). For unknown reasons, people with FD show marked differences in disease severity despite carrying an identical, homozygous point mutation in IKBKAP, encoding IκB kinase complex-associated protein. Here we present disease-related phenotypes in human pluripotent stem cells (PSCs) that capture FD severity. Cells from individuals with severe but not mild disease show impaired specification of NC derivatives, including autonomic and sensory neurons. In contrast, cells from individuals with severe and mild FD show defects in peripheral neuron survival, indicating that neurodegeneration is the main culprit for cases of mild FD. Although genetic repair of the FD-associated mutation reversed early developmental NC defects, sensory neuron specification was not restored, indicating that other factors may contribute to disease severity. Whole-exome sequencing identified candidate modifier genes for individuals with severe FD. Our study demonstrates that PSC-based modeling is sensitive in recapitulating disease severity, which presents an important step toward personalized medicine.


Subject(s)
Autonomic Nervous System/physiopathology , Dysautonomia, Familial/physiopathology , Induced Pluripotent Stem Cells , Sensory Receptor Cells/cytology , Adolescent , Adult , Autonomic Nervous System/cytology , Autonomic Nervous System/growth & development , Carrier Proteins/genetics , Case-Control Studies , Cell Survival/genetics , Child , Dysautonomia, Familial/genetics , Female , Genotype , Humans , Male , Models, Neurological , Mutation , Neural Crest/cytology , Neurons/cytology , Phenotype , Sequence Analysis, DNA , Severity of Illness Index , Transcriptional Elongation Factors , Young Adult
12.
J Vis Exp ; (109): e53447, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26967678

ABSTRACT

Human cardiac tissue engineering can fundamentally impact therapeutic discovery through the development of new species-specific screening systems that replicate the biofidelity of three-dimensional native human myocardium, while also enabling a controlled level of biological complexity, and allowing non-destructive longitudinal monitoring of tissue contractile function. Initially, human engineered cardiac tissues (hECT) were created using the entire cell population obtained from directed differentiation of human pluripotent stem cells, which typically yielded less than 50% cardiomyocytes. However, to create reliable predictive models of human myocardium, and to elucidate mechanisms of heterocellular interaction, it is essential to accurately control the biological composition in engineered tissues. To address this limitation, we utilize live cell sorting for the cardiac surface marker SIRPα and the fibroblast marker CD90 to create tissues containing a 3:1 ratio of these cell types, respectively, that are then mixed together and added to a collagen-based matrix solution. Resulting hECTs are, thus, completely defined in both their cellular and extracellular matrix composition. Here we describe the construction of defined hECTs as a model system to understand mechanisms of cell-cell interactions in cell therapies, using an example of human bone marrow-derived mesenchymal stem cells (hMSC) that are currently being used in human clinical trials. The defined tissue composition is imperative to understand how the hMSCs may be interacting with the endogenous cardiac cell types to enhance tissue function. A bioreactor system is also described that simultaneously cultures six hECTs in parallel, permitting more efficient use of the cells after sorting.


Subject(s)
Cell Separation/methods , Myocytes, Cardiac/cytology , Tissue Engineering/methods , Cell Differentiation/physiology , Cell- and Tissue-Based Therapy/methods , Extracellular Matrix , Fibroblasts/cytology , Humans , Myocardium/cytology , Pluripotent Stem Cells/cytology
13.
PLoS One ; 9(7): e101316, 2014.
Article in English | MEDLINE | ID: mdl-25010565

ABSTRACT

The use of human stem cell-derived cardiomyocytes to study atrial biology and disease has been restricted by the lack of a reliable method for stem cell-derived atrial cell labeling and purification. The goal of this study was to generate an atrial-specific reporter construct to identify and purify human stem cell-derived atrial-like cardiomyocytes. We have created a bacterial artificial chromosome (BAC) reporter construct in which fluorescence is driven by expression of the atrial-specific gene sarcolipin (SLN). When purified using flow cytometry, cells with high fluorescence specifically express atrial genes and display functional calcium handling and electrophysiological properties consistent with atrial cardiomyocytes. Our data indicate that SLN can be used as a marker to successfully monitor and isolate hiPSC-derived atrial-like cardiomyocytes. These purified cells may find many applications, including in the study of atrial-specific pathologies and chamber-specific lineage development.


Subject(s)
Flow Cytometry/methods , Heart Atria/cytology , Induced Pluripotent Stem Cells/cytology , Muscle Proteins/genetics , Myocytes, Cardiac/cytology , Proteolipids/genetics , Calcium/metabolism , Cell Differentiation , Chromosomes, Artificial, Bacterial/genetics , Electrophysiological Phenomena , Gene Expression , Genes, Reporter/genetics , Humans , Induced Pluripotent Stem Cells/metabolism
14.
Nat Biotechnol ; 29(11): 1011-8, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020386

ABSTRACT

To identify cell-surface markers specific to human cardiomyocytes, we screened cardiovascular cell populations derived from human embryonic stem cells (hESCs) against a panel of 370 known CD antibodies. This screen identified the signal-regulatory protein alpha (SIRPA) as a marker expressed specifically on cardiomyocytes derived from hESCs and human induced pluripotent stem cells (hiPSCs), and PECAM, THY1, PDGFRB and ITGA1 as markers of the nonmyocyte population. Cell sorting with an antibody against SIRPA allowed for the enrichment of cardiac precursors and cardiomyocytes from hESC/hiPSC differentiation cultures, yielding populations of up to 98% cardiac troponin T-positive cells. When plated in culture, SIRPA-positive cells were contracting and could be maintained over extended periods of time. These findings provide a simple method for isolating populations of cardiomyocytes from human pluripotent stem cell cultures, and thereby establish a readily adaptable technology for generating large numbers of enriched cardiomyocytes for therapeutic applications.


Subject(s)
Antigens, Differentiation/metabolism , Cell Separation/methods , Flow Cytometry/methods , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Receptors, Immunologic/metabolism , Antigens, Surface/analysis , Biomarkers/metabolism , Cell Differentiation , Gene Expression Regulation , Humans , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism
15.
Cell Stem Cell ; 8(2): 228-40, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21295278

ABSTRACT

Efficient differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to a variety of lineages requires step-wise approaches replicating the key commitment stages found during embryonic development. Here we show that expression of PdgfR-α segregates mouse ESC-derived Flk-1 mesoderm into Flk-1(+)PdgfR-α(+) cardiac and Flk-1(+)PdgfR-α(-) hematopoietic subpopulations. By monitoring Flk-1 and PdgfR-α expression, we found that specification of cardiac mesoderm and cardiomyocytes is determined by remarkably small changes in levels of Activin/Nodal and BMP signaling. Translation to human ESCs and iPSCs revealed that the emergence of cardiac mesoderm could also be monitored by coexpression of KDR and PDGFR-α and that this process was similarly dependent on optimal levels of Activin/Nodal and BMP signaling. Importantly, we found that individual mouse and human pluripotent stem cell lines require optimization of these signaling pathways for efficient cardiac differentiation, illustrating a principle that may well apply in other contexts.


Subject(s)
Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nodal Protein/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Flow Cytometry , Humans , Mice , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Development ; 135(14): 2455-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550708

ABSTRACT

The c-Myc protein has been implicated in playing a pivotal role in regulating the expression of a large number of genes involved in many aspects of cellular function. Consistent with this view, embryos lacking the c-myc gene exhibit severe developmental defects and die before midgestation. Here, we show that Sox2Cre-mediated deletion of the conditional c-myc(flox) allele specifically in the epiblast (hence trophoectoderm and primitive endoderm structures are wild type) rescues the majority of developmental abnormalities previously characterized in c-myc knockout embryos, indicating that they are secondary defects and arise as a result of placental insufficiency. Epiblast-restricted c-Myc-null embryos appear morphologically normal and do not exhibit any obvious proliferation defects. Nonetheless, these embryos are severely anemic and die before E12. c-Myc-deficient embryos exhibit fetal liver hypoplasia, apoptosis of erythrocyte precursors and functionally defective definitive hematopoietic stem/progenitor cells. Specific deletion of c-myc(flox) in hemogenic or hepatocytic lineages validate the hematopoietic-specific requirement of c-Myc in the embryo proper and provide in vivo evidence to support a synergism between hematopoietic and liver development. Our results reveal for the first time that physiological levels of c-Myc are essential for cell survival and demonstrate that, in contrast to most other embryonic lineages, erythroblasts and hematopoietic stem/progenitor cells are particularly dependent on c-Myc function.


Subject(s)
Erythroblasts/physiology , Hematopoietic Stem Cells/physiology , Placenta/physiology , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/physiology , Alleles , Animals , Cell Survival , Erythroblasts/cytology , Female , Hematopoietic Stem Cells/cytology , Immunohistochemistry , Mice , Mice, Transgenic , Models, Biological , Morphogenesis , Pregnancy , Proto-Oncogene Proteins c-myc/genetics
17.
Genesis ; 44(8): 355-60, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847871

ABSTRACT

Here we describe the generation of the Nes-Cre1 transgenic mouse line in which Cre recombinase expression is controlled by the rat nestin promoter and intron 2 enhancer. This line has previously been used for conditional loss-of-function studies of various genes in the central nervous system and first branchial arch ectoderm. Here we report the detailed temporal and spatial recombination pattern of Nes-Cre1 using three different reporters of Cre-mediated recombination, ROSA26R (R26R), Z/AP, and Z/EG. Cre/loxP recombination was detected in embryos as early as the head-fold stage. By embryonic day (E)15.5 recombination occurred in virtually all cells of the nervous system and unexpectedly also in somite-derived tissues and kidneys. Tissues with little or no recombination included heart, liver, thymus, and lung. This study suggests that Nes-Cre1-mediated recombination occurs in progenitor cell types present in the neuroectoderm, the developing mesonephros, and the somites.


Subject(s)
Integrases/metabolism , Kidney/embryology , Nervous System/embryology , Recombination, Genetic , Somites/metabolism , Animals , Integrases/genetics , Intermediate Filament Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...