Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters








Publication year range
1.
Nat Commun ; 14(1): 41, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596806

ABSTRACT

During embryogenesis, haematopoietic and endothelial lineages emerge closely in time and space. It is thought that the first blood and endothelium derive from a common clonal ancestor, the haemangioblast. However, investigation of candidate haemangioblasts in vitro revealed the capacity for mesenchymal differentiation, a feature more compatible with an earlier mesodermal precursor. To date, no evidence for an in vivo haemangioblast has been discovered. Using single cell RNA-Sequencing and in vivo cellular barcoding, we have unravelled the ancestral relationships that give rise to the haematopoietic lineages of the yolk sac, the endothelium, and the mesenchyme. We show that the mesodermal derivatives of the yolk sac are produced by three distinct precursors with dual-lineage outcomes: the haemangioblast, the mesenchymoangioblast, and a previously undescribed cell type: the haematomesoblast. Between E5.5 and E7.5, this trio of precursors seeds haematopoietic, endothelial, and mesenchymal trajectories.


Subject(s)
Hemangioblasts , Yolk Sac , Hematopoiesis/genetics , Clone Cells , Endothelium , Cell Differentiation
3.
J Cell Biol ; 153(5): 985-98, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11381084

ABSTRACT

We have isolated a murine cDNA encoding a 9-kD protein, Chisel (Csl), in a screen for transcriptional targets of the cardiac homeodomain factor Nkx2-5. Csl transcripts were detected in atria and ventricles of the heart and in all skeletal muscles and smooth muscles of the stomach and pulmonary veins. Csl protein was distributed throughout the cytoplasm in fetal muscles, although costameric and M-line localization to the muscle cytoskeleton became obvious after further maturation. Targeted disruption of Csl showed no overt muscle phenotype. However, ectopic expression in C2C12 myoblasts induced formation of lamellipodia in which Csl protein became tethered to membrane ruffles. Migration of these cells was retarded in a monolayer wound repair assay. Csl-expressing myoblasts differentiated and fused normally, although in the presence of insulin-like growth factor (IGF)-1 they showed dramatically enhanced fusion, leading to formation of large dysmorphogenic "myosacs." The activities of transcription factors nuclear factor of activated T cells (NFAT) and myocyte enhancer-binding factor (MEF)2, were also enhanced in an IGF-1 signaling-dependent manner. The dynamic cytoskeletal localization of Csl and its dominant effects on cell shape and behavior and transcription factor activity suggest that Csl plays a role in the regulatory network through which muscle cells coordinate their structural and functional states during growth, adaptation, and repair.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Muscle Proteins/metabolism , Muscles/cytology , Muscles/drug effects , Nuclear Proteins , Xenopus Proteins , Aging/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcineurin/metabolism , Cell Differentiation , Cell Fusion , Cell Line , Cell Size/drug effects , Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , MEF2 Transcription Factors , Mice , Mice, Knockout , Molecular Sequence Data , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscles/embryology , Muscles/metabolism , Myogenic Regulatory Factors , NFATC Transcription Factors , Organ Specificity , Physical Chromosome Mapping , Protein Transport , RNA, Messenger/analysis , RNA, Messenger/genetics , Transcription Factors/metabolism , Wound Healing
4.
Circ Res ; 87(10): 888-95, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11073884

ABSTRACT

Heterozygous mutations in the cardiac homeobox gene, NKX2-5, underlie familial cases of atrial septal defect (ASD) with severe atrioventricular conduction block. In this study, mice heterozygous for Nkx2-5-null alleles were assessed for analogous defects. Although ASD occurred only rarely, atrial septal dysmorphogenesis was evident as increased frequencies of patent foramen ovale and septal aneurysm, and decreased length of the septum primum flap valve. These parameters were compounded by genetic background effects, and in the 129/Sv strain, septal dysmorphogenesis bordered on ASD in 17% of Nkx2-5 heterozygotes. In a proportion of neonatal heterozygotes, as well as in adults with ASD, we found that the size of the foramen ovale was significantly enlarged and altered in shape, potentially exposing the normally thin septum primum to excessive hemodynamic forces. Therefore, defective morphogenesis of the septum secundum may be one contributing factor in the generation of patent foramen ovale, septal aneurysm, and certain ASDs. Mild prolongation of P-R interval in females and an increased frequency of stenotic bicuspid aortic valves were also features of the Nkx2-5 heterozygous phenotype. Our data demonstrate that the complex effects of Nkx2-5 haploinsufficiency in mice are weaker but convergent with those in humans. As in the mouse, the phenotype of human NKX2-5 mutations may be modulated by interacting alleles.


Subject(s)
Heart Septal Defects/genetics , Heart Valves/abnormalities , Heterozygote , Homeodomain Proteins/genetics , Mutation/genetics , Transcription Factors , Xenopus Proteins , Alleles , Animals , Animals, Newborn , Blood Flow Velocity , Echocardiography , Electrocardiography , Genes, Homeobox , Heart Septal Defects/diagnostic imaging , Heart Septal Defects/pathology , Heart Septal Defects, Atrial/diagnostic imaging , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/pathology , Heart Valves/diagnostic imaging , Heart Valves/pathology , Homeobox Protein Nkx-2.5 , Mice , Mice, Inbred Strains , Mice, Transgenic , Mitral Valve/abnormalities , Mitral Valve/diagnostic imaging , Mitral Valve/pathology , Mitral Valve Stenosis/diagnostic imaging , Mitral Valve Stenosis/genetics , Mitral Valve Stenosis/pathology
5.
Dev Biol ; 224(2): 152-67, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10926756

ABSTRACT

Regulated emigration of blood-borne leukocytes plays a defining role in lymphoid organ development, immune surveillance, and inflammatory responses. We report here that mice deficient in the homeobox gene Nkx2-3, expressed in developing visceral mesoderm, show a complex intestinal malabsorption phenotype and striking abnormalities of gut-associated lymphoid tissue and spleen suggestive of deranged leukocyte homing. Mutant Peyer's patches were reduced in number and size, intestinal villi contained few IgA(+) plasma cells, and mutant spleens were small and often atrophic, showing fused periarterial lymphoid sheaths, partially merged T and B cell zones, an absent marginal zone, and a dearth of macrophages in red pulp. Semiquantitative RT-PCR analysis and immunohistochemistry revealed down-regulation of mucosal addressin cell adhesion molecule-1 (MAdCAM-1) in endothelial cells in which Nkx2-3 is normally expressed. MAdCAM-1 is a member of the immunoglobulin superfamily, acting as an endothelial cell ligand for leukocyte homing receptors L-selectin and alpha4beta7 integrin. Our data suggest a role for a homeodomain factor in establishing the developmental and positional cues in endothelia that regulate leukocyte homing through local control of cellular adhesion and identify MAdCAM-1 as a candidate target gene of Nkx2-3.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Immunoglobulins/genetics , Mucoproteins/genetics , Receptors, Lymphocyte Homing/genetics , Zebrafish Proteins , Animals , Base Sequence , Cell Adhesion Molecules , DNA Primers , Endothelium/cytology , Endothelium/metabolism , Homeodomain Proteins/genetics , Lac Operon , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Viscera
6.
Dev Biol ; 223(2): 266-78, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10882515

ABSTRACT

In this study we challenge the generally accepted view that cardiac chambers form from an array of segmental primordia arranged along the anteroposterior axis of the linear and looping heart tube. We traced the spatial pattern of expression of genes encoding atrial natriuretic factor, sarcoplasmic reticulum calcium ATPase, Chisel, Irx5, Irx4, myosin light chain 2v, and beta-myosin heavy chain and related these to morphogenesis. Based on the patterns we propose a two-step model for chamber formation in the embryonic heart. First, a linear heart forms, which is composed of "primary" myocardium that nonetheless shows polarity in phenotype and gene expression along its anteroposterior and dorsoventral axes. Second, specialized ventricular chamber myocardium is specified at the ventral surface of the linear heart tube, while distinct left and right atrial myocardium forms more caudally on laterodorsal surfaces. The process of looping aligns these primordial chambers such that they face the outer curvature. Myocardium of the inner curvature, as well as that of inflow tract, atrioventricular canal, and outflow tract, retains the molecular signature originally found in linear heart tube myocardium. Evidence for distinct transcriptional programs which govern compartmentalization in the forming heart is seen in the patterns of expression of Hand1 for the dorsoventral axis, Irx4 and Tbx5 for the anteroposterior axis, and Irx5 for the distinction between primary and chamber myocardium.


Subject(s)
Heart Atria/embryology , Heart Ventricles/embryology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/isolation & purification , Basic Helix-Loop-Helix Transcription Factors , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/isolation & purification , Connexin 43/genetics , Connexin 43/isolation & purification , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Homeodomain Proteins/genetics , Homeodomain Proteins/isolation & purification , In Situ Hybridization , Models, Biological , Models, Structural , Morphogenesis/genetics , Myosins/genetics , Myosins/isolation & purification , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/isolation & purification , Transcription, Genetic
7.
Genesis ; 26(4): 259-64, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10748464

ABSTRACT

The mouse Cer1 (mCer1, Cer-l, Cerr1) gene encodes one member of a family of cytokines structurally and functionally related to the Xenopus head-inducing factor, Cerberus (xCer). We generated a mouse line in which the Cer1 gene was inactivated by replacing the first coding exon with a lacZ reporter gene. Mice homozygous for this allele (Cer1(lacZ)) showed no apparent perturbation of embryogenesis or later development. However, the lacZ reporter revealed a number of hitherto uncharacterised sites of Cer1 expression in late fetal and adult tissues. Preliminary analysis suggests that Cer1 is not essential for their morphogenesis, differentiation, or homeostasis.


Subject(s)
Arabidopsis Proteins , Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Lac Operon , Plant Proteins/genetics , Animals , Genes, Reporter , Mice , Mice, Transgenic
8.
J Mol Cell Cardiol ; 31(4): 837-43, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10329211

ABSTRACT

LIM proteins perform critical roles in development and tissue differentiation. The skeletal muscle LIM protein 1 (SLIM1) comprises four and a half LIM domains. Northern blot analysis demonstrated high level expression of SLIM1 mRNA in adult human skeletal muscle with intermediate expression in adult heart and lower expression in other tissues. Western blot analysis using specific affinity-purified anti-SLIM1 antipeptide antibodies demonstrated a 32 kDa polypeptide in the aorta and atria of rabbit heart, but not in vena cava, interventricular septum or ventricular muscle. SLIM1 was also demonstrated in rabbit skeletal muscle. In situ hybridization of whole mouse embryos confirmed the cardiac expression of SLIM1 was restricted to the cardiac outflow tract from embryonic day 8.5-11. No expression was seen in atrial or ventricular muscle. SLIM1 mRNA was also demonstrated in the hindbrain, neural tube and somites. The localized expression of SLIM1 to the outflow tract of the embryonic heart implies an important role for the protein in the development of this region and possibly in congenital heart anomalies involving the separation and formation of the aortic and pulmonary trunks.


Subject(s)
Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Adult , Animals , Base Sequence , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Tissue Distribution
9.
Mech Dev ; 77(2): 173-84, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9831647

ABSTRACT

We report that DAN, a potential cell cycle regulator and tumour suppressor, is a secreted glycoprotein related to Xenopus cerberus. DAN, cerberus, its mouse relative Cer-1/cer-l/Cerberus-like/Cerr1, and the recently described factor DRM/Gremlin, appear to be members of the cystine knot superfamily, which includes TGFbetas and BMPs. Like cerberus and mCer-1, DAN-induced cement glands as well as markers of anterior neural tissue and endoderm in Xenopus animal cap assays, features of BMP signalling blockade. During mouse embryogenesis, Dan was expressed from E8.5 in cranial mesenchyme and somites, then later in limb and facial mesenchyme. The pattern in somites was highly dynamic, with transcripts initially localized to the caudal half of the nascent epithelial somite, then, after maturation, to sclerotomal cells adjacent to the neural tube. Dan was also expressed in the developing myotome. The expression domains include sites in which BMP inhibition is known to be important for development. Thus, DAN appears to be a secreted factor belonging to the cystine knot superfamily, and one of a growing number of antagonists acting to modulate BMP signalling during development.


Subject(s)
Gene Expression Regulation, Developmental , Proteins/genetics , Proteins/metabolism , Xenopus Proteins , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cystine , Cytokines , Dimerization , Embryo, Nonmammalian , Embryonic Induction/genetics , Glycosylation , Head/embryology , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Limb Buds , Mesoderm , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Somites/metabolism , Xenopus laevis/genetics , Xenopus laevis/growth & development
10.
Dev Dyn ; 213(1): 105-13, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9733105

ABSTRACT

We report the cloning, chromosomal localization, and analysis of the expression pattern of epicardin, a member of the basic helix-loop-helix (bHLH) family of transcription factors. Within its bHLH domain, the human and murine epicardin genes were most similar to paraxis, a bHLH gene important for segmentation of embryonic paraxial mesoderm. In situ hybridization studies revealed strong epicardin expression in murine embryos at 9.5 days postcoitum (dpc) in a region of the septum transversum at the base of the heart known as the proepicardial organ. This mesenchymal structure extends villous projections from which epicardial precursor cells emerge and migrate out over the surface of the myocardium. Strong expression was seen in individual migratory cells and clusters at 9.5 dpc and in a continuous epicardial cell layer in more mature hearts. Also from 9.5 dpc, epicardin transcripts were seen in endocardial cushions of the atrioventricular canal and outflow tract, in skeletal myoblasts within branchial arches and in condensing mesenchyme of gut, kidney, urinary tract, gonads, spleen, and lung. Northern analysis showed that expression persisted in mature visceral organs and heart, but was transient in skeletal muscle. The central role played by bHLH factors in pathways for tissue determination in the embryo suggests a function for epicardin in specification of select mesodermal cell populations associated with heart, cranial skeletal muscle, gut, and urogenital system.


Subject(s)
Helix-Loop-Helix Motifs , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Blotting, Northern , Branchial Region/embryology , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Digestive System/embryology , Female , Gene Expression , Gonads/embryology , Humans , Kidney/embryology , Lung/embryology , Male , Mesoderm , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pericardium/embryology , Sequence Homology, Amino Acid
11.
Mech Dev ; 73(1): 125-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9545560

ABSTRACT

NK-2 class homeobox genes are candidate patterning and lineage regulators in diverse organisms. We report here the embryonic expression pattern of murine member, Nkx2-6. In keeping with its vertebrate relatives, Nkx2-6 was transcribed in ventrolateral embryonic structures. Expression was first detected at E8.0 in endodermal walls of the foregut pocket, tissue destined to become pharyngeal floor. From E8.0-10.5, transcripts were concentrated in pharyngeal pouches and juxtaposed arch ectoderm and mesoderm, as well as in more caudal gut segments. Expression was also seen at opposite poles of the developing heart from E8-8.5 in posterior myocardial progenitors, then sinus venosa and dorsal pericardium, and from E9.5 in outflow tract myocardium.


Subject(s)
Endoderm/metabolism , Genes, Homeobox/genetics , Heart/embryology , Homeodomain Proteins/genetics , Intestines/embryology , Animals , Drosophila Proteins , Homeodomain Proteins/biosynthesis , In Situ Hybridization , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Transcription Factors
12.
Dev Biol ; 194(2): 135-51, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9501024

ABSTRACT

Xenopus cerberus (Xcer) is a cytokine expressed in anterior mesendoderm overlapping and surrounding Spemann's gastrula organiser. When misexpressed in blastomeres, Xcer can induce ectopic heads with well-defined brain, cement gland, olfactory placodes, cyclopic eye, and occasionally liver and heart. We report here the identification of mCer-1, a murine gene related to cerberus. Both mCer-1 and Xcer appear to belong to the cystine knot superfamily, which includes TGF beta s and BMPs. In Xenopus animal cap assays, mCer-1 and Xcer induced cement glands and markers of anterior neural tissue and endoderm, characteristic of BMP inhibition. Furthermore, both antagonised the ventrolateral mesoderm-inducing activity of coexpressed BMP4. In mouse embryos, mCer-1 was expressed at early gastrulation in a stripe of primitive endoderm along the future anterior side of the egg cylinder, a region essential for anterior patterning. A second phase of expression was detected in anterior embryonic mesendoderm, and by late-streak stages most of the anterior half of the embryo was positive, except for the node and cardiac progenitors. Expression was later seen in the cranial portion of the two most-recently formed somites and in two stripes within presomitic mesoderm. In embryos lacking Otx2, a homeogene with a demonstrated role in anterior patterning, mCer-1 was still expressed in an anterior zone, although often abnormally. The data suggest that mCer-1 shares structural, functional, and expression characteristics with Xcer and may participate in patterning the anterior of the embryo and nascent somite region, in part, through a BMP-inhibitory mechanism.


Subject(s)
Body Patterning/genetics , Proteins/physiology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cytokines , Databases, Factual , Dimerization , Embryonic Induction , Endoderm/metabolism , Gene Expression Regulation, Developmental , Gene Library , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otx Transcription Factors , Proteins/genetics , Sequence Alignment , Trans-Activators/genetics , Trans-Activators/metabolism , Xenopus Proteins
13.
Dev Biol ; 187(2): 183-99, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9242416

ABSTRACT

Embryonic and fetal stages of skeletal muscle development are characterized by the differential expression of a number of muscle-specific genes. These include the products of independent promoters at the fast myosin light chain 1F/3F locus. In the mouse embryo MLC1F transcripts accumulate in embryonic skeletal muscle from E9, 4-5 days before high-level accumulation of MLC3F transcripts. A 3' enhancer can activate MLC1F and MLC3F promoters in differentiated muscle cells in vitro and in transgenic mice; both promoters, however, are activated at the time of MLC1F transcript accumulation. We now demonstrate the presence of a second muscle-specific enhancer at this locus, located in the intron separating the MLC1F and MLC3F promoters. Transgenic mice containing the intronic, but lacking the 3' enhancer, express high levels of an nlacZ reporter gene from the MLC3F promoter in adult fast skeletal muscle fibers. In contrast to the 3' enhancer, the intronic element is inactive both in embryonic muscle cells in vivo and in embryonic myocyte cultures. The intronic enhancer is activated at the onset of fetal development in both primary and secondary muscle fibers, at the time of endogenous MLC3F transcript accumulation. Late-activated MLC3F transgenes thus provide a novel in toto marker of fetal myogenesis. These results suggest that temporal regulation of transcription at the MLC1F/3F locus is controlled by separate enhancers which are differentially activated during embryonic and fetal development.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Muscle, Skeletal/embryology , Myosin Light Chains/genetics , Animals , Base Sequence , Cell Differentiation , Embryo, Mammalian , Fetus , Fluorescent Antibody Technique , In Situ Hybridization , Introns , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Analysis, DNA , Time Factors , Transcription, Genetic
14.
Genes Dev ; 11(11): 1357-69, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9192865

ABSTRACT

One of the first morphological manifestations of left/right (L/R) asymmetry in mammalian embryos is a pronounced rightward looping of the linear heart tube. The direction of looping is thought to be controlled by signals from an embryonic L/R axial system. We report here that morphological L/R asymmetry in the murine heart first became apparent at the linear tube stage as a leftward displacement of its caudal aspect. Beginning at the same stage, the basic helix-loop-helix (bHLH) factor gene eHand was expressed in a strikingly left-dominant pattern in myocardium, reflecting an intrinsic molecular asymmetry. In hearts of embryos lacking the homeobox gene Nkx2-5, which do not loop, left-sided eHand expression was abolished. However, expression was unaffected in Sc1-/- hearts that loop poorly because of hematopoietic insufficiency, and was right-sided in hearts of inv/inv embryos that display situs inversus. The data predict that eHand expression is enhanced in descendants of the left heart progenitor pool as one response to inductive signaling from the L/R axial system, and that eHand controls intrinsic morphogenetic pathways essential for looping. One aspect of the intrinsic response to L/R information falls under Nkx2-5 homeobox control.


Subject(s)
DNA-Binding Proteins/biosynthesis , Heart/embryology , Helix-Loop-Helix Motifs , Homeodomain Proteins/genetics , Transcription Factors/biosynthesis , Xenopus Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Genotype , Heart/anatomy & histology , Homeobox Protein Nkx-2.5 , In Situ Hybridization , Mice , Models, Genetic , Morphogenesis/genetics , Mutation , Tissue Distribution
16.
Dev Biol ; 173(1): 200-12, 1996 Jan 10.
Article in English | MEDLINE | ID: mdl-8575622

ABSTRACT

Cardiac actin is an early marker of cardiac and skeletal muscle lineages in the mouse. After birth, the gene is down-regulated in skeletal muscle. High-level expression of the murine cardiac actin gene in skeletal myotubes in vitro involves distal (-7.8/-7.0 kb) and proximal (-5.4/-3.5 kb) enhancer sequences as well as the proximal promoter (-0.7/+0.1 kb). Transgenic mice carrying an nlacZ reporter gene under the control of different fragments of the upstream region of the cardiac actin gene were generated. This analysis led to the conclusions that (1) the proximal promoter is a weak but tissue specific element in vivo, (2) consistent high-level expression in skeletal muscle depends on the presence of at least one of the enhancers, (3) expression in adult cardiac muscle requires a cardiac enhancer located in the (-5.4/-0.7 kb) region, and (4) a construct containing these three elements gives a strong specific expression of the transgene in the heart throughout the life of the animal and in embryonic skeletal muscle. All transgenes tested reproduce the down-regulation observed in adult skeletal muscle for the cardiac actin gene. Nonuniform expression of these transgenes in the heart may mark cardiomyocytes derived from different cardiac progenitors.


Subject(s)
Actins/biosynthesis , Gene Expression Regulation, Developmental , Heart/embryology , Muscle, Skeletal/embryology , Regulatory Sequences, Nucleic Acid , Actins/genetics , Animals , Enhancer Elements, Genetic , Female , Genes, Reporter , Lac Operon , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Promoter Regions, Genetic , Tissue Distribution , Transcription, Genetic
17.
Mol Cell Biol ; 14(5): 3504-13, 1994 May.
Article in English | MEDLINE | ID: mdl-8164695

ABSTRACT

A DNase I-hypersensitive site analysis of the 5'-flanking region of the mouse alpha-cardiac actin gene with muscle cell lines derived from C3H mice shows the presence of two such sites, at about -5 and -7 kb. When tested for activity in cultured cells with homologous and heterologous promoters, both sequences act as muscle-specific enhancers. Transcription from the proximal promoter of the alpha-cardiac actin gene is increased 100-fold with either enhancer. The activity of the distal enhancer in C2/7 myotubes is confined to an 800-bp fragment, which contains multiple E boxes. In transfection assays, this sequence does not give detectable transactivation by any of the myogenic factors even though one of the E boxes is functionally important. Bandshift assays showed that MyoD and myogenin can bind to this E box. However, additional sequences are also required for activity. We conclude that in the case of this muscle enhancer, myogenic factors alone are not sufficient to activate transcription either directly via an E box or indirectly through activation of genes encoding other muscle factors. In BALB/c mice, in which cardiac actin mRNA levels are 8- to 10-fold lower, the alpha-cardiac actin locus is perturbed by a 9.5-kb insertion (I. Garner, A. J. Minty, S. Alonso, P. J. Barton, and M. E. Buckingham, EMBO J. 5:2559-2567, 1986). This is located at -6.5 kb, between the two enhancers. The insertion therefore distances the distal enhancer from the promoter and from the proximal enhancer of the bona fide cardiac actin gene, probably thus perturbing transcriptional activity.


Subject(s)
Actins/genetics , DNA/genetics , Enhancer Elements, Genetic , Muscles/metabolism , Myocardium/metabolism , RNA, Messenger/biosynthesis , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/metabolism , Cloning, Molecular , DNA/isolation & purification , DNA/metabolism , DNA Probes , Deoxyribonuclease I , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Oligonucleotide Probes , Restriction Mapping , Transfection
18.
Nucleic Acids Res ; 17(12): 4745-55, 1989 Jun 26.
Article in English | MEDLINE | ID: mdl-2501761

ABSTRACT

Enhancer activity of the rabbit immunoglobulin kappa light chain gene intron conserved region (KICR) was examined in mouse myeloma cells using transient expression experiments. Compared to the homologous region of the mouse kappa light chain gene, the rabbit KICR shows nearly no stimulatory effect on expression of the indicator gene, cat. Experiments with mouse-rabbit chimeric KICRs indicated that differences in the region around the NF-kappa B binding site are responsible for the impaired activity of the rabbit KICR whereas mouse sequences covering the kappa E2 and kappa E3 motifs can be replaced by the equivalent rabbit fragment without affecting enhancer function. Creation of a perfect mouse NF-kappa B target sequence in the rabbit gene only partially restores enhancer activity. Furthermore, mouse and rabbit DNA fragments encompassing the NF-kappa B target sequence behave in an identical manner in an electrophoretic mobility shift assay. The results indicate species-related functional differences in the immunoglobulin kappa light chain gene enhancer and suggest that although the NF-kappa B binding site plays a crucial role in enhancer activity surrounding gene elements are also necessary for full enhancer effect.


Subject(s)
Enhancer Elements, Genetic , Immunoglobulin kappa-Chains/genetics , Plasmacytoma/genetics , Animals , Base Sequence , Chimera , DNA-Binding Proteins/genetics , Immunoglobulin kappa-Chains/metabolism , Introns , Mice , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Plasmacytoma/immunology , Plasmacytoma/metabolism , Rabbits , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL