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










Database
Language
Publication year range
1.
J Cardiovasc Transl Res ; 5(5): 654-65, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22566269

ABSTRACT

Recent reports indicate that the adult mammalian heart is capable of limited, but measurable, cardiomyocyte turnover. While the lineage origin of the newly formed cardiomyocytes is not entirely understood, mounting evidence suggest that the epicardium and subepicardium may represent an important source of cardiac stem or progenitor cells. Stem cell niches are characterized by low oxygen tension, where stem cells preferentially utilize cytoplasmic glycolysis to meet their energy demands. However, it is unclear if the heart harbors similar hypoxic regions, or whether these regions house metabolically distinct cardiac progenitor populations. Here we identify the epicardium and subepicardium as the cardiac hypoxic niche based on [corrected] capillary density quantification, and localization of Hif-1α in the uninjured heart. We further demonstrate that this hypoxic microenvironment houses a metabolically distinct population of glycolytic progenitor cells. Finally, we show that Hif-1α regulates the glycolytic phenotype and progenitor properties of these cells. These findings highlight important anatomical and functional properties of the epicardial and subepicardial microenvironment, and the potential role of hypoxia signaling in regulation of cardiac progenitors.


Subject(s)
Cellular Microenvironment , Energy Metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Pericardium/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Hypoxia , Cell Lineage , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gene Expression Regulation , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Oxidative Stress , Perfusion , Phenotype , RNA Interference , Signal Transduction , Transfection
2.
Am J Hum Genet ; 87(2): 289-96, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20691403

ABSTRACT

The focal facial dermal dysplasias (FFDDs) are a group of inherited developmental disorders in which the characteristic diagnostic feature is bitemporal scar-like lesions that resemble forceps marks. To date, the genetic defects underlying these ectodermal dysplasias have not been determined. To identify the gene defect causing autosomal-recessive Setleis syndrome (type III FFDD), homozygosity mapping was performed with genomic DNAs from five affected individuals and 26 members of the consanguineous Puerto Rican (PR) family originally described by Setleis and colleagues. Microsatellites D2S1397 and D2S2968 were homozygous in all affected individuals, mapping the disease locus to 2q37.3. Haplotype analyses of additional markers in the PR family and a consanguineous Arab family further limited the disease locus to approximately 3 Mb between D2S2949 and D2S2253. Of the 29 candidate genes in this region, the bHLH transcription factor, TWIST2, was initially sequenced on the basis of its known involvement in murine facial development. Homozygous TWIST2 nonsense mutations, c.324C>T and c.486C>T, were identified in the affected members of the Arab and PR families, respectively. Characterization of the expressed mutant proteins, p.Q65X and p.Q119X, by electrophoretic mobility shift assays and immunoblot analyses indicated that they were truncated and unstable. Notably, Setleis syndrome patients and Twist2 knockout mice have similar facial features, indicating the gene's conserved role in mammalian development. Although human TWIST2 and TWIST1 encode highly homologous bHLH transcription factors, the finding that TWIST2 recessive mutations cause an FFDD and dominant TWIST1 mutations cause Saethre-Chotzen craniocynostosis suggests that they function independently in skin and bone development.


Subject(s)
Abnormalities, Multiple/genetics , Codon, Nonsense/genetics , Homozygote , Repressor Proteins/genetics , Twist-Related Protein 1/genetics , Amino Acid Sequence , Animals , Base Sequence , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 3/genetics , Facies , Female , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/chemistry , Pedigree , Phenotype , Puerto Rico , Repressor Proteins/chemistry , Sequence Alignment , Syndrome , Twist-Related Protein 1/chemistry , United Arab Emirates
3.
Invest Ophthalmol Vis Sci ; 51(11): 5561-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20574024

ABSTRACT

PURPOSE: Twist2 is a member of a family of bHLH transcription factors critical for normal mesenchymal proliferation and differentiation. In this study, the authors analyzed the role of Twist2 in the eye and cornea through examination of a Twist2 loss-of-function mouse mutant. METHODS: Twist2 expression during eye development in the mouse was investigated using RT-PCR and mRNA slide in situ hybridization. Lineage tracing was performed using Cre reporter mice. Morphometric analyses were performed, and cell proliferation and cell death were investigated by immunohistochemistry using Ki67 and cleaved caspase 3 antibodies, respectively. RESULTS: In the mouse, Twist2 is expressed first in the periocular mesenchyme and subsequently in the corneal stroma and endothelium of the developing eye. Loss of Twist2 function leads to corneal thinning and a reduced population of stromal keratocytes. The reduction in the stromal cell population can be traced back to embryonic stages during which the proliferation of stromal progenitor cells is impaired and to the reduced number of proliferating cells in the corneal limbus postnatally. Adult Twist2-null mice display enophthalmia and blepharophimosis. Corneal thinning in mutant mice is not accompanied by glaucoma, an association reported in human patients. CONCLUSIONS: Twist2 is required for normal corneal keratocyte proliferation and eyelid morphogenesis in the mouse. Loss of Twist2 function leads to corneal thinning because of the reduction in stromal keratocyte proliferation.


Subject(s)
Cell Proliferation , Cornea/embryology , Cornea/pathology , Corneal Stroma/embryology , Repressor Proteins/physiology , Twist-Related Protein 1/physiology , Animals , Animals, Newborn , Apoptosis , Blepharophimosis/genetics , Blepharophimosis/pathology , Caspase 3/metabolism , Cell Differentiation/physiology , Corneal Stroma/metabolism , Enophthalmos/genetics , Enophthalmos/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Helix-Loop-Helix Motifs/physiology , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
PLoS Biol ; 6(12): e316, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19090621

ABSTRACT

Basic helix-loop-helix (bHLH) transcription factors play critical roles in lymphoid and erythroid development; however, little is known about their role in myeloid lineage development. In this study, we identify the bHLH transcription factor Twist-2 as a key negative regulator of myeloid lineage development, as manifested by marked increases in mature myeloid populations of macrophages, neutrophils, and basophils in Twist-2-deficient mice. Mechanistic studies demonstrate that Twist-2 inhibits the proliferation as well as differentiation of granulocyte macrophage progenitors (GMP) by interacting with and inhibiting the transcription factors Runx1 and C/EBPalpha. Moreover, Twist-2 was found to have a contrasting effect on cytokine production: inhibiting the production of proinflammatory cytokines such as interleukin-12 (IL-12) and interferon-gamma (IFNgamma) while promoting the regulatory cytokine IL-10 by myeloid cells. The data from further analyses suggest that Twist-2 activates the transcription factor c-Maf, leading to IL-10 expression. In addition, Twist-2 was found to be essential for endotoxin tolerance. Thus, this study reveals the critical role of Twist-2 in regulating the development of myeloid lineages, as well as the function and inflammatory responses of mature myeloid cells.


Subject(s)
Myeloid Progenitor Cells/physiology , Repressor Proteins/physiology , Twist-Related Protein 1/physiology , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/physiology , Cell Lineage , Core Binding Factor Alpha 2 Subunit/metabolism , Endotoxins/pharmacology , Interferon-gamma/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-12/antagonists & inhibitors , Mice , Mice, Knockout , Proto-Oncogene Proteins c-maf/biosynthesis , Repressor Proteins/biosynthesis , Twist-Related Protein 1/biosynthesis
5.
J Exp Med ; 203(8): 1891-901, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16831897

ABSTRACT

Type I interferons (IFNs) are pleiotropic cytokines with antiviral and immunomodulatory properties. The immunosuppressive actions of type I IFNs are poorly understood, but IFN-mediated suppression of TNFalpha production has been implicated in the regulation of inflammation and contributes to the effectiveness of type I IFNs in the treatment of certain autoimmune and inflammatory diseases. In this study, we investigated mechanisms by which type I IFNs suppress induction of TNFalpha production by immune complexes, Fc receptors, and Toll-like receptors. Suppression of TNFalpha production was mediated by induction and activation of the Axl receptor tyrosine kinase and downstream induction of Twist transcriptional repressors that bind to E box elements in the TNF promoter and suppress NF-kappaB-dependent transcription. Twist expression was activated by the Axl ligand Gas6 and by protein S and apoptotic cells. These results implicate Twist proteins in regulation of TNFalpha production by antiinflammatory factors and pathways, and provide a mechanism by which type I IFNs and Axl receptors suppress inflammatory cytokine production.


Subject(s)
Inflammation/immunology , Interferon-alpha/pharmacology , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Twist-Related Protein 1/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Macrophages/drug effects , Mice , Models, Biological , Oncogene Proteins/genetics , Proto-Oncogene Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fc/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Axl Receptor Tyrosine Kinase
6.
Dev Cell ; 6(3): 423-35, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15030764

ABSTRACT

Runx2 is necessary and sufficient for osteoblast differentiation, yet its expression precedes the appearance of osteoblasts by 4 days. Here we show that Twist proteins transiently inhibit Runx2 function during skeletogenesis. Twist-1 and -2 are expressed in Runx2-expressing cells throughout the skeleton early during development, and osteoblast-specific gene expression occurs only after their expression decreases. Double heterozygotes for Twist-1 and Runx2 deletion have none of the skull abnormalities observed in Runx2(+/-) mice, a Twist-2 null background rescues the clavicle phenotype of Runx2(+/-) mice, and Twist-1 or -2 deficiency leads to premature osteoblast differentiation. Furthermore, Twist-1 overexpression inhibits osteoblast differentiation without affecting Runx2 expression. Twist proteins' antiosteogenic function is mediated by a novel domain, the Twist box, which interacts with the Runx2 DNA binding domain to inhibit its function. In vivo mutagenesis confirms the antiosteogenic function of the Twist box. Thus, relief of inhibition by Twist proteins is a mandatory event precluding osteoblast differentiation.


Subject(s)
Cell Differentiation/physiology , Myogenic Regulatory Factors/physiology , Nuclear Proteins/physiology , Osteoblasts/physiology , Osteogenesis/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Aging/metabolism , Animals , Animals, Newborn , Blotting, Northern/methods , Blotting, Western/methods , Cells, Cultured , Chlorocebus aethiops , Core Binding Factor Alpha 1 Subunit , DNA Mutational Analysis/methods , Electrophoretic Mobility Shift Assay/methods , Embryo, Mammalian , Gene Expression Regulation/physiology , Heterozygote , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myogenic Regulatory Factors/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Precipitin Tests/methods , Proline/genetics , Protein Structure, Tertiary/physiology , RNA/analysis , Rats , Repressor Proteins/genetics , Serine/genetics , Skeleton , Staining and Labeling , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/physiology , Transfection/methods , Twist-Related Protein 1
7.
Development ; 130(13): 3063-74, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12756187

ABSTRACT

Human craniosynostosis syndromes, resulting from activating or neomorphic mutations in fibroblast growth factor receptor 2 (FGFR2), underscore an essential role for FGFR2 signaling in skeletal development. Embryos harboring homozygous null mutations in FGFR2 die prior to skeletogenesis. To address the role of FGFR2 in normal bone development, a conditional gene deletion approach was adopted. Homologous introduction of cre recombinase into the Dermo1 (Twist2) gene locus resulted in robust expression of CRE in mesenchymal condensations giving rise to both osteoblast and chondrocyte lineages. Inactivation of a floxed Fgfr2 allele with Dermo1-cre resulted in mice with skeletal dwarfism and decreased bone density. Although differentiation of the osteoblast lineage was not disturbed, the proliferation of osteoprogenitors and the anabolic function of mature osteoblasts were severely affected.


Subject(s)
Bone Development/physiology , Fibroblast Growth Factors/metabolism , Osteoblasts/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Repressor Proteins , Signal Transduction/physiology , Animals , Bone Density , Bone and Bones/diagnostic imaging , Cell Differentiation/physiology , Cell Lineage , Chondrocytes/cytology , Chondrocytes/physiology , Craniosynostoses/genetics , Craniosynostoses/metabolism , Gene Deletion , Humans , In Situ Hybridization , Integrases/genetics , Integrases/metabolism , Mice , Mice, Knockout , Osteoblasts/cytology , Phenotype , Radiography , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1 , Viral Proteins/genetics , Viral Proteins/metabolism
9.
Cell ; 112(2): 169-80, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12553906

ABSTRACT

During Drosophila embryogenesis, the dorsal transcription factor activates the expression of twist, a transcription factor required for mesoderm formation. We show here that the mammalian twist proteins, twist-1 and -2, are induced by a cytokine signaling pathway that requires the dorsal-related protein RelA, a member of the NF-kappaB family of transcription factors. Twist-1 and -2 repress cytokine gene expression through interaction with RelA. Mice homozygous for a twist-2 null allele or doubly heterozygous for twist-1 and -2 alleles show elevated expression of proinflammatory cytokines, resulting in perinatal death from cachexia. These findings reveal an evolutionarily conserved signaling circuit in which twist proteins regulate cytokine signaling by establishing a negative feedback loop that represses the NF-kappaB-dependent cytokine pathway.


Subject(s)
Cytokines/genetics , Feedback, Physiological , Gene Expression Regulation, Developmental , NF-kappa B/antagonists & inhibitors , Nuclear Proteins/metabolism , Transcription Factors , Animals , Apoptosis/drug effects , Cachexia/genetics , Cytokines/pharmacology , Genes, Lethal , Inflammation/genetics , Mice , Mice, Knockout , Mutation/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Signal Transduction/drug effects , Skin/pathology , Transcription Factor RelA , Transcription, Genetic , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology , Twist-Related Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...