Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Immunol ; 12(2): 129-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186366

ABSTRACT

The nuclear adaptor Ldb1 functions as a core component of multiprotein transcription complexes that regulate differentiation in diverse cell types. In the hematopoietic lineage, Ldb1 forms a complex with the non-DNA-binding adaptor Lmo2 and the transcription factors E2A, Scl and GATA-1 (or GATA-2). Here we demonstrate a critical and continuous requirement for Ldb1 in the maintenance of both fetal and adult mouse hematopoietic stem cells (HSCs). Deletion of Ldb1 in hematopoietic progenitors resulted in the downregulation of many transcripts required for HSC maintenance. Genome-wide profiling by chromatin immunoprecipitation followed by sequencing (ChIP-Seq) identified Ldb1 complex-binding sites at highly conserved regions in the promoters of genes involved in HSC maintenance. Our results identify a central role for Ldb1 in regulating the transcriptional program responsible for the maintenance of HSCs.


Subject(s)
Adult Stem Cells/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Fetal Development , Hematopoietic Stem Cells/metabolism , Adoptive Transfer , Adult Stem Cells/cytology , Adult Stem Cells/immunology , Adult Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Female , Fetal Development/genetics , Fetal Development/immunology , Gene Expression Regulation, Developmental/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , High-Throughput Nucleotide Sequencing , LIM Domain Proteins , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Protein Binding , Regulatory Elements, Transcriptional/genetics , Regulatory Elements, Transcriptional/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
2.
J Biol Chem ; 285(16): 12344-54, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20139090

ABSTRACT

A fundamental biologic principle is that diverse biologic signals are channeled through shared signaling cascades to regulate development. Large scaffold proteins that bind multiple proteins are capable of coordinating shared signaling pathways to provide specificity to activation of key developmental genes. Although much is known about transcription factors and target genes that regulate cardiomyocyte differentiation, less is known about scaffold proteins that couple signals at the cell surface to differentiation factors in developing heart cells. Here we show that AKAP13 (also known as Brx-1, AKAP-Lbc, and proto-Lbc), a unique protein kinase A-anchoring protein (AKAP) guanine nucleotide exchange region belonging to the Dbl family of oncogenes, is essential for cardiac development. Cardiomyocytes of Akap13-null mice had deficient sarcomere formation, and developing hearts were thin-walled and mice died at embryonic day 10.5-11.0. Disruption of Akap13 was accompanied by reduced expression of Mef2C. Consistent with a role of AKAP13 upstream of MEF2C, Akap13 siRNA led to a reduction in Mef2C mRNA, and overexpression of AKAP13 augmented MEF2C-dependent reporter activity. The results suggest that AKAP13 coordinates Galpha(12) and Rho signaling to an essential transcription program in developing cardiomyocytes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Fetal Heart/embryology , Fetal Heart/metabolism , Guanine Nucleotide Exchange Factors/metabolism , A Kinase Anchor Proteins/antagonists & inhibitors , A Kinase Anchor Proteins/deficiency , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Female , Fetal Heart/abnormalities , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , In Situ Hybridization , MEF2 Transcription Factors , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Minor Histocompatibility Antigens , Models, Cardiovascular , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Pregnancy , RNA, Small Interfering/genetics , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Signal Transduction , rho GTP-Binding Proteins/metabolism
3.
Dev Biol ; 325(1): 24-32, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18930042

ABSTRACT

IFT172, also known as Selective Lim-domain Binding protein (SLB), is a component of the intraflagellar transport (IFT) complex. In order to evaluate the biological role of the Ift172 gene, we generated a loss-of-function mutation in the mouse. The resulting Slb mutant embryos die between E12.5 and 13.0, and exhibit severe cranio-facial malformations, failure to close the cranial neural tube, holoprosencephaly, heart edema and extensive hemorrhages. Cilia outgrowth in cells of the neuroepithelium is initiated but the axonemes are severely truncated and do not contain visible microtubules. Morphological and molecular analyses revealed a global brain-patterning defect along the dorsal-ventral (DV) and anterior-posterior (AP) axes. We demonstrate that Ift172 gene function is required for early regulation of Fgf8 at the midbrain-hindbrain boundary and maintenance of the isthmic organizer. In addition, Ift172 is required for proper function of the embryonic node, the early embryonic organizer and for formation of the head organizing center (the anterior mesendoderm, or AME). We propose a model suggesting that forebrain and mid-hindbrain growth and AP patterning depends on the early function of Ift172 at gastrulation. Our data suggest that the formation and function of the node and AME in the mouse embryo relies on an indispensable role of Ift172 in cilia morphogenesis and cilia-mediated signaling.


Subject(s)
Body Patterning , Brain/embryology , Cilia/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mammals/embryology , Adaptor Proteins, Signal Transducing , Animals , Biomarkers/metabolism , Brain/pathology , Brain/ultrastructure , Cell Death , Cilia/ultrastructure , Cytoskeletal Proteins , Embryo, Mammalian/abnormalities , Endoderm/embryology , Endoderm/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Targeting , Hedgehog Proteins/metabolism , Holoprosencephaly/embryology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Nodal Protein/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
4.
Stem Cells ; 26(6): 1490-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18388304

ABSTRACT

LIM-domain binding protein 1 (Ldb1) is a multiadaptor protein that mediates the action of transcription factors, including LIM-homeodomain proteins. To elucidate the functional role of Ldb1 in the neuronal differentiation of embryonic stem (ES) cells, we have generated Ldb1-null mutant (Ldb1-/-) ES cells and examined neuronal differentiation potentials in vitro using two different neuronal differentiation protocols. When subjected to a five-stage protocol that recapitulates in vivo conditions of neuronal differentiation, wild-type ES cells differentiated into a wide spectrum of neuronal cell types. However, Ldb1-/- ES cells did not differentiate into neuronal cells; instead, they differentiated into sarcomeric alpha-actinin-positive muscle cells. In contrast, when an adherent monolayer culture procedure (which is based on the default mechanism of neural induction and eliminates environmental influences) was applied, both wild-type and Ldb1-/- ES cells differentiated into MAP2-positive mature neurons. Comparison of the results obtained when two different neuronal differentiation protocols were used suggests that Ldb1-/- ES cells have an innate potential to differentiate into neuronal cells, but this potential can be inhibited by environmental influences. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Embryonic Stem Cells/cytology , Neurons/cytology , Neurons/physiology , Animals , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic Stem Cells/physiology , Immunohistochemistry , LIM Domain Proteins , Mice , Mice, Knockout , Prosencephalon/cytology , Prosencephalon/embryology , Prosencephalon/physiology , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
5.
Mol Cancer ; 5: 57, 2006 Nov 05.
Article in English | MEDLINE | ID: mdl-17083744

ABSTRACT

BACKGROUND: The MUC1 gene encodes a mucin glycoprotein(s) which is basally expressed in most epithelial cells. In breast adenocarcinoma and a variety of epithelial tumors its transcription is dramatically upregulated. Of particular relevance to breast cancer, steroid hormones also stimulate the expression of the MUC1 gene. The MUC1 gene directs expression of several protein isoforms, which participate in many crucial cell processes. Although the MUC1 gene plays a critical role in cell physiology and pathology, little is known about its promoter organization and transcriptional regulation. The goal of this study was to provide insight into the structure and transcriptional activity of the MUC1 promoter. RESULTS: Using TRANSFAC and TSSG soft-ware programs the transcription factor binding sites of the MUC1 promoter were analyzed and a map of transcription cis-elements was constructed. The effect of different MUC1 promoter regions on MUC1 gene expression was monitored. Different regions of the MUC1 promoter were analyzed for their ability to control expression of specific MUC1 isoforms. Differences in the expression of human MUC1 gene transfected into mouse cells (heterologous artificial system) compared to human cells (homologous natural system) were observed. The role of estrogen on MUC1 isoform expression in human breast cancer cells, MCF-7 and T47D, was also analyzed. It was shown for the first time that synthesis of MUC1/SEC is dependent on estrogen whereas expression of MUC1/TM did not demonstrate such dependence. Moreover, the estrogen receptor alpha, ERalpha, could bind in vitro estrogen responsive cis-elements, EREs, that are present in the MUC1 promoter. The potential roles of different regions of the MUC1 promoter and ER in regulation of MUC1 gene expression are discussed. CONCLUSION: Analysis of the structure and transcriptional activity of the MUC1 promoter performed in this study helps to better understand the mechanisms controlling transcription of the MUC1 gene. The role of different regions of the MUC1 promoter in expression of the MUC1 isoforms and possible function of ERalpha in this process has been established. The data obtained in this study may help in development of molecular modalities for controlled regulation of the MUC1 gene thus contributing to progress in breast cancer gene therapy.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Estrogen Receptor alpha/physiology , Gene Expression Regulation, Neoplastic , Mucins/genetics , Promoter Regions, Genetic , Animals , Antigens, Neoplasm/metabolism , Binding Sites , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogens/physiology , Humans , Mice , Mucin-1 , Mucins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Response Elements , Transcription Factors/metabolism , Transcription, Genetic
6.
Stem Cells Dev ; 25(13): 986-94, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27171818

ABSTRACT

LIM domain-binding protein 1 (Ldb1) is a nuclear cofactor that interacts with LIM homeodomain proteins to form multiprotein complexes that are important for transcription regulation. Ldb1 has been shown to play essential roles in various processes during mouse embryogenesis. To determine the role of Ldb1 in mid- and hindbrain development, we have generated a conditional mutant with a specific deletion of the Ldb1 in the Engrailed-1-expressing region of the developing mid- and hindbrain. Our study showed that the deletion impaired the expression of signaling molecules, such as fibroblast growth factor 8 (FGF8) and Wnt1, in the isthmic organizer and the expression of Shh in the ventral midbrain. The midbrain and the cerebellum were severely reduced in size, and the midbrain dopaminergic (mDA) neurons were missing in the mutant. These defects are identical to the phenotype that has been observed previously in mice with a deletion of the LIM homeodomain gene Lmx1b. Our results thus provide genetic evidence supporting that Ldb1 and Lmx1b function cooperatively to regulate mid- and hindbrain development. In addition, we found that mouse embryonic stem cells lacking Ldb1 failed to generate several types of differentiated neurons, including the mDA neurons, serotonergic neurons, cholinergic neurons, and olfactory bulb neurons, indicating an essential cell-autonomous role for Ldb1 in the development of these neurons.


Subject(s)
DNA-Binding Proteins/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , LIM Domain Proteins/metabolism , Mesencephalon/cytology , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cerebellum/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mesencephalon/embryology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mutation/genetics
7.
PLoS One ; 9(4): e95256, 2014.
Article in English | MEDLINE | ID: mdl-24755910

ABSTRACT

Islet1 belongs to Lim homeobox (Lhx) gene family which encodes transcription factors that have been conserved in evolution. They form complexes with other transcriptional regulators, among them obligatory co-factors encoded by Ldb genes. Isl1 (Islet1), Lhx and Ldb1 genes play a crucial role in organ patterning, cell fate determination and cell differentiation in both embryonic and adult tissues. In this study we analyzed expression pattern of Isl1 and its co-factor Ldb1 in small intestine. We also studied the biological role of Ldb1 in gut endoderm. Quantitative PCR analysis revealed a relatively high level of expression of Lhx1, Isl1, Isl2, Lmx1a, Ldb1 and Ldb2 mRNAs in the gut tissue as compared to the level of less abundant detectable Lmx1b mRNA. Immunohistochemical studies demonstrated a unique pattern of Ldb1 and Islet1 proteins in the crypt compartment. Ldb1 is produced at a low level in majority of crypt cells; but, its abundant expression was demonstrated for some single cells. Islet1 is also expressed in single cells of the crypt. Double staining experiments with Ldb1 and Isl1 antibodies showed that both genes are co-expressed in certain cells of the crypt. Further analysis revealed the Ldb1-expressing cells in the gut are both of endodermal and mesodermal origin. Proliferation studies using antibodies to phospho-histone H3 and Ki-67 antigens, as well as long-term BrdU labeling, showed that cells prominently expressing Ldb1/Islet1 are quiescent but do not belong to any known terminally differentiated cell lineages. They may represent a group of stem-like cells in the crypt. Further experiments by cell lineage tracing should be performed to better characterize this cell population. Functional studies of mice with Ldb1 gene ablated in gut endoderm revealed no specific role of Ldb1 in that tissue.


Subject(s)
Cell Cycle , DNA-Binding Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Compartmentation , Cell Proliferation , Endoderm/metabolism , Gene Targeting , Integrases/metabolism , Intestine, Small/embryology , Intestine, Small/metabolism , Mice, Inbred C57BL , Mutation/genetics , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Development ; 136(8): 1375-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19304889

ABSTRACT

Vertebrate limb development is controlled by three signaling centers that regulate limb patterning and growth along the proximodistal (PD), anteroposterior (AP) and dorsoventral (DV) limb axes. Coordination of limb development along these three axes is achieved by interactions and feedback loops involving the secreted signaling molecules that mediate the activities of these signaling centers. However, it is unknown how these signaling interactions are processed in the responding cells. We have found that distinct LIM homeodomain transcription factors, encoded by the LIM homeobox (LIM-HD) genes Lhx2, Lhx9 and Lmx1b integrate the signaling events that link limb patterning and outgrowth along all three axes. Simultaneous loss of Lhx2 and Lhx9 function resulted in patterning and growth defects along the AP and the PD limb axes. Similar, but more severe, phenotypes were observed when the activities of all three factors, Lmx1b, Lhx2 and Lhx9, were significantly reduced by removing their obligatory co-factor Ldb1. This reveals that the dorsal limb-specific factor Lmx1b can partially compensate for the function of Lhx2 and Lhx9 in regulating AP and PD limb patterning and outgrowth. We further showed that Lhx2 and Lhx9 can fully substitute for each other, and that Lmx1b is partially redundant, in controlling the production of output signals in mesenchymal cells in response to Fgf8 and Shh signaling. Our results indicate that several distinct LIM-HD transcription factors in conjunction with their Ldb1 co-factor serve as common central integrators of distinct signaling interactions and feedback loops to coordinate limb patterning and outgrowth along the PD, AP and DV axes after limb bud formation.


Subject(s)
Body Patterning , Extremities/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Cell Proliferation , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Transcription Factors/genetics
9.
Development ; 133(11): 2149-54, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16672341

ABSTRACT

The Dkk family of secreted cysteine-rich proteins regulates Wnt/beta-catenin signaling by interacting with the Wnt co-receptor Lrp5/6. Here, we show that Dkk2-mediated repression of the Wnt/beta-catenin pathway is essential to promote differentiation of the corneal epithelial progenitor cells into a non-keratinizing stratified epithelium. Complete transformation of the corneal epithelium into a stratified epithelium that expresses epidermal-specific differentiation markers and develops appendages such as hair follicles is achieved in the absence of the Dkk2 gene function. We show that Dkk2 is a key regulator of the corneal versus epidermal fate of the ocular surface epithelium.


Subject(s)
Cornea/growth & development , Cornea/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Biomarkers , Cornea/embryology , Epidermis/growth & development , Epidermis/metabolism , Epithelium/embryology , Epithelium/growth & development , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Keratinocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Phenotype
10.
Development ; 130(22): 5471-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14507786

ABSTRACT

The epithelial b variant of Fgfr2 is active in the entire surface ectoderm of the early embryo, and later in the limb ectoderm and AER, where it is required for limb outgrowth. As limb buds do not form in the absence of Fgfr2, we used chimera analysis to investigate the mechanism of action of this receptor in limb development. ES cells homozygous for a loss-of-function mutation of Fgfr2 that carry a beta-galactosidase reporter were aggregated with normal pre-implantation embryos. Chimeras with a high proportion of mutant cells did not form limbs, whereas those with a moderate proportion formed limb buds with a lobular structure and a discontinuous AER. Where present, the AER did not contain mutant cells, although mutant cells did localize to the adjacent surface ectoderm and limb mesenchyme. In the underlying mesenchyme of AER-free areas, cell proliferation was reduced, and transcription of Shh and Msx1 was diminished. En1 expression in the ventral ectoderm was discontinuous and exhibited ectopic dorsal localization, whereas Wnt7a expression was diminished in the dorsal ectoderm but remained confined to that site. En1 and Wnt7a were not expressed in non-chimeric Fgfr2-null mutant embryos, revealing that they are downstream of Fgfr2. In late gestation chimeras, defects presented in all three limb segments as bone duplications, bone loss or ectopic outgrowths. We suggest that Fgfr2 is required for AER differentiation, as well as for En1 and Wnt7a expression. This receptor also mediates signals from the limb mesenchyme to the limb ectoderm throughout limb development, affecting the position and morphogenesis of precursor cells in the dorsal and ventral limb ectoderm, and AER.


Subject(s)
Cell Differentiation/physiology , Ectoderm/metabolism , Extremities/embryology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Chimera/embryology , Mice , Receptor, Fibroblast Growth Factor, Type 2
SELECTION OF CITATIONS
SEARCH DETAIL