Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 132(6)2019 03 28.
Article in English | MEDLINE | ID: mdl-30765465

ABSTRACT

Purified vascular endothelial cell (EC) growth factor receptor-2 (VEGFR2) auto-phosphorylates upon VEGF-A occupation in vitro, arguing that VEGR2 confers its mitotic and viability signaling in and of itself. Herein, we show that, in ECs, VEGFR2 function requires concurrent C3a/C5a receptor (C3ar1/C5ar1) and IL-6 receptor (IL-6R)-gp130 co-signaling. C3ar1/C5ar1 or IL-6R blockade totally abolished VEGFR2 auto-phosphorylation, downstream Src, ERK, AKT, mTOR and STAT3 activation, and EC cell cycle entry. VEGF-A augmented production of C3a/C5a/IL-6 and their receptors via a two-step p-Tyk2/p-STAT3 process. Co-immunoprecipitation analyses, confocal microscopy, ligand pulldown and bioluminescence resonance energy transfer assays all indicated that the four receptors are physically interactive. Angiogenesis in murine day 5 retinas and in adult tissues was accelerated when C3ar1/C5ar1 signaling was potentiated, but repressed when it was disabled. Thus, C3ar1/C5ar1 and IL-6R-gp130 joint activation is needed to enable physiological VEGFR2 function.


Subject(s)
Cytokine Receptor gp130/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Proliferation , Endothelial Cells/metabolism , Interleukin-6/metabolism , Mice , Neovascularization, Physiologic , Signal Transduction , Vascular Endothelial Growth Factors/metabolism
2.
J Am Heart Assoc ; 7(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29858371

ABSTRACT

BACKGROUND: Limb ischemia resulting from peripheral vascular disease is a common cause of morbidity. Vessel occlusion limits blood flow, creating a hypoxic environment that damages distal tissue, requiring therapeutic revascularization. Hypoxia-inducible factors (HIFs) are key transcriptional regulators of hypoxic vascular responses, including angiogenesis and arteriogenesis. Despite vascular smooth muscle cells' (VSMCs') importance in vessel integrity, little is known about their functional responses to hypoxia in peripheral vascular disease. This study investigated the role of VSMC HIF in mediating peripheral ischemic responses. METHODS AND RESULTS: We used ArntSMKO mice with smooth muscle-specific deletion of aryl hydrocarbon receptor nuclear translocator (ARNT, HIF-1ß), required for HIF transcriptional activity, in a femoral artery ligation model of peripheral vascular disease. ArntSMKO mice exhibit impaired perfusion recovery despite normal collateral vessel dilation and angiogenic capillary responses. Decreased blood flow manifests in extensive tissue damage and hypoxia in ligated limbs of ArntSMKO mice. Furthermore, loss of aryl hydrocarbon receptor nuclear translocator changes the proliferation, migration, and transcriptional profile of cultured VSMCs. ArntSMKO mice display disrupted VSMC morphologic features and wrapping around arterioles and increased vascular permeability linked to decreased local blood flow. CONCLUSIONS: Our data demonstrate that traditional vascular remodeling responses are insufficient to provide robust peripheral tissue reperfusion in ArntSMKO mice. In all, this study highlights HIF responses to hypoxia in arteriole VSMCs critical for the phenotypic and functional stability of vessels that aid in the recovery of blood flow in ischemic peripheral tissues.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Gene Expression Regulation , Ischemia/genetics , Lower Extremity/blood supply , Muscle, Smooth, Vascular/metabolism , Peripheral Vascular Diseases/genetics , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/biosynthesis , Blotting, Western , Cells, Cultured , Disease Models, Animal , Immunohistochemistry , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Muscle, Smooth, Vascular/pathology , Peripheral Vascular Diseases/metabolism , Peripheral Vascular Diseases/pathology , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
Mol Cell Biol ; 38(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29203644

ABSTRACT

Macrophages are strategically distributed in mammalian tissues and play an essential role in priming the immune response. However, macrophages need to constantly strike a balance between activation and inhibition states to avoid a futile inflammatory reaction. Here, we identify the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as a potent repressor of macrophage proinflammatory activation. Gain- and loss-of-function studies revealed that CITED2 is required for optimal peroxisome proliferator-activated receptor gamma (PPARγ) activation and attendant select anti-inflammatory gene expression in macrophages. More importantly, deficiency of CITED2 resulted in significant attenuation of rosiglitazone-induced PPARγ activity, PPARγ recruitment to target gene promoters, and anti-inflammatory target gene expression in macrophages. Interestingly, deficiency of Cited2 strikingly heightened proinflammatory gene expression through stabilization of hypoxia-inducible factor 1 alpha (HIF1α) protein in macrophages. Further, overexpression of Egln3 or inhibition of HIF1α in Cited2-deficient macrophages completely reversed elevated proinflammatory cytokine/chemokine gene expression. Importantly, mice bearing a myeloid cell-specific deletion of Cited2 were highly susceptible to endotoxin-induced sepsis symptomatology and mortality. Collectively, our observations identify CITED2 as a novel negative regulator of macrophage proinflammatory activation that protects the host from inflammatory insults.


Subject(s)
Macrophage Activation/physiology , Macrophages/immunology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apoptosis Regulatory Proteins , Cells, Cultured , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , PPAR gamma/metabolism , RAW 264.7 Cells
4.
Vasc Med ; 19(6): 429-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25398385

ABSTRACT

The recruitment and homing of circulating bone marrow-derived cells include endothelial progenitor cells (EPCs) that are critical to neovascularization and tissue regeneration of various vascular pathologies. We report here that conditional inactivation of hypoxia-inducible factor's (HIF) transcriptional activity in the endothelium of adult mice (Arnt(ΔiEC) mice) results in a disturbance of infiltrating cells, a hallmark of neoangiogenesis, during the early phases of wound healing. Cutaneous biopsy punches show distinct migration of CD31(+) cells into wounds of control mice by 36 hours. However, a significant decline in numbers of infiltrating cells with immature vascular markers, as well as decreased transcript levels of genes associated with their expression and recruitment, were identified in wounds of Arnt(ΔiEC) mice. Matrigel plug assays further confirmed neoangiogenic deficiencies alongside a reduction in numbers of proangiogenic progenitor cells from bone marrow and peripheral blood samples of recombinant vascular endothelial growth factor-treated Arnt(ΔiEC) mice. In addition to HIF's autocrine requirements in endothelial cells, our data implicate that extrinsic microenvironmental cues provided by endothelial HIF are pivotal for early migration of proangiogenic cells, including those involved in wound healing.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Wound Healing/physiology , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/physiology , Skin/metabolism , Skin/pathology , Vascular Endothelial Growth Factor A/metabolism
5.
J Biol Chem ; 289(17): 12016-12028, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24599951

ABSTRACT

Regulation of endothelial cell biology by the Notch signaling pathway (Notch) is essential to vascular development, homeostasis, and sprouting angiogenesis. Although Notch determines cell fate and differentiation in a wide variety of cells, the molecular basis of upstream regulation of Notch remains poorly understood. Our group and others have implicated the Krüppel-like factor family of transcription factors as critical regulators of endothelial function. Here, we show that Krüppel-like factor 4 (KLF4) is a central regulator of sprouting angiogenesis via regulating Notch. Using a murine model in which KLF4 is overexpressed exclusively in the endothelium, we found that sustained expression of KLF4 promotes ineffective angiogenesis leading to diminished tumor growth independent of endothelial cell proliferation or cell cycling effects. These tumors feature increased vessel density yet are hypoperfused, leading to tumor hypoxia. Mechanistically, we show that KLF4 differentially regulates expression of Notch receptors, ligands, and target genes. We also demonstrate that KLF4 limits cleavage-mediated activation of Notch1. Finally, we rescue Notch target gene expression and the KLF4 sprouting angiogenesis phenotype by supplementation of DLL4 recombinant protein. Identification of this hitherto undiscovered role of KLF4 implicates this transcription factor as a critical regulator of Notch, tumor angiogenesis, and sprouting angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , Kruppel-Like Transcription Factors/physiology , Neovascularization, Pathologic , Receptors, Notch/metabolism , Signal Transduction , Animals , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Gene Knockdown Techniques , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Transgenic
6.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24265312

ABSTRACT

CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.


Subject(s)
Embryonic Stem Cells/metabolism , Glycolysis/physiology , Mitochondria/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic/physiology , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Hypoxia/physiology , Embryonic Stem Cells/cytology , Glucose/genetics , Glucose/metabolism , Hexokinase/biosynthesis , Hexokinase/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidation-Reduction , Repressor Proteins/genetics , Trans-Activators/genetics
7.
J Biol Chem ; 287(34): 29088-100, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22761414

ABSTRACT

Cited2 (CBP/p300-interacting transactivator with glutamic acid (E)/aspartic acid (D)-rich tail 2) is a transcriptional modulator critical for the development of multiple organs. Although many Cited2-mediated phenotypes and molecular events have been well characterized using in vivo genetic murine models, Cited2-directed cell fate decision in embryonic stem cells (ESCs) remains elusive. In this study, we examined the role of Cited2 in the maintenance of stemness and pluripotency of murine ESCs by a gene-targeting approach. Cited2 knock-out (Cited2(Δ/-), KO) ESCs display defective differentiation. Loss of Cited2 in differentiating ESCs results in delayed silencing of the genes involved in the maintenance of pluripotency and self-renewal of stem cells (Oct4, Klf4, Sox2, and c-Myc) and the disturbance in cardiomyocyte, hematopoietic, and neuronal differentiation. In addition, Cited2 KO ESCs experience a delayed induction of cardiomyocyte differentiation-associated proteins, NFAT3 (along with the reduced expression of NFAT3 target genes, Nkx2.5 and ß-MHC), N-cadherin, and smooth muscle actin. CITED2 is recruited to the Oct4 promoter to regulate its expression during early ESC differentiation. This is the first demonstration that Cited2 controls ESC pluripotency and differentiation via direct regulation of Oct4 gene expression.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Cadherins/biosynthesis , Cadherins/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Mice , Myocytes, Cardiac/cytology , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/genetics , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
8.
Angiogenesis ; 15(3): 409-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22484908

ABSTRACT

Hypoxia inducible factor (HIF) is a master heterodimeric transcriptional regulator of oxygen (O(2)) homeostasis critical to proper angiogenic responses. Due to the distinctive coexpression of HIF-1α and HIF-2α subunits in endothelial cells, our goal was to examine the genetic elimination of HIF transcriptional activity in response to physiological hypoxic conditions by using a genetic model in which the required HIF-ß subunit (ARNT, Aryl hydrocarbon Receptor Nuclear Translocator) to HIF transcriptional responses was depleted. Endothelial cells (ECs) and aortic explants were isolated from Arnt ( loxP/loxP ) mice and infected with Adenovirus-Cre/GFP or control-GFP. We observed that moderate levels of 2.5 % O(2) promoted vessel sprouting, growth, and branching in control aortic ring assays while growth from Adenovirus-Cre infected explants was compromised. Primary Adenovirus-Cre infected EC cultures featured adverse migration and tube formation phenotypes. Primary pulmonary or cardiac ARNT-deleted ECs also failed to proliferate and survive in response to 8 or 2.5 % O(2) and hydrogen peroxide treatment. Our data demonstrates that ARNT promotes EC migration and vessel outgrowth and is indispensible for the proliferation and preservation of ECs in response to the physiological environmental cue of hypoxia. Thus, these results demonstrate that ARNT plays a critical intrinsic role in ECs and support an important collaboration between HIF-1 and HIF-2 transcriptional activity in these cells.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Cell Hypoxia , Endothelial Cells/metabolism , Animals , Aorta/growth & development , Apoptosis , Base Sequence , Cell Proliferation , Cell Survival , Cells, Cultured , DNA Primers , Endothelial Cells/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , In Vitro Techniques , Mice , Neovascularization, Physiologic , Real-Time Polymerase Chain Reaction
9.
Stem Cells ; 28(4): 799-809, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20135683

ABSTRACT

Adaptive responses to low oxygen (O(2)) tension (hypoxia) are mediated by the heterodimeric transcription factor hypoxia inducible factor (HIF). When stabilized by hypoxia, bHLH-PAS alpha- and beta- (HIF-1beta or ARNT) HIF complex regulate the expression of multiple genes, including vascular endothelial growth factor (VEGF). To investigate the mechanism(s) through which hypoxia contributes to blood vessel development, we used embryonic stem cell (ESC) differentiation cultures that develop into embryoid bodies (EBs) mimicking early embryonic development. Significantly, low O(2) levels promote vascular development and maturation in wild-type (WT) ESC cultures measured by an increase in the numbers of CD31(+) endothelial cells (ECs) and sprouting angiogenic EBs, but refractory in Arnt(-/-) and Vegf(-/-) ESC cultures. Thus, we propose that hypoxia promotes the production of ECs and contributes to the development and maturation of vessels. Our findings further demonstrate that hypoxia alters the temporal expression of VEGF receptors Flk-1 (VEGFR-2) and the membrane and soluble forms of the antagonistic receptor Flt-1 (VEGFR-1). Moreover, these receptors are distinctly expressed in differentiating Arnt(-/-) and Vegf(-/-) EBs. These results support existing models in which VEGF signaling is tightly regulated during specific biologic events, but also provide important novel evidence that, in response to physiologic hypoxia, HIF mediates a distinct stoichiometric pattern of VEGF receptors throughout EB differentiation analogous to the formation of vascular networks during embryogenesis.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neovascularization, Physiologic , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/deficiency , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Mice , Mice, Knockout , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism
10.
Dev Cell ; 11(1): 81-92, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16824955

ABSTRACT

Hypoxia inducible factors (HIFs) regulate adaptive responses to changes in oxygen (O(2)) tension during embryogenesis, tissue ischemia, and tumorigenesis. Because HIF-deficient embryos exhibit a number of developmental defects, the precise role of HIF in early vascular morphogenesis has been uncertain. Using para-aortic splanchnopleural (P-Sp) explant cultures, we show that deletion of the HIF-beta subunit (ARNT) results in defective hematopoiesis and the inhibition of both vasculogenesis and angiogenesis. These defects are rescued upon the addition of wild-type Sca-1(+) hematopoietic cells or recombinant VEGF. Arnt(-/-) embryos exhibit reduced levels of VEGF protein and increased numbers of apoptotic hematopoietic cells. These results suggest that HIF coordinates early endothelial cell emergence and vessel development by promoting hematopoietic cell survival and paracrine growth factor production.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Blood Vessels/embryology , Hematopoietic Cell Growth Factors/physiology , Animals , Apoptosis , Aryl Hydrocarbon Receptor Nuclear Translocator/deficiency , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Base Sequence , Bone Marrow Cells/physiology , Coculture Techniques , DNA/genetics , Embryonic Development/drug effects , Embryonic Development/physiology , Female , Hematopoiesis , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Mice, Knockout , Neovascularization, Physiologic , Pregnancy , Recombinant Proteins/pharmacology , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
11.
Development ; 131(18): 4623-34, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15342485

ABSTRACT

Hypoxia Inducible Factor (HIF), consisting of HIF1alpha and ARNT (HIF1beta) subunits, activates multiple genes in response to oxygen (O(2)) deprivation. Arnt(-/-) mice exhibit substantial defects in blood cell and vessel development. We demonstrate that hypoxia accelerates the expression of Brachyury (a mesoderm-specific transcription factor), BMP4 (a mesoderm-promoting growth factor) and FLK1 (a marker of hemangioblasts, the bipotential progenitor of endothelial and hematopoietic cells) in differentiating ES cell cultures. Significantly, proliferation of embryonic hemangioblasts (BL-CFCs) is regulated by hypoxia, as Arnt(+/+) ES cells generate increased numbers of FLK1(+) cells, and BL-CFCs with accelerated kinetics in response to low O(2). This response is HIF-dependent as Arnt(-/-) ES cells produce fewer FLK1(+) cells and BL-CFCs, under both normoxic and hypoxic conditions. Interestingly, this defect is rescued when Arnt(-/-) ES cells are co-cultured with Arnt(+/+) ES cells. Vegf(+/-)or Vegf(-/-) ES cells generate proper numbers of FLK1(+) cells but fewer BL-CFCs, suggesting that additional factors regulated by HIF (other than VEGF) are involved in these early events. Thus, hypoxic responses are important for the establishment of various progenitor cells, including early mesoderm and its differentiation into hemangioblasts. Together these data suggest that ineffective responses to hypoxia in Arnt(-/-) embryos abrogate proper cardiovascular development during early embryogenesis, including the pathways controlling hemangioblast differentiation.


Subject(s)
Cell Differentiation , Hypoxia/physiopathology , Mesoderm/cytology , Mesoderm/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Cell Count , Cell Division , Cells, Cultured , Coculture Techniques , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fetal Proteins/genetics , Gene Deletion , Growth Substances/pharmacology , Hypoxia/genetics , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Kinetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Stem Cells/cytology , Stem Cells/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
EMBO J ; 21(9): 2220-30, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980719

ABSTRACT

Mature macrophages, neutrophils and lymphoid cells do not develop in PU.1(-/-) mice. In contrast, mice lacking the highly related protein Spi-B generate all hematopoietic lineages but display a B-cell receptor signaling defect. These distinct phenotypes could result from functional differences between PU.1 and Spi-B or their unique temporal and tissue-specific expression (PU.1: myeloid and B cells; Spi-B: B cells only). To address this question, we introduced the Spi-B cDNA into the murine PU.1 locus by homologous recombination. In the absence of PU.1, Spi-B rescued macrophage and granulocyte development when assayed by in vitro differentiation of embryonic stem cells. Adherent, CD11b(+)/F4/80(+) cells capable of phagocytosis were detected in PU.1(Spi-B/Spi-B) embryoid bodies, and myeloid colonies were present in hematopoietic progenitor assays. Despite its ability to rescue myeloid differentiation, Spi-B did not rescue lymphoid development in a RAG-2(-/-) complementation assay. These results demonstrate an important difference between PU.1 and Spi-B. Careful comparison of these Ets factors will delineate important functional domains of PU.1 involved in lymphocyte lineage commitment and/or maturation.


Subject(s)
Cell Lineage/physiology , DNA-Binding Proteins/physiology , Lymphocytes/physiology , Myeloid Cells/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Cell Lineage/genetics , Cells, Cultured , Gene Targeting , Macrophages/physiology , Mice , Proto-Oncogene Proteins c-ets , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...