Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Dev Dyn ; 250(8): 1074-1095, 2021 08.
Article in English | MEDLINE | ID: mdl-33522658

ABSTRACT

BACKGROUND: Cardio-facio-cutaneous (CFC) syndrome is a human multiple congenital anomaly syndrome that is caused by activating heterozygous mutations in either BRAF, MEK1, or MEK2, three protein kinases of the Ras/mitogen-activated protein kinase (MAPK) pathway. CFC belongs to a group of syndromes known as RASopathies. Skeletal muscle hypotonia is a ubiquitous phenotype of RASopathies, especially in CFC syndrome. To better understand the underlying mechanisms for the skeletal myopathy in CFC, a mouse model with an activating BrafL597V allele was utilized. RESULTS: The activating BrafL597V allele resulted in phenotypic alterations in skeletal muscle characterized by a reduction in fiber size which leads to a reduction in muscle size which are functionally weaker. MAPK pathway activation caused inhibition of myofiber differentiation during embryonic myogenesis and global transcriptional dysregulation of developmental pathways. Inhibition in differentiation can be rescued by MEK inhibition. CONCLUSIONS: A skeletal myopathy was identified in the CFC BrafL597V mouse validating the use of models to study the effect of Ras/MAPK dysregulation on skeletal myogenesis. RASopathies present a novel opportunity to identify new paradigms of myogenesis and further our understanding of Ras in development. Rescue of the phenotype by inhibitors may help advance the development of therapeutic options for RASopathy patients.


Subject(s)
Ectodermal Dysplasia/genetics , Failure to Thrive/genetics , Heart Defects, Congenital/genetics , Mitogen-Activated Protein Kinases/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Proto-Oncogene Proteins B-raf/genetics , Alleles , Animals , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Facies , Failure to Thrive/metabolism , Failure to Thrive/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Phenotype , Proto-Oncogene Proteins B-raf/metabolism
2.
Oncotarget ; 6(28): 26278-90, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26172302

ABSTRACT

Colorectal cancer (CRC) is the third most prevalent cancer type worldwide with a mortality rate of approximately 50%. Elevated cell-surface expression of truncated carbohydrate structures such as Tn antigen (GalNAcα-Ser/Thr) is frequently observed during tumor progression. We have previously demonstrated that the C-type lectin macrophage galactose-type lectin (MGL), expressed by human antigen presenting cells, can distinguish healthy tissue from CRC through its specific recognition of Tn antigen. Both MGL binding and oncogenic BRAF mutations have been implicated in establishing an immunosuppressive microenvironment. Here we aimed to evaluate whether MGL ligand expression has prognostic value and whether this was correlated to BRAF(V600E) mutation status. Using a cohort of 386 colon cancer patients we demonstrate that high MGL binding to stage III tumors is associated with poor disease-free survival, independent of microsatellite instability or adjuvant chemotherapy. In vitro studies using CRC cell lines showed an association between MGL ligand expression and the presence of BRAF(V600E). Administration of specific BRAF(V600E) inhibitors resulted in decreased expression of MGL-binding glycans. Moreover, a positive correlation between induction of BRAF(V600E) and MGL binding to epithelial cells of the gastrointestinal tract was found in vivo using an inducible BRAF(V600E) mouse model. We conclude that the BRAF(V600E) mutation induces MGL ligand expression, thereby providing a direct link between oncogenic transformation and aberrant expression of immunosuppressive glycans. The strong prognostic value of MGL ligands in stage III colon cancer patients, i.e. when tumor cells disseminate to lymph nodes, further supports the putative immune evasive role of MGL ligands in metastatic disease.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Lectins, C-Type/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Aged, 80 and over , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Disease-Free Survival , Female , Genetic Predisposition to Disease , HT29 Cells , Humans , Kaplan-Meier Estimate , Ligands , Male , Mice, Transgenic , Middle Aged , Neoplasm Staging , Phenotype , Proportional Hazards Models , Signal Transduction , Time Factors , Treatment Outcome , Tumor Escape , Up-Regulation
3.
Mol Pharmacol ; 86(2): 211-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830940

ABSTRACT

Both human ether-à-go-go-related gene (hERG1) and the closely related human ether-à-go-go (hEAG1) channel are aberrantly expressed in a large proportion of human cancers. In the present study, we demonstrate that transfection of hERG1 into mouse fibroblasts is sufficient to induce many features characteristic of malignant transformation. An important finding of this work is that this transformation could be reversed by chronic incubation (for 2-3 weeks) with the hERG channel blocker dofetilide (100 nM), whereas more acute applications (for 1-2 days) were ineffective. The hERG1 expression resulted in a profound loss of cell contact inhibition, multiple layers of overgrowing cells, and high saturation densities. Cells also changed from fibroblast-like to a more spindle-shaped morphology, which was associated with a smaller cell size, a dramatic increase in cell polarization, a reduction in the number of actin stress fibers, and less punctate labeling of focal adhesions. Analysis of single-cell migration and scratch-wound closure clearly demonstrated that hERG1-expressing cells migrated more rapidly than vector-transfected control cells. In contrast to previous studies on hEAG1, there were no increases in rates of proliferation, or loss of growth factor dependency; however, hERG1-expressing cells were capable of substrate-independent growth. Allogeneic transplantation of hERG1-expressing cells into nude mice resulted in an increased incidence of tumors. In contrast to hEAG1, the mechanism of cellular transformation is dependent on ion conduction. Trafficking-deficient and conduction-deficient hERG1 mutants also prevented cellular transformation. These results provide evidence that hERG1 expression is sufficient to induce cellular transformation by a mechanism distinct from hEAG1. The most important conclusion of this study is that selective hERG1 channel blockers have therapeutic potential in the treatment of hERG1-expressing cancers.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Potassium Channel Blockers/pharmacology , Actins/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Fibroblasts/drug effects , Focal Adhesions/metabolism , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Stress Fibers/metabolism , Transfection
4.
Mol Cancer Res ; 11(12): 1530-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24152792

ABSTRACT

UNLABELLED: Mutational activation of BRAF leading to expression of the BRAF(V600E) oncoprotein was recently identified in a high percentage of specific hematopoietic neoplasms in monocyte/histiocyte and mature B-cell lineages. Although BRAF(V600E) is a driver oncoprotein and pharmacologic target in solid tumors such as melanoma, lung, and thyroid cancer, it remains unknown whether BRAF(V600E) is an appropriate therapeutic target in hematopoietic neoplasms. To address this critical question, we generated a mouse model expressing inducible BRAF(V600E) in the hematopoietic system, and evaluated the efficacy of pathway-targeted therapeutics against primary hematopoietic cells. In this model, BRAF(V600E) expression conferred cytokine-independent growth to monocyte/macrophage-lineage progenitors leading to aberrant in vivo and in vitro monocyte/macrophage expansion. Furthermore, transplantation of BRAF(V600E)-expressing bone marrow cells promoted an in vivo pathology most notable for monocytosis in hematopoietic tissues and visceral organs. In vitro analysis revealed that MAP-ERK kinase inhibition, but not RAF inhibition, effectively suppressed cytokine-independent clonal growth of monocyte/macrophage-lineage progenitors. However, combined RAF and phosphoinositide 3-kinase (PI3K) inhibition effectively inhibited cytokine-independent colony formation, suggesting autocrine PI3K pathway activation. Taken together, these results provide evidence that constitutively activated BRAF(V600E) drives aberrant proliferation of monocyte-lineage cells. IMPLICATIONS: This study supports the development of pathway-targeted therapeutics in the treatment of BRAF(V600E)-expressing hematopoietic neoplasms in the monocyte/histiocyte lineage.


Subject(s)
Drug Resistance, Neoplasm , Indoles/pharmacology , Monocyte-Macrophage Precursor Cells/physiology , Monocytes/physiology , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/pharmacology , raf Kinases/antagonists & inhibitors , Animals , Benzamides/pharmacology , Bone Marrow Transplantation , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Erythropoiesis , Furans/pharmacology , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocyte-Macrophage Precursor Cells/drug effects , Monocytes/drug effects , Myelopoiesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction
5.
Biochem Biophys Res Commun ; 426(2): 190-5, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22925892

ABSTRACT

Mice homozygous for several Tln2 gene targeted alleles are viable and fertile. Here we show that although the expression of talin2 protein is drastically reduced in muscle from these mice, other tissues continue to express talin2 albeit at reduced levels. We therefore generated a Tln2 allele lacking the entire coding sequence (Tln2(cd)). Tln2(cd/cd) mice were viable and fertile, and the genotypes of Tln2(cd/+) intercrosses were at the expected Mendelian ratio. Tln2(cd/cd) mice showed no major difference in body mass or the weight of the major organs compared to wild-type, although they displayed a mildly dystrophic phenotype. Moreover, Tln2(cd/cd) mouse embryo fibroblasts showed no obvious defects in cell adhesion, migration or proliferation. However, the number of Tln2(cd/cd) pups surviving to adulthood was variable suggesting that such mice have an underlying defect.


Subject(s)
Embryonic Development/genetics , Fertility , Talin/physiology , Animals , Body Weight , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Fibroblasts/physiology , Gene Deletion , Male , Mice , Mice, Knockout , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Talin/genetics
6.
Development ; 139(3): 465-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190642

ABSTRACT

Knock out of intestinal Cdx2 produces different effects depending upon the developmental stage at which this occurs. Early in development it produces histologically ordered stomach mucosa in the midgut. Conditional inactivation of Cdx2 in adult intestinal epithelium, as well as specifically in the Lgr5-positive stem cells, of adult mice allows long-term survival of the animals but fails to produce this phenotype. Instead, the endodermal cells exhibit cell-autonomous expression of gastric genes in an intestinal setting that is not accompanied by mesodermal expression of Barx1, which is necessary for gastric morphogenesis. Cdx2-negative endodermal cells also fail to express Sox2, a marker of gastric morphogenesis. Maturation of the stem cell niche thus appears to be associated with loss of ability to express positional information cues that are required for normal stomach development. Cdx2-negative intestinal crypts produce subsurface cystic vesicles, whereas untargeted crypts hypertrophy to later replace the surface epithelium. These observations are supported by studies involving inactivation of Cdx2 in intestinal crypts cultured in vitro. This abolishes their ability to form long-term growing intestinal organoids that differentiate into intestinal phenotypes. We conclude that expression of Cdx2 is essential for differentiation of gut stem cells into any of the intestinal cell types, but they maintain a degree of cell-autonomous plasticity that allows them to switch on a variety of gastric genes.


Subject(s)
Endoderm/growth & development , Intestinal Mucosa/growth & development , Intestine, Small/growth & development , Animals , CDX2 Transcription Factor , Cell Differentiation/genetics , Cells, Cultured , Female , Gastric Mucosa/growth & development , Gene Knockout Techniques , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis/genetics , SOXB1 Transcription Factors/biosynthesis , Stem Cells/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Dev Biol ; 349(2): 494-502, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21081121

ABSTRACT

Using Tln1(fl/fl);CreER mice, we show that tamoxifen-induced inactivation of the talin1 gene throughout the embryo produces an angiogenesis phenotype that is restricted to newly forming blood vessels. The phenotype has a rapid onset in early embryos, resulting in vessel defects by 48 h and death of the embryo within 72 h. Very similar vascular defects were obtained using a Tie2-Cre endothelial cell-specific Tln1 knockout, a phenotype that was rescued by expression of a Tln1 mini-gene in endothelial cells. We show that endothelial cells, unlike most other cell types, do not express talin2, which can compensate for loss of talin1, and demonstrate for the first time that endothelial cells in vivo lacking talin1 are unable to undergo the cell spreading and flattening required to form vessels.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Talin/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Embryo, Mammalian , Endothelial Cells/physiology , Gene Silencing/drug effects , Histological Techniques , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron , Talin/genetics , Tamoxifen
8.
EMBO Mol Med ; 2(11): 458-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20941790

ABSTRACT

The majority of human colorectal cancers (CRCs) are initiated by mutations arising in the adenomatous polyposis coli (APC) tumour suppressor gene. However, a new class of non-APC mutated CRCs has been defined that have a serrated histopathology and carry the (V600E)BRAF oncogene. Here we have investigated the pathogenesis of serrated CRCs by expressing (V600E)Braf in the proliferative cells of the mouse gastrointestinal tract. We show that the oncogene drives an initial burst of Mek-dependent proliferation, leading to the formation of hyperplastic crypts. This is associated with ß-catenin nuclear localization by a mechanism involving Mapk/Erk kinase (Mek)-dependent, Akt-independent phosphorylation of Gsk3ß. However, hyperplastic crypts remain dormant for prolonged periods due to the induction of crypt senescence accompanied by upregulation of senescence-associated ß-galactosidase and p16(Ink4a). We show that tumour progression is associated with down-regulation of p16(Ink4a) through enhanced CpG methylation of exon 1 and knockout of Cdkn2a confirms this gene is a barrier to tumour progression. Our studies identify (V600E)BRAF as an early genetic driver mutation in serrated CRCs and indicate that, unlike APC-mutated cancers, this subtype arises by the bypassing of a (V600E)Braf driven oncogene-induced senescence programme.


Subject(s)
Aging , Colorectal Neoplasms/physiopathology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Gastrointestinal Tract/physiopathology , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Amino Acid Substitution/genetics , Animals , Cell Nucleus/chemistry , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Gastrointestinal Tract/pathology , Gene Expression Profiling , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hyperplasia/pathology , Mice , Up-Regulation , beta Catenin/metabolism
9.
Eur J Cell Biol ; 89(9): 661-73, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605055

ABSTRACT

Talin binds to and activates integrins and is thought to couple them to cytoskeletal actin. However, functional studies on talin have been restricted by the fact that most cells express two talin isoforms. Here we show that human umbilical vein endothelial cells (HUVEC) express only talin1, and that talin1 knockdown inhibited focal adhesion (FA) assembly preventing the cells from maintaining a spread morphology, a phenotype that was rescued by GFP-mouse talin1. Thus HUVEC offer an ideal model system in which to conduct talin structure/function studies. Talin contains an N-terminal FERM domain (comprised of F1, F2 and F3 domains) and a C-terminal flexible rod. The F3 FERM domain binds beta-integrin tails, and mutations in F3 that inhibited integrin binding (W359A) or activation (L325R) severely compromised the ability of GFP-talin1 to rescue the talin1 knockdown phenotype despite the presence of a second integrin-binding site in the talin rod. The talin rod contains several actin-binding sites (ABS), and mutations in the C-terminal ABS that reduced actin-binding impaired talin1 function, whereas those that increased binding resulted in more stable FAs. The results show that both the N-terminal integrin and C-terminal actin-binding functions of talin are essential to cell spreading and FA assembly. Finally, mutations that relieve talin auto-inhibition resulted in the rapid and excessive production of FA, highlighting the importance of talin regulation within the cell.


Subject(s)
Endothelial Cells/metabolism , Integrins/metabolism , Talin/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Focal Adhesions/physiology , Gene Knockdown Techniques , Humans , Integrins/chemistry , Integrins/genetics , Mice , Phenotype , Talin/chemistry , Talin/genetics , Transfection , Umbilical Veins/cytology , Up-Regulation
10.
FEBS J ; 276(6): 1610-28, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19220457

ABSTRACT

Talins are large adaptor proteins that link the integrin family of adhesion molecules to F-actin. In vertebrates, there are two talin genes. Talin 1 is essential for integrin-mediated cell adhesion; the role of talin 2 is unclear. Here we report a detailed analysis of mammalian talin 2. This reveals the existence of a previously unrecognized promoter associated with a CpG island, and separated from the first coding exon by numerous alternatively spliced noncoding exons spanning > 200 kb. Analysis of a mouse gene trap line shows that this promoter accounts for most of the talin 2 expression in adult tissues. We also demonstrate that testis and kidney express truncated talin 2 isoforms that lack the N-terminal half of the protein, and provide evidence for the developmentally regulated expression of the short testis-specific talin 2 isoform in elongating spermatids. Finally, we identify four tissue-specific alternative splicing events within the coding region of talin 2.


Subject(s)
RNA, Messenger/genetics , Talin/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , CpG Islands , DNA , Exons , Gene Knockdown Techniques , Humans , Kidney/metabolism , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Spermatids/metabolism , Talin/chemistry , Talin/genetics , Talin/metabolism
11.
Mol Cell ; 31(6): 862-72, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18922468

ABSTRACT

The CRAF protein kinase regulates proliferative, differentiation, and survival signals from activated RAS proteins to downstream effectors, most often by inducing MEK/ERK activation. A well-established model of CRAF regulation involves RAS-mediated translocation of CRAF to the plasma membrane, where it is activated by a series of events including phosphorylation. Here we have discovered a new mode of regulation that occurs prior to this step. By creating a kinase-defective version of CRAF in mice or by use of the RAF inhibitor sorafenib, we show that CRAF must first undergo autophosphorylation of serine 621 (S621). Autophosphorylation occurs in cis, does not involve MEK/ERK activation, and is essential to ensure the correct folding and stability of the protein. In the absence of S621 phosphorylation, CRAF is degraded by the proteasome by mechanisms that do not uniquely rely on the E3 ubiquitin ligase CHIP.


Subject(s)
Phosphoserine/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-raf/metabolism , Animals , Enzyme Activation , Enzyme Stability , Fibroblasts/enzymology , Mice , Models, Biological , Phenotype , Phosphorylation , Protein Folding , Ubiquitination
12.
FEBS Lett ; 582(17): 2555-60, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18577384

ABSTRACT

Null mutation or haploinsufficiency of Cdx2 results in the development of heterotopic lesions with a gastric phenotype in the midgut endoderm. Conversely transgenic expression of Cdx2 in the stomach causes the endoderm to differentiate into intestinal-type mucosa. We demonstrate that the mesoderm adjacent to intestinal heterotopic areas expresses stomach specific Barx1 while the surrounding mesoderm is Barx1 negative. We conclude that the initiation of gut histodifferentiation lies in the endodermal expression of Cdx2 and that endodermal/mesodermal cross-talk involving Barx1 with appropriate feedback loops results in the development of the postnatal gut phenotype.


Subject(s)
Endoderm/metabolism , Homeodomain Proteins/metabolism , Intestines/embryology , Morphogenesis , Stomach/embryology , Transcription Factors/metabolism , Animals , CDX2 Transcription Factor , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Female , Gastric Mucosa/metabolism , HMGB Proteins/metabolism , Homeodomain Proteins/genetics , Intestinal Mucosa/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Mutant Strains , Mice, Transgenic , Morphogenesis/genetics , SOXB1 Transcription Factors , Transcription Factors/genetics
13.
PLoS One ; 3(4): e1929, 2008 Apr 16.
Article in English | MEDLINE | ID: mdl-18414652

ABSTRACT

AIMS: Cultured cardiac explants produce a heterogeneous population of cells including a distinctive population of refractile cells described here as small round cardiac explant derived cells (EDCs). The aim of this study was to explore the source, morphology and cardiogenic potential of EDCs. METHODS: Transgenic MLC2v-Cre/ZEG, and actin-eGFP mice were used for lineage-tracing of EDCs in vitro and in vivo. C57B16 mice were used as cell transplant recipients of EDCs from transgenic hearts, as well as for the general characterisation of EDCs. The activation of cardiac-specific markers were analysed by: immunohistochemistry with bright field and immunofluorescent microscopy, electron microscopy, PCR and RT-PCR. Functional engraftment of transplanted cells was further investigated with calcium transient studies. RESULTS: Production of EDCs was highly dependent on the retention of blood-derived cells or factors in the cultured explants. These cells shared some characteristics of cardiac myocytes in vitro and survived engraftment in the adult heart in vivo. However, EDCs failed to differentiate into functional cardiac myocytes in vivo as demonstrated by the absence of stimulation-evoked intracellular calcium transients following transplantation into the peri-infarct zone. CONCLUSIONS: This study highlights that positive identification based upon one parameter alone such as morphology or immunofluorescene is not adequate to identify the source, fate and function of adult cardiac explant derived cells.


Subject(s)
Heart/anatomy & histology , Myocardium/cytology , Myocytes, Cardiac/cytology , Animals , Calcium/metabolism , Cell Lineage , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Perfusion , Polymerase Chain Reaction , Transgenes
14.
Blood ; 106(3): 833-40, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15784729

ABSTRACT

Raf kinases play an integral role in the classic mitogen-activated protein (MAP) kinase (Raf/MEK/extracellular signal-related kinase [ERK]) intracellular signaling cascade, but their role in specific developmental processes is largely unknown. Using a genetic approach, we have identified a role for B-Raf during hematopoietic progenitor cell development and during megakaryocytopoiesis. Fetal liver and in vitro embryonic stem (ES) cell-derived myeloid progenitor development is quantitatively impaired in the absence of B-Raf. Biochemical data suggest that this phenotype is due to the loss of a normally occurring rise in B-Raf expression and associated ERK1/2 activation during hematopoietic progenitor cell formation. However, the presence of B-raf-/- ES cell-derived myeloid progenitors in the bone marrow of adult chimeric mice indicates the lack of an obligate cell-autonomous requirement for B-Raf in myeloid progenitor development. The lack of B-Raf also impairs megakaryocytopoiesis. Thrombopoietin (Tpo)-induced in vitro expansion of ES cell-derived megakaryocyte-lineage cells fails to occur in the absence of B-Raf. Moreover, this quantitative in vitro defect in megakaryocyte-lineage expansion is mirrored by chimeric mice data that show reduced B-raf-/- genotype contribution in megakaryocytes relative to its contribution in myeloid progenitors. Together, these data suggest that B-Raf plays a cell-autonomous role in megakaryocytopoiesis and a permissive role in myeloid progenitor development.


Subject(s)
Myelopoiesis , Proto-Oncogene Proteins B-raf/physiology , Animals , Cell Lineage , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , MAP Kinase Signaling System , Mice , Mice, Knockout , Proto-Oncogene Proteins B-raf/deficiency , Proto-Oncogene Proteins B-raf/genetics , Stem Cells , Thrombopoiesis
15.
Cancer Cell ; 7(3): 219-26, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15766660

ABSTRACT

To investigate the role of signaling by the small GTPase Ral, we have generated mice deficient for RalGDS, a guanine nucleotide exchange factor that activates Ral. We show that RalGDS is dispensable for mouse development but plays a substantial role in Ras-induced oncogenesis. Lack of RalGDS results in reduced tumor incidence, size, and progression to malignancy in multistage skin carcinogenesis, and reduced transformation by Ras in tissue culture. RalGDS does not appear to participate in the regulation of cell proliferation, but instead controls survival of transformed cells. Experiments performed in cells isolated from skin tumors suggest that RalGDS mediates cell survival through the activation of the JNK/SAPK pathway. These studies identify RalGDS as a key component in Ras-dependent carcinogenesis in vivo.


Subject(s)
Cell Transformation, Neoplastic , Signal Transduction/physiology , Skin Neoplasms , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Disease Progression , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , ral GTP-Binding Proteins/genetics , ral Guanine Nucleotide Exchange Factor/genetics , ras Proteins/metabolism
16.
Mol Cell Biol ; 24(13): 5937-52, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199148

ABSTRACT

Recent data have shown that the BRAF gene is mutated at a high frequency in human malignancies. We have analyzed the migratory characteristics of B-raf(-/-) mouse embryonic fibroblasts (MEFs) and compared these with the organization of the actin cytoskeleton and the activity of signaling pathways that are known to influence this organization. Disruption of B-raf significantly reduced the levels of phospho-ERK1/2 and, surprisingly, induced an approximately 1.5-fold increase in cell migration. Consistent with these findings, the high level of actin stress fibers normally present in MEFs was considerably reduced following disruption of B-raf, and the F-actin content of B-raf(-/-) cells was less than half that of B-raf(+/+) cells. Phosphorylation of the myosin light chain on Thr18/Ser19 residues was not reduced in B-raf(-/-) cells. Rather, reduced ROCKII expression and attenuated phosphorylation of ADF/cofilin on serine 3 occurred. Normal stress fiber and phosphocofilin levels were restored by the expression of human B-Raf and catalytically active MEK and by the overexpression of LIM kinase (LIMK). These results have important implications for the role of the B-Raf/ERK signaling pathway in regulating cell motility in normal and malignant cells. They suggest that B-Raf is involved in invasiveness by regulating the proper assembly of actin stress fibers and contractility through a ROCKII/LIMK/cofilin signaling pathway.


Subject(s)
Fibroblasts/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/physiology , Signal Transduction , Stress Fibers/ultrastructure , Actin Depolymerizing Factors , Actins , Animals , Cell Line , Cell Movement , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Fibroblasts/cytology , Humans , Intracellular Signaling Peptides and Proteins , Lim Kinases , Mice , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf/genetics , Stress Fibers/physiology , Transfection , rho-Associated Kinases
17.
Blood ; 103(7): 2568-70, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14576068

ABSTRACT

Thrombopoietin stimulates extracellular signal-related kinase 1/2 (ERK1/2) phosphorylation in megakaryocytes, and the classic mitogen-activated protein (MAP) kinase (Raf/mitogen-induced extracellular kinase [MEK]/ERK) pathway has been implicated directly and indirectly to play a critical role in megakaryocytopoiesis. However, the involvement of specific Raf family members in megakaryocytopoiesis is unknown. raf-1(-/-) mice were therefore used to directly determine the role of Raf-1 in megakaryocytopoiesis. Surprisingly, raf-1(-/-) mice have a modestly higher platelet count than their raf-1(+/+) littermates. Nonetheless, the absence of Raf-1 does not alter thrombopoietin-induced expansion of primary megakaryocyte-lineage cells, the development of apoptotic megakaryocytes in the presence or absence of thrombopoietin, or the development of megakaryocyte DNA ploidy distribution. Moreover, raf-1(-/-) megakaryocytes do not have a compensatory increase in A-Raf or B-Raf expression, and thrombopoietin-induced ERK1/2 phosphorylation is similar in raf-1(-/-) and raf-1(+/+) megakaryocytes. These unexpected findings demonstrate that Raf-1 is dispensable for megakaryocytopoiesis, and for thrombopoietin-induced ERK1/2 activation in primary megakaryocyte-lineage cells.


Subject(s)
Erythropoiesis/physiology , Megakaryocytes/cytology , Megakaryocytes/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/physiology , Thrombopoietin/pharmacology , Animals , Crosses, Genetic , Megakaryocytes/drug effects , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Proto-Oncogene Proteins c-raf/deficiency , Proto-Oncogene Proteins c-raf/genetics , Thrombocytopenia/genetics
18.
Biochim Biophys Acta ; 1653(1): 25-40, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12781369

ABSTRACT

A recent report has shown that activating mutations in the BRAF gene are present in a large percentage of human malignant melanomas and in a proportion of colon cancers. The vast majority of these mutations represent a single nucleotide change of T-A at nucleotide 1796 resulting in a valine to glutamic acid change at residue 599 within the activation segment of B-Raf. This exciting new discovery is the first time that a direct association between any RAF gene and human cancer has been reported. Raf proteins are also indirectly associated with cancer as effectors of activated Ras proteins, oncogenic forms of which are present in approximately one-third of all human cancers. BRAF and RAS mutations are rarely both present in the same cancers but the cancer types with BRAF mutations are similar to those with RAS mutations. This has been taken as evidence that the inappropriate regulation of the downstream ERKs (the p42/p44 MAP kinases) is a major contributing factor in the development of these cancers. Recent studies in mice with targeted mutations of the raf genes have confirmed that B-Raf is a far stronger activator of ERKs than its better studied Raf-1 homologue, even in cell types in which the protein is barely expressed. The explanation for this lies in a number of key differences in the regulation of B-Raf and Raf-1 activity. Constitutive phosphorylation of serine 445 of B-Raf leads to this protein having a higher basal kinase activity than Raf-1. Phosphorylation of threonine 598 and serine 601 within the activation loop of B-Raf at the plasma membrane also regulates its activity. The V599E mutation is thought to mimic these phosphorylations, resulting in a protein with high activity, leading to constitutive ERK activation. B-Raf now provides a critical new target to which drugs for treating malignant melanoma can be developed and, with this in mind, it is now important to gain clear insight into the biochemical properties of this relatively little characterised protein.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-raf/genetics , ras Proteins/metabolism , Amino Acid Sequence , Animals , Humans , MAP Kinase Signaling System , Molecular Sequence Data , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf/metabolism , Sequence Homology, Amino Acid , Signal Transduction
19.
Cell Growth Differ ; 13(7): 307-13, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12133899

ABSTRACT

Recent studies indicate that kinase suppressor of Ras (KSR)is a scaffold protein for the Ras/Raf/MEK/ERK signaling cascade in mammals. To help determine the in vivo function of KSR, we have examined the tissue-specific distribution of this protein in the embryonic and adult mouse using a rat monoclonal antibody raised against the mouse protein. Western blot analysis indicates that the protein is expressed at highest levels in the adult brain. It is also expressed at low levels in bladder, ovary, testis, and lung, but the protein is not detectable in any other adult tissue. However, reverse transcription-PCR analysis shows that Ksr transcripts are detected in all adult tissues except the liver. A variant containing a differentially spliced exon in the CA4 domain is observed in brain, cerebellum, ovary, and intestine. The protein is also expressed throughout the E6.5 embryo and at high levels in the neuroepithelium of the E10.5 embryo. At this embryonic stage, expression is also detected at lower levels in the limb and tail buds as well as in the myocardium.


Subject(s)
Brain/embryology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental/physiology , Protein Kinases/metabolism , Viscera/embryology , ras Proteins/metabolism , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fetus , Gonads/cytology , Gonads/embryology , Gonads/metabolism , Immunohistochemistry , Lung/cytology , Lung/embryology , Lung/metabolism , Mice , Protein Kinases/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/physiology , Urinary Bladder/cytology , Urinary Bladder/embryology , Urinary Bladder/metabolism , Viscera/cytology , Viscera/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...