Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Clin Exp Allergy ; 47(2): 176-189, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27649066

ABSTRACT

BACKGROUND: Allergic asthma is characterized by inflammation and airway remodelling. Airway remodelling with excessive deposition of extracellular matrix (ECM) and larger smooth muscle mass are correlated with increased airway responsiveness and asthma severity. Calpain is a family of calcium-dependent endopeptidases, which plays an important role in ECM remodelling. However, the role of calpain in airway smooth muscle remodelling remains unknown. OBJECTIVE: To investigate the role of calpain in asthmatic airway remodelling as well as the underlying mechanism. METHODS: The mouse asthma model was made by ovalbumin sensitization and challenge. Calpain conditional knockout mice were studied in the model. Airway smooth muscle cells (ASMCs) were isolated from smooth muscle bundles in airway of rats. Cytokines IL-4, IL-5, TNF-α, and TGF-ß1, and serum from patients with asthma were selected to treated ASMCs. Collagen-I synthesis, cell proliferation, and phosphorylation of Akt in ASMCs were analysed. RESULTS: Inhibition of calpain using calpain knockout mice attenuated airway smooth muscle remodelling in mouse asthma models. Cytokines IL-4, IL-5, TNF-α, and TGF-ß1, and serum from patients with asthma increased collagen-I synthesis, cell proliferation, and phosphorylation of Akt in ASMCs, which were blocked by the calpain inhibitor MDL28170. Moreover, MDL28170 reduced cytokine-induced increases in Rictor protein, which is the most important component of mammalian target of rapamycin complex 2 (mTORC2). Blockage of the mTORC2 signal pathway prevented cytokine-induced phosphorylation of Akt, collagen-I synthesis, and cell proliferation of ASMCs and attenuated airway smooth muscle remodelling in mouse asthma models. CONCLUSIONS AND CLINICAL RELEVANCE: Our results indicate that calpain mediates cytokine-induced collagen-I synthesis and proliferation of ASMCs via the mTORC2/Akt signalling pathway, thereby regulating airway smooth muscle remodelling in asthma.


Subject(s)
Airway Remodeling , Asthma/metabolism , Asthma/pathology , Calpain/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Muscle, Smooth/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Airway Remodeling/drug effects , Airway Remodeling/genetics , Animals , Asthma/immunology , Calpain/antagonists & inhibitors , Calpain/genetics , Cell Proliferation , Collagen Type I/biosynthesis , Cytokines/metabolism , Dipeptides/pharmacology , Disease Models, Animal , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism
3.
Oncogene ; 34(31): 4109-17, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-25347743

ABSTRACT

The non-receptor tyrosine kinase Fer belongs to a distinct subfamily of F-BAR domain containing kinases implicated in vesicular trafficking and signaling downstream of adhesion and growth factor receptors. Targeted inactivation of the fer gene in a transgenic mouse model of HER2(+), breast cancer was associated with delayed tumor onset and reduced proliferative rates in tumor cells. Fer deficiency was associated with increased rates of epidermal growth factor (EGF)-induced epidermal growth factor receptor (EGFR) internalization and amplified Ras-Raf-Mek-Erk (Ras-MAPK) signaling in primary mammary tumor epithelial cells, as well as increased cytotoxic and anti-proliferative sensitivity to the dual EGFR/HER2 inhibitor Lapatinib (LPN). These observations suggest a model in which accelerated ligand-induced EGFR internalization in Fer-deficient cells hypersensitizes the Ras-MAPK pathway to EGF, resulting in MAPK signal amplification to levels that induce cytostasis, rather than proliferation. Thus, Ras-MAPK cytostatic signaling delays HER2 tumor initiation and increases LPN cytotoxicity in Fer-deficient model systems. Taken together, these data suggest that targeting Fer alone, or in combination with LPN, may be of therapeutic benefit in HER2(+) breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein-Tyrosine Kinases/genetics , ras Proteins/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation/genetics , Female , Mice , Mice, Transgenic , Protein Transport , Protein-Tyrosine Kinases/deficiency , Receptor, ErbB-2/genetics , Up-Regulation/physiology
4.
Mol Biol Cell ; 25(19): 3037-48, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25143396

ABSTRACT

Dysferlin and calpain are important mediators of the emergency response to repair plasma membrane injury. Our previous research revealed that membrane injury induces cleavage of dysferlin to release a synaptotagmin-like C-terminal module we termed mini-dysferlinC72. Here we show that injury-activated cleavage of dysferlin is mediated by the ubiquitous calpains via a cleavage motif encoded by alternately spliced exon 40a. An exon 40a-specific antibody recognizing cleaved mini-dysferlinC72 intensely labels the circumference of injury sites, supporting a key role for dysferlinExon40a isoforms in membrane repair and consistent with our evidence suggesting that the calpain-cleaved C-terminal module is the form specifically recruited to injury sites. Calpain cleavage of dysferlin is a ubiquitous response to membrane injury in multiple cell lineages and occurs independently of the membrane repair protein MG53. Our study links calpain and dysferlin in the calcium-activated vesicle fusion of membrane repair, placing calpains as upstream mediators of a membrane repair cascade that elicits cleaved dysferlin as an effector. Of importance, we reveal that myoferlin and otoferlin are also cleaved enzymatically to release similar C-terminal modules, bearing two C2 domains and a transmembrane domain. Evolutionary preservation of this feature highlights its functional importance and suggests that this highly conserved C-terminal region of ferlins represents a functionally specialized vesicle fusion module.


Subject(s)
Calpain/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Synaptotagmins/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins/metabolism , Calpain/genetics , Carrier Proteins , Cells, Cultured , Dysferlin , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Membrane Fusion/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle Proteins/genetics , Protein Isoforms , Protein Structure, Tertiary , Sequence Alignment , Tripartite Motif Proteins
5.
Gut ; 54(8): 1091-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16009680

ABSTRACT

BACKGROUND AND AIMS: Fer kinase is activated by a number of growth factors and cytokines, and phosphorylates cortactin during cell shape change induced cortical actin reorganisation. In addition, Fer participates in cytoskeletal interactions mediated by cadherins, platelet endothelial cell adhesion molecule 1 (PECAM-1), and integrins, and has recently been implicated in limiting the innate immune response. Here we examined the role of Fer in modulating leucocyte recruitment and epithelial barrier function in the gut in response to lipopolysaccharide (LPS). METHODS: Mice targeted with a kinase inactivating mutation (FerDR) or strain matched wild-type (129Sv/J) mice were studied after intraperitoneal injection of LPS. Intravital microscopy was used to examine intestinal leucocyte kinetics, and leucocyte infiltration was assessed by fluorescence activated cell sorting. Systemic inflammation was assessed by measuring lung myeloperoxidase activity. Epithelial barrier function was assessed in vivo using blood to lumen 51Cr-EDTA clearance, with or without antibody based depletion of circulating neutrophils. RESULTS: LPS induced a significant increase in leucocyte adhesion and neutrophil infiltration into the intestinal tissue, and increased blood to lumen 51Cr-EDTA clearance. Pretreatment with neutrophil depleting antibody completely abrogated this response in wild-type mice. In FerDR mice, LPS induced leucocyte adhesion within the intestinal venules was exacerbated and associated with a trend towards increased neutrophil transmigration relative to wild-type mice. Surprisingly, LPS induced epithelial barrier permeability was increased 2.5-fold in FerDR mice relative to wild-type mice, and this barrier defect was only partly attenuated by depleting circulating neutrophils by >93 %. CONCLUSIONS: Fer plays a role in regulating LPS induced epithelial barrier dysfunction in vivo through both neutrophil dependent and neutrophil independent mechanisms.


Subject(s)
Epithelial Cells/immunology , Intestines/immunology , Leukocytes/immunology , Proto-Oncogene Proteins/physiology , Animals , Cell Adhesion/immunology , Cell Movement/immunology , Immunity, Cellular , Intestine, Small/immunology , Leukocyte Count/methods , Lipopolysaccharides/immunology , Mice , Neutrophils/immunology , Permeability , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins/immunology
6.
Cell Death Differ ; 12(5): 512-22, 2005 May.
Article in English | MEDLINE | ID: mdl-15933726

ABSTRACT

We have shown that C2 ceramide, a cell-permeable analog of this lipid second messenger, triggers an NF-kappaB dependent survival pathway that counteracts cell death. Activation of NF-kappaB and subsequent induction of prosurvival genes relies on calpain activity and is prevented on silencing of the calpain small subunit (Capn4) that is required for the function of ubiquitous calpains. We have demonstrated that p105 (NF-kappaB1) and its proteolytic product p50 can be targets of micro- and milli-calpain in vitro and that a p50 deletion mutant, lacking both the N- and the C-terminal ends, is resistant to calpain-mediated degradation. Capn4 silencing results in stabilization of endogenous p105 and p50 in diverse human cell lines. Furthermore, p105 processing and activation of NF-kappaB survival genes in response to C2 ceramide is impaired in Capn4-/- mouse embryonic fibroblasts defective in calpain activity. Altogether, these data argue for the existence of a ceramide-calpain-NF-kappaB axis with prosurvival functions.


Subject(s)
Calpain/metabolism , Cell Survival/physiology , Ceramides/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , Annexin A5/metabolism , Apoptosis , Calpain/antagonists & inhibitors , Cell Death/drug effects , Cell Line, Tumor , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , HeLa Cells , Humans , Mice/embryology , Mice, Knockout , NIH 3T3 Cells , Propidium/metabolism
7.
J Thromb Haemost ; 2(11): 2009-19, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15550033

ABSTRACT

The Fps/Fes tyrosine kinase has been implicated in the regulation of hematopoiesis and inflammation. Mice expressing an activated variant of Fps/Fes (MFps) encoded by a gain-of-function mutant transgenic fps/fes allele (fps(MF)) exhibited hematological phenotypes, which suggested that Fps/Fes can direct hematopoietic lineage output. These mice also displayed marked hypervascularity and multifocal-hemangiomas which implicated this kinase in the regulation of angiogenesis. Here we explored the potential involvement of Fps/Fes in the regulation of hemostasis through effects on blood cells and the vascular endothelium. Hematological parameters of fps(MF) mice were characterized by peripheral blood analysis, histology, and transmission electron microscopy. Hemostasis parameters and platelet functions were assessed by flow cytometry and measurements of activated partial thromboplastin time, prothrombin time, thrombin clot time, platelet aggregation, bleeding times and in vitro fibrinolytic assays. Hematological and morphological analyses showed that fps(MF) mice displayed mild thrombocytopenia, anemia, red cell abnormalities and numerous hemostatic defects, including hypofibrinogenemia, hyper-fibrinolysis, impaired whole blood aggregation and a mild bleeding diathesis. fps(MF) mice displayed a complex array of hemostatic perturbations which are reminiscent of hemostatic disorders such as disseminated intravascular coagulation (DIC) and of hemangioma-associated pathologies such as Kasabach-Merritt phenomenon (KMS). These studies suggest that Fps/Fes influences both angiogenic and hemostatic function through regulatory effects on the endothelium.


Subject(s)
Hemostasis , Neovascularization, Physiologic , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Anemia/etiology , Animals , Blood Coagulation Disorders/etiology , Blood Platelets/pathology , Endothelium, Vascular/pathology , Hemolysis , Mice , Mice, Transgenic , P-Selectin/analysis , Phenotype , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fes , Thrombocytopenia/etiology , Thrombopoiesis
8.
J Thromb Haemost ; 2(5): 820-32, 2004 May.
Article in English | MEDLINE | ID: mdl-15099290

ABSTRACT

BACKGROUND: Fps/Fes is a cytoplasmic tyrosine kinase that is abundantly expressed in the myeloid, endothelial, epithelial, neuronal and platelet lineages. Genetic manipulation in mice has uncovered potential roles for this kinase in hematopoiesis, innate immunity, inflammation and angiogenesis. OBJECTIVE: We have utilized a genetic approach to explore the role of Fps/Fes in angiogenesis. METHODS: A hypervascular line of mice generated by expression of a 'gain-of-function' human fps/fes transgene (fps(MF)) encoding a myristoylated variant of Fps (MFps) was used in these studies. The hypervascular phenotype of this line was extensively characterized by intravital microscopy and biochemical approaches. RESULTS: fps(MF) mice exhibited 1.6-1.7-fold increases in vascularity which was attributable to increases in the number of secondary vessels. Vessels were larger, exhibited varicosities and disorganized patterning, and were found to have defects in histamine-induced permeability. Biochemical characterization of endothelial cell (EC) lines derived from fps(MF) mice revealed that MFps was hypersensitive to activation by vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). CONCLUSIONS: MFps mediates enhanced sensitization to VEGF and PDGF signaling in ECs. We propose that this hypersensitization contributes to excessive angiogenic signaling and that this underlies the observed hypervascular phenotype of fps(MF) mice. These phenotypes recapitulate important aspects of the vascular defects observed in both VEGF and angiopoietin-1 transgenic mice. The fps/fes proto-oncogene product therefore represents a novel player in the regulation of angiogenesis, and the fps(MF) line of mice constitutes a unique new murine model for the study of this process.


Subject(s)
Blood Vessels/abnormalities , Neovascularization, Pathologic , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Vascular Endothelial Growth Factor A/pharmacology , Animals , Blood Vessels/drug effects , Blood Vessels/growth & development , Brain/blood supply , Cell Line , Coronary Vessels , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Hemodynamics/drug effects , Humans , Mice , Mice, Transgenic , Neovascularization, Pathologic/chemically induced , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-fes
9.
J Thromb Haemost ; 1(5): 1062-70, 2003 May.
Article in English | MEDLINE | ID: mdl-12871378

ABSTRACT

Fps/Fes and Fer proto-oncoproteins are structurally related non-receptor protein-tyrosine kinases implicated in signaling downstream from cytokines, growth factors and immune receptors. We show that Fps/Fes and Fer are expressed in human and mouse platelets, and are activated following stimulation with collagen and collagen-related peptide (CRP), suggesting a role in GPVI receptor signaling. Fer was also activated following stimulation with thrombin and a protease-activated receptor4 (PAR4)-activating peptide, suggesting a role in signaling downstream from the G protein-coupled PAR4. There were no detectable perturbations in CRP-induced activation of Syk, PLCgamma2, cortactin, Erk, Jnk, Akt or p38 in platelets from mice lacking Fps/Fes, Fer, or both kinases. Platelets lacking Fps/Fes, from a targeted fps/fes null strain of mice, showed increased rates and amplitudes of collagen-induced aggregation, relative to wild-type platelets. P-Selectin expression was also elevated on the surface of Fps/Fes-null platelets in response to CRP. Fer-deficient platelets, from mice targeted with a kinase-inactivating mutation, disaggregated more rapidly than wild-type platelets in response to ADP. This report provides the first evidence that Fps/Fes and Fer are expressed in platelets and become activated downstream from the GPVI collagen receptor, and that Fer is activated downstream from a G-protein coupled receptor. Furthermore, using targeted mouse models we show that deficiency in Fps/Fes or Fer resulted in disregulated platelet aggregation and disaggregation, demonstrating a role for these kinases in regulating platelet functions.


Subject(s)
Fusion Proteins, gag-onc/physiology , Platelet Aggregation , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/enzymology , Collagen/pharmacology , Fusion Proteins, gag-onc/analysis , Humans , Mice , Mice, Transgenic , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins , Protein-Tyrosine Kinases/analysis , Proto-Oncogene Proteins/analysis , Receptors, Cell Surface , Signal Transduction
10.
Mol Cell Biol ; 22(1): 257-69, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11739739

ABSTRACT

v-Src-induced oncogenic transformation is characterized by alterations in cell morphology, adhesion, motility, survival, and proliferation. To further elucidate some of the signaling pathways downstream of v-Src that are responsible for the transformed cell phenotype, we have investigated the role that the calpain-calpastatin proteolytic system plays during oncogenic transformation induced by v-Src. We recently reported that v-Src-induced transformation of chicken embryo fibroblasts is accompanied by calpain-mediated proteolytic cleavage of the focal adhesion kinase (FAK) and disassembly of the focal adhesion complex. In this study we have characterized a positive feedback loop whereby activation of v-Src increases protein synthesis of calpain II, resulting in degradation of its endogenous inhibitor calpastatin. Reconstitution of calpastatin levels by overexpression of exogenous calpastatin suppresses proteolytic cleavage of FAK, morphological transformation, and anchorage-independent growth. Furthermore, calpastatin overexpression represses progression of v-Src-transformed cells through the G(1) stage of the cell cycle, which correlates with decreased pRb phosphorylation and decreased levels of cyclins A and D and cyclin-dependent kinase 2. Calpain 4 knockout fibroblasts also exhibit impaired v-Src-induced morphological transformation and anchorage-independent growth. Thus, modulation of the calpain-calpastatin proteolytic system plays an important role in focal adhesion disassembly, morphological transformation, and cell cycle progression during v-Src-induced cell transformation.


Subject(s)
CDC2-CDC28 Kinases , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Cell Transformation, Neoplastic , Oncogene Protein pp60(v-src)/metabolism , Actins/metabolism , Animals , Calcium-Binding Proteins/genetics , Calpain/antagonists & inhibitors , Calpain/genetics , Cell Division/physiology , Cell Size , Cells, Cultured , Chick Embryo , Cyclin A/metabolism , Cyclin D , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Cytoskeleton/metabolism , Feedback, Physiological/physiology , Fibroblasts/physiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Focal Adhesions/metabolism , Genes, myc , Genes, ras , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Oncogene Protein p65(gag-jun)/metabolism , Oncogene Proteins v-fos/metabolism , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Retinoblastoma Protein/metabolism , Temperature
11.
J Biol Chem ; 276(51): 48382-8, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11602605

ABSTRACT

The physiological functions and substrates of the calcium-dependent protease calpain remain only partly understood. The mu- and m-calpains consist of a mu- or m-80-kDa large subunit (genes Capn1 and Capn2), and a common 28-kDa small subunit (Capn4). To assess the role of calpain in migration, we used fibroblasts obtained from Capn4(-/-) mouse embryos. The cells lacked calpain activity on casein zymography and did not generate the characteristic calpain-generated spectrin breakdown product that is observed in wild-type cells. Capn4(-/-) cells had decreased migration rates and abnormal organization of the actin cytoskeleton with a loss of central stress fibers. Interestingly, these cells extended numerous thin projections and displayed delayed retraction of membrane protrusions and filopodia. The number of focal adhesions was decreased in Capn4(-/-) cells, but the cells had prominent vinculin-containing focal complexes at the cell periphery. The levels of the focal adhesion proteins, alpha-actinin, focal adhesion kinase (FAK), spectrin, talin, and vinculin, were the same in Capn4(+/+) and Capn4(-/-) cells. FAK, alpha-actinin, and vinculin were not cleaved in either cell type plated on fibronectin. However, proteolysis of the focal complex component, talin, was detected in the wild-type cells but not in the Capn4(-/-) cells, suggesting that calpain cleavage of talin is important during cell migration. Moreover, talin cleavage was again observed when calpain activity was partially restored in Capn4(-/-) embryonic fibroblasts by stable transfection with a vector expressing the rat 28-kDa calpain small subunit. The results demonstrate unequivocally that calpain is a critical regulator of cell migration and of the organization of the actin cytoskeleton and focal adhesions.


Subject(s)
Actins/metabolism , Calpain/physiology , Cell Movement/physiology , Cytoskeleton/metabolism , Embryo, Mammalian/metabolism , Animals , Antigens, Polyomavirus Transforming/physiology , Calpain/genetics , Cell Line , Cell Line, Transformed , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Hydrolysis , Mice , Rats , Talin/metabolism
12.
J Exp Med ; 194(5): 581-9, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11535627

ABSTRACT

A somatic mutation in the X-linked phosphatidylinositol glycan class A (PIGA) gene causes the loss of glycosyl phosphatidylinositol (GPI)-linked proteins on blood cells from patients with paroxysmal nocturnal hemoglobinuria. Because all blood cell lineages may be affected it is thought that the mutation occurs in a hematopoietic stem cell. In transgenic mice, germline transmission of an inactive Piga gene is embryonic lethal. To inactivate the murine Piga gene in early hematopoiesis we therefore chose conditional gene inactivation using the Cre/loxP system. We expressed Cre recombinase under the transcription regulatory sequences of the human c-fes gene. FES-Cre inactivated PIGA in hematopoietic cells of mice carrying a floxed Piga allele (LF mice). PIGA(-) cells were found in all hematopoietic lineages of definitive but not primitive hematopoiesis. Their proportions were low in newborn mice but subsequently increased continuously to produce for the first time mice that have almost exclusively PIGA(-) blood cells. The loss of GPI-linked proteins occurred mainly in c-kit(+)CD34(+)Lin(-) progenitor cells before the CFU-GEMM stage. Using bone marrow reconstitution experiments with purified PIGA(-) cells we demonstrate that LF mice have long-term bone marrow repopulating cells that lack GPI-linked proteins, indicating that recombination of the floxed Piga allele occurs in the hematopoietic stem cell.


Subject(s)
Gene Expression Regulation, Developmental , Glycosylphosphatidylinositols/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Integrases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/metabolism , Viral Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Colony-Forming Units Assay , Female , Fetal Death , Hemoglobinuria, Paroxysmal/genetics , Humans , Integrases/genetics , Mice , Mice, Transgenic , Polymerase Chain Reaction , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fes , Proto-Oncogenes , Recombination, Genetic , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Viral Proteins/genetics
13.
DNA Cell Biol ; 20(4): 203-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11403717

ABSTRACT

Eph receptors and their membrane-associated ephrin ligands regulate cell-cell interactions during development. The biochemical and biologic functions of this receptor tyrosine kinase family are still being elucidated but include roles in nervous system segmentation, axon pathfinding, and angiogenesis. To isolate murine orthologs of three zebrafish Eph family members (zek1, zek2, and zek3), we have used a degenerate RT-PCR-based cloning method specific for members of the Eph family. Although this method was effective for isolation of Eph receptor cDNAs, including members of both the A and B subfamilies, our results suggested that zek1 may not have a murine ortholog. The isolated cDNAs were also used to generate RNA in situ hybridization probes to examine the expression patterns of murine EphA2, A3, A4, A7, B1, B2, and B4 in 9.5-dpc mouse embryos. In addition to the expected abundant expression of these Eph receptors in the developing CNS and the presence of EphB receptors in vascular tissues, several of the EphA receptors were expressed in discrete regions of the developing vasculature. These results suggest a role for both EphA and EphB receptors in vascular development.


Subject(s)
Blood Vessels/embryology , Central Nervous System/embryology , Cloning, Molecular/methods , Gene Expression Regulation, Developmental , Multigene Family , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Eph Family , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish Proteins , Amino Acid Sequence , Animals , Blood Vessels/metabolism , Blotting, Southern , Central Nervous System/metabolism , DNA, Complementary/genetics , Evolution, Molecular , Female , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , Gene Library , Gestational Age , Mice , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nucleic Acid Hybridization , Phylogeny , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Vertebrates/genetics , Zebrafish/genetics
14.
Exp Cell Res ; 266(1): 87-94, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11339827

ABSTRACT

The subcellular localizations of the Fps/Fes and closely related Fer cytoplasmic tyrosine kinases were studied using green fluorescent protein (GFP) fusions and confocal fluorescence microscopy. In contrast to previous reports, neither kinase localized to the nucleus. Fer was diffusely cytoplasmic throughout the cell cycle. Fps/Fes also displayed a diffuse cytoplasmic localization, but in addition it showed distinct accumulations in cytoplasmic vesicles as well as in a perinuclear region consistent with the Golgi. This localization was very similar to that of TGN38, a known marker of the trans Golgi. The localization of Fps/Fes and TGN38 were both perturbed by brefeldin A, a fungal metabolite that disrupts the Golgi apparatus. Fps/Fes was also found to colocalize to various extents with several Rab proteins, which are members of the monomeric G-protein superfamily involved in vesicular transport between specific subcellular compartments. Using Rabs that are involved in endocytosis (Rab5B and Rab7) or exocytosis (Rab1A and Rab3A), we showed that Fps/Fes is localized in both pathways. These results suggest that Fps/Fes may play a general role in the regulation of vesicular trafficking.


Subject(s)
Cell Compartmentation/physiology , Cytoplasm/metabolism , Fusion Proteins, gag-onc/metabolism , Glycoproteins , Membrane Proteins , Protein Transport/physiology , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/metabolism , Transport Vesicles/metabolism , Animals , COS Cells/cytology , COS Cells/metabolism , Cell Cycle/physiology , Cytoplasm/ultrastructure , Golgi Apparatus/metabolism , Immunohistochemistry , Intracellular Membranes/metabolism , Membrane Glycoproteins/metabolism , Transport Vesicles/ultrastructure , rab GTP-Binding Proteins/metabolism
15.
Mol Cell Biol ; 21(2): 603-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134346

ABSTRACT

The ubiquitous Fer protein-tyrosine kinase has been proposed to regulate diverse processes such as cell growth, cell adhesion, and neurite outgrowth. To gain insight into the biological function of Fer, we have targeted the fer locus with a kinase-inactivating missense mutation (fer(D743R)). Mice homozygous for this mutation develop normally, have no overt phenotypic differences from wild-type mice, and are fertile. Since these mice lack both Fer and the testis-specific FerT kinase activities, these proteins are clearly not essential for development and survival. No differences were observed in overall cellularity of bone marrow, spleen, or thymus in the absence of Fer activity. While most platelet-derived growth factor (PDGF)-induced tyrosine phosphorylation was unchanged in fer(D743R) homozygous embryonic fibroblasts, cortactin phosphorylation was reduced. However, Fer kinase activity was not required for PDGF-induced Stat3, p120(ctn), or epidermal growth factor (EGF)-induced beta-catenin phosphorylation. Also, no defects were observed in changes to the actin cytoskeleton, adherens junctions, or focal adhesions in PDGF- or EGF-stimulated fer(D743R) homozygous embryonic fibroblasts. Therefore, Fer likely serves a redundant role in regulating cell growth, cell adhesion, retinal development, and spermatogenesis but is required for efficient phosphorylation of cortactin.


Subject(s)
Fertility/genetics , Gene Deletion , Microfilament Proteins/metabolism , Proto-Oncogene Proteins/deficiency , Trans-Activators , Actins/metabolism , Alleles , Animals , Catenins , Cell Adhesion Molecules/metabolism , Cortactin , Cytoskeletal Proteins/metabolism , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epidermal Growth Factor/pharmacology , Fibroblasts , Gene Targeting , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Phenotype , Phosphoproteins/metabolism , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Survival Rate , beta Catenin , Delta Catenin
16.
Mol Cell Biol ; 20(12): 4474-81, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825211

ABSTRACT

Calpains are a family of Ca(2+)-dependent intracellular cysteine proteases, including the ubiquitously expressed micro- and m-calpains. Both mu- and m-calpains are heterodimers, consisting of a distinct large 80-kDa catalytic subunit, encoded by the genes Capn1 and Capn2, and a common small 28-kDa regulatory subunit (Capn4). The physiological roles and possible functional distinctions of mu- and m-calpains remain unclear, but suggested functions include participation in cell division and migration, integrin-mediated signal transduction, apoptosis, and regulation of cellular control proteins such as cyclin D1 and p53. Homozygous disruption of murine Capn4 eliminated both mu- and m-calpain activities, but this did not affect survival and proliferation of cultured embryonic stem cells or embryonic fibroblasts, or the early stages of organogenesis. However, mutant embryos died at midgestation and displayed defects in the cardiovascular system, hemorrhaging, and accumulation of erythroid progenitors.


Subject(s)
Calpain/genetics , Gene Expression Regulation, Developmental , Animals , Cell Division/genetics , Embryonic and Fetal Development/genetics , Gene Deletion , Mice
17.
Mol Cell Biol ; 19(11): 7436-46, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523632

ABSTRACT

The fps/fes proto-oncogene encodes a cytoplasmic protein-tyrosine kinase that is functionally implicated in the survival and terminal differentiation of myeloid progenitors and in signaling from several members of the cytokine receptor superfamily. To gain further insight into the physiological function of fps/fes, we targeted the mouse locus with a kinase-inactivating missense mutation. Mutant Fps/Fes protein was expressed at normal levels in these mice, but it lacked detectable kinase activity. Homozygous mutant animals were viable and fertile, and they showed no obvious defects. Flow cytometry analysis of bone marrow showed no statistically significant differences in the levels of myeloid, erythroid, or B-cell precursors. Subtle abnormalities observed in mutant mice included slightly elevated total leukocyte counts and splenomegaly. In bone marrow hematopoietic progenitor cell colony-forming assays, mutant mice gave slightly elevated numbers and variable sizes of CFU-granulocyte macrophage in response to interleukin-3 (IL-3) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Tyrosine phosphorylation of Stat3 and Stat5A in bone marrow-derived macrophages was dramatically reduced in response to GM-CSF but not to IL-3 or IL-6. This suggests a distinct nonredundant role for Fps/Fes in signaling from the GM-CSF receptor that does not extend to the closely related IL-3 receptor. Lipopolysaccharide-induced Erk1/2 activation was also reduced in mutant macrophages. These subtle molecular phenotypes suggest a possible nonredundant role for Fps/Fes in myelopoiesis and immune responses.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Milk Proteins , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Fertility , Homozygote , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutation, Missense , Organ Size , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-3/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction , Spleen , Trans-Activators/metabolism
18.
Biochim Biophys Acta ; 1388(1): 247-52, 1998 Oct 14.
Article in English | MEDLINE | ID: mdl-9774740

ABSTRACT

The calpains comprise a family of heterodimeric (80+28 kDa) Ca2+-dependent cysteine proteases, probably having roles in signal transduction and cytoskeletal remodelling. We describe cloning and sequencing of the 28 kDa calpain subunit cDNA from mouse (coding for 268 amino acids), and characterization of its gene. The gene spans 7 kb and contains 11 exons. The promoter region, like those of other calpain genes, lacks an obvious TATA box, but contains several Sp1 binding sites.


Subject(s)
Calpain/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , DNA, Complementary/analysis , DNA, Complementary/isolation & purification , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic
19.
Exp Cell Res ; 237(2): 452-5, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9434641

ABSTRACT

DNA topoisomerase II alpha is the intracellular target for several important chemotherapeutic agents, and drug-resistant human tumor cell lines have been described in which deletions in the C-proximal region of this enzyme are associated with its cytoplasmic localization. We have identified multiple potential bipartite nuclear localization signal (NLS) sequences in this region using a modified definition of the motif, and in the present study, we have expressed five of these as fusion proteins with beta-galactosidase. Only one sequence (spanning amino acids 1454 to 1497) was sufficient to cause strong nuclear localization. Subsequent mutation analyses indicated that this NLS sequence was bipartite and that both domains contain more than two basic amino acids. Substitution of the lysine residue at position 1492 in the second basic domain with glutamine resulted in a fusion protein that localized inefficiently to the nucleus, indicating that all three basic residues in this domain are necessary. Our results confirm that a broader definition is required to detect all potential bipartite NLS motifs in a polypeptide sequence, although functional tests are still essential for identification of those sequences actually capable of directing nuclear localization.


Subject(s)
DNA Topoisomerases, Type II/chemistry , Nuclear Localization Signals , Amino Acid Sequence , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Structure-Activity Relationship
20.
Fertil Steril ; 63(5): 1088-93, 1995 May.
Article in English | MEDLINE | ID: mdl-7720923

ABSTRACT

OBJECTIVE: To evaluate the direct effect of photodynamic treatment with 5-aminolevulinic acid (ALA) on preimplantation mouse embryos in an in vitro setting. DESIGN: Preimplantation mouse embryos were incubated with or without ALA for 5 hours and followed immediately by light exposure for 0, 5, or 15 minutes. Comparison of the viability and blastocyst formation was made among different treatment groups. SETTING: A conventional laboratory setting with embyro culture facilities. INTERVENTIONS: Female CD1 mice were superovulated with pregnant mare serum gonadotropin and hCG before mating. Four-and eight-cell embryos and compacted morulae were flushed from the oviducts and incubated with 0, 0.1, 0.5, 1.0, or 5.0 mM ALA for 5 hours. Embryos subsequently were exposed to photoactivating light for 0, 5, or 15 minutes. MAIN OUTCOME MEASURES: Microscopic assessment of embryos quality at 12 hours and determination of the percentage of embryos progressing to the blastocyst stage at 36 or 60 hours. RESULTS: Incubation of embryos with 0.1, 0.5, 1.0, 5.0 mM ALA without light resulted in 87.3% +/- 1.6%, 84.9% +/- 3.4%, 81.4% +/- 1.8%, and 82.8% +/- 4.7% of the embryos developing to blastocysts, respectively. In the absence of ALA, light exposure for 0, 5, or 15 minutes resulted in 93.8% +/- 2.3%, 92.3% +/- 2.2%, and 85.9% +/- 1.7% blastocyst formation. Combining treatment of ALA at the same concentrations with light resulted in 33.3% +/- 2.1%, 0.7% +/- 0.9%, 0%, 0% (5-minute light), 13.3% +/- 1.0%, 0%, 1.6% +/- 1.3%, 0% (15-minute light) blastocyst formation, respectively. When gross morphology was used to assess embryo viability at 12 hours, similar results were observed. Measurement of the fluorescent spectrum of embryos incubated with ALA indicated that protoporphyrin IX had been formed. CONCLUSION: Photodynamic ablation of mouse embryos was achieved with ALA under in vitro conditions. These results indicate that preimplantation mouse embryos are capable of converting ALA to the photosensitizer, protoporphyrin IX, and are susceptible to subsequent photoablation. A photodynamic effect on the embryo may be important to the successful application of this technique to the treatment of human ectopic pregnancy.


Subject(s)
Aminolevulinic Acid/toxicity , Blastocyst/drug effects , Light , Animals , Blastocyst/radiation effects , Chorionic Gonadotropin/pharmacology , Female , Gonadotropins, Equine/pharmacology , Mice , Morula/drug effects , Morula/radiation effects , Photochemistry , Pregnancy , Protoporphyrins/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...