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










Publication year range
1.
Cell Genom ; 4(3): 100510, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38447574

ABSTRACT

CRISPR-Cas9 short guide RNA (sgRNA) library screening is a powerful approach to understand the molecular mechanisms of biological phenomena. However, its in vivo application is currently limited. Here, we developed our previously established in vitro revival screening method into an in vivo one to identify factors involved in spermatogenesis integrity by utilizing sperm capacitation as an indicator. By introducing an sgRNA library into testicular cells, we successfully pinpointed the retinal degeneration 3 (Rd3) gene as a significant factor in spermatogenesis. Single-cell RNA sequencing (scRNA-seq) analysis highlighted the high expression of Rd3 in round spermatids, and proteomics analysis indicated that Rd3 interacts with mitochondria. To search for cell-type-specific signaling pathways based on scRNA-seq and proteomics analyses, we developed a computational tool, Hub-Explorer. Through this, we discovered that Rd3 modulates oxidative stress by regulating mitochondrial distribution upon ciliogenesis induction. Collectively, our screening system provides a valuable in vivo approach to decipher molecular mechanisms in biological processes.


Subject(s)
RNA, Guide, CRISPR-Cas Systems , Semen , Male , Humans , Testis , Spermatids , Spermatogenesis/genetics
2.
Nat Commun ; 14(1): 5592, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37696806

ABSTRACT

The "eat me" signal, phosphatidylserine is exposed on the surface of dying cells by phospholipid scrambling. Previously, we showed that the Xkr family protein Xkr4 is activated by caspase-mediated cleavage and binding of the XRCC4 fragment. Here, we show that extracellular calcium is an additional factor needed to activate Xkr4. The constitutively active mutant of Xkr4 is found to induce phospholipid scrambling in an extracellular, but not intracellular, calcium-dependent manner. Importantly, other Xkr family members also require extracellular calcium for activation. Alanine scanning shows that D123 and D127 of TM1 and E310 of TM3 coordinate calcium binding. Moreover, lysine scanning demonstrates that the E310K mutation-mediated salt bridge between TM1 and TM3 bypasses the requirement of calcium. Cysteine scanning proves that disulfide bond formation between TM1 and TM3 also activates phospholipid scrambling without calcium. Collectively, this study shows that extracellular calcium functions as a molecular glue for TM1 and TM3 of Xkr proteins for activation, thus demonstrating a regulatory mechanism for multi-transmembrane region-containing proteins.


Subject(s)
Alanine , Calcium , Biological Transport , Caspases , Phosphatidylserines
3.
Cell Rep ; 34(12): 108876, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761359

ABSTRACT

ATP-competitive inhibitors have been developed as promising anti-cancer agents. However, drug-resistance frequently occurs, and the underlying mechanisms are not fully understood. Here, we show that the activation of c-Src and its downstream phosphorylation cascade can be paradoxically induced by Src-targeted and RTK-targeted kinase inhibitors. We reveal that inhibitor binding induces a conformational change in c-Src, leading to the association of the active form c-Src with focal adhesion kinase (FAK). Reduction of the inhibitor concentration results in the dissociation of inhibitors from the c-Src-FAK complex, which allows c-Src to phosphorylate FAK and initiate FAK-Grb2-mediated Erk signaling. Furthermore, a drug-resistant mutation in c-Src, which reduces the affinity of inhibitors for c-Src, converts Src inhibitors into facilitators of cell proliferation by enhancing the phosphorylation of FAK and Erk in c-Src-mutated cells. Our data thus reveal paradoxical enhancement of cell growth evoked by target-based kinase inhibitors, providing potentially important clues for the future development of effective and safe cancer treatment.


Subject(s)
Drug Resistance, Neoplasm , src-Family Kinases/metabolism , Animals , Base Sequence , Dasatinib/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , GRB2 Adaptor Protein/metabolism , Humans , Ligands , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Models, Biological , Mutation/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Xenopus , src Homology Domains , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
4.
Mol Cell ; 81(7): 1397-1410.e9, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33725486

ABSTRACT

Phospholipid scrambling in dying cells promotes phosphatidylserine exposure, a critical process for efferocytosis. We previously identified the Xkr family protein Xkr4 as a phospholipid-scrambling protein, but its activation mechanisms remain unknown. Here we show that Xkr4 is activated in two steps: dimer formation by caspase-mediated cleavage and structural change caused by activating factors. To identify the factors, we developed a new screening system, "revival screening," using a CRISPR sgRNA library. Applying this system, we identified the nuclear protein XRCC4 as the single candidate for the Xkr4 activator. Upon apoptotic stimuli, XRCC4, contained in the DNA repair complex, is cleaved by caspases, and its C-terminal fragment with an intrinsically disordered region is released into the cytoplasm. Protein interaction screening showed that the fragment interacts directly with the Xkr4 dimer to activate it. This study demonstrates that caspase-mediated cleavage releases a nuclear protein fragment for direct regulation of lipid dynamics on the plasma membrane.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Membrane/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Phospholipids/metabolism , Proteolysis , Animals , Apoptosis Regulatory Proteins/genetics , Caspases/genetics , Cell Line, Tumor , Cell Membrane/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Phospholipids/genetics , Protein Multimerization
5.
Neurosci Res ; 167: 30-37, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33476682

ABSTRACT

Phospholipids are asymmetrically distributed at the plasma membrane. Phosphatidylserine (PtdSer) is exclusively located in the inner leaflet of the cell membrane while phosphatidylcholine (PtdCho) and glycolipids are mainly located in the outer leaflet of the membrane. However, this asymmetry is disrupted in various physiological situations, and PtdSer is exposed on the cell surface. In platelets, exposed PtdSer functions as a scaffold for the coagulation reaction, while in dead cells, exposed PtdSer serves as an "Eat-me" signal for efferocytosis. In the developing brain, synaptic connections are over-formed during the fetal period, but about half of the neurons are removed by apoptosis, and synaptic and dendritic compartments of living neurons are also removed by phagocytes. During these processes, glial cells such as microglia and astrocyte engulf unwanted dead cells and compartments in living cells using several phagocytic receptors, recognizing PtdSer by direct binding or an indirect way using secreted molecules. Based on recent findings, we will discuss how the compartments in living neurons are eliminated for the neuronal circuit plasticity.


Subject(s)
Phosphatidylserines , Phospholipids , Apoptosis , Brain , Cell Membrane
6.
Genes Cells ; 23(3): 214-224, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29431243

ABSTRACT

The immunoglobulin (Ig)-like cell adhesion molecule nectin-like molecule (Necl)-5/poliovirus receptor is up-regulated in many types of cancer cells and implicated in their abnormally enhanced cell proliferation and movement. We previously showed that Necl-5 cis-interacts with the platelet-derived growth factor (PDGF) receptor ß through the extracellular region and enhances its signaling. Although this cis-interaction does not affect the PDGF-induced tyrosine phosphorylation of the receptor, the interaction of the cytoplasmic region of Necl-5 with sprouty2 and the regulation of its activity are required for the enhancement of the PDGF receptor ß signaling by Necl-5. We investigated here the more detailed mechanism for this cis-interaction of Necl-5 with the PDGF receptor ß. Necl-5 contains three Ig-like domains and the PDGF receptor ß contains five Ig-like domains at their extracellular regions. We showed here that the third Ig-like domain of Necl-5 cis-interacted with the fifth Ig-like domain of the PDGF receptor ß. The recombinant protein of the third Ig-like domain of Necl-5 inhibited the cis-interaction of full-length Necl-5 with the PDGF receptor ß and the PDGF-induced activation of the ERK signaling pathway that was enhanced by Necl-5. These results revealed the novel roles of the third Ig-like domain of Necl-5 and the fifth Ig-like domain of the PDGF receptor ß in its signaling.


Subject(s)
Immunoglobulin Domains , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Virus/metabolism , Animals , Binding, Competitive , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NIH 3T3 Cells , Phosphorylation , Protein Binding , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
7.
Sci Rep ; 7(1): 11375, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900130

ABSTRACT

The ligand-induced dimerization of cell surface single-transmembrane receptors is essential for their activation. However, physiological molecules that inhibit their dimerization and activation have not been identified. ErbB3 dimerizes with ErbB2 upon binding of heregulin (HRG) to ErbB3, causing the ErbB2-catalyzed tyrosine phosphorylation of ErbB3, which leads to the activation of the signalling pathways for cell movement and survival. Genetic disorders of this receptor cause tumorigenesis and metastasis of cancers. We show here that nectin-like molecule-4/cell adhesion molecule 4, known to serve as a tumour suppressor, interacts with ErbB3 in the absence of HRG and inhibits the HRG-induced dimerization of ErbB3 with ErbB2 and its activation. The third immunoglobulin-like domain of nectin-like molecule-4 cis-interacts with the extracellular domain 3 of ErbB3. We describe here a novel regulatory mechanism for the activation and signalling of cell surface single-transmembrane receptors.


Subject(s)
Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Cell Adhesion Molecules/chemistry , Cell Line , Extracellular Space/metabolism , Humans , Immunoglobulins/chemistry , Ligands , Neuregulin-1/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Receptor, ErbB-2/chemistry , Receptor, ErbB-3/chemistry
8.
J Biol Chem ; 292(17): 6895-6909, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28258213

ABSTRACT

Cell-surface cytokine receptors are regulated by their cis-interacting stimulatory and inhibitory co-receptors. We previously showed that the Ig-like cell-adhesion molecule nectin-4 cis-interacts with the prolactin receptor through the extracellular region and stimulates prolactin-induced prolactin receptor activation and signaling, resulting in alveolar development in the mouse mammary gland. However, it remains unknown how this interaction stimulates these effects. We show here that the cis-interaction of the extracellular region of nectin-4 with the prolactin receptor was not sufficient for eliciting these effects and that the cytoplasmic region of nectin-4 was also required for this interaction. The cytoplasmic region of nectin-4 directly interacted with suppressor of cytokine signaling 1 (SOCS1), but not SOCS3, JAK2, or STAT5a, and inhibited the interaction of SOCS1 with JAK2, eventually resulting in the increased phosphorylation of STAT5a. The juxtamembrane region of nectin-4 interacted with the Src homology 2 domain of SOCS1. Both the interaction of nectin-4 with the extracellular region of the prolactin receptor and the interaction of SOCS1 with the cytoplasmic region of nectin-4 were required for the stimulatory effect of nectin-4 on the prolactin-induced prolactin receptor activation. The third Ig-like domain of nectin-4 and the second fibronectin type III domain of the prolactin receptor were involved in this cis-interaction, and both the extracellular and transmembrane regions of nectin-4 and the prolactin receptor were required for this direct interaction. These results indicate that nectin-4 serves as a stimulatory co-receptor for the prolactin receptor by regulating the feedback inhibition of SOCS1 in the JAK2-STAT5a signaling pathway.


Subject(s)
Cell Adhesion Molecules/metabolism , Janus Kinase 2/metabolism , Receptors, Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Cytoplasm/metabolism , Female , Fibronectins/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mammary Glands, Animal/metabolism , Mice , Mutation , Phosphorylation , Prolactin/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
9.
J Biol Chem ; 291(11): 5817-5831, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26757815

ABSTRACT

Mammary gland development is induced by the actions of various hormones to form a structure consisting of collecting ducts and milk-secreting alveoli, which comprise two types of epithelial cells known as luminal and basal cells. These cells adhere to each other by cell adhesion apparatuses whose roles in hormone-dependent mammary gland development remain largely unknown. Here we identified a novel cell adhesion apparatus at the boundary between the luminal and basal cells in addition to desmosomes. This apparatus was formed by the trans-interaction between the cell adhesion molecules nectin-4 and nectin-1, which were expressed in the luminal and basal cells, respectively. Nectin-4 of this apparatus further cis-interacted with the prolactin receptor in the luminal cells to enhance the prolactin-induced prolactin receptor signaling for alveolar development with lactogenic differentiation. Thus, a novel nectin-mediated cell adhesion apparatus regulates the prolactin receptor signaling for mammary gland development.


Subject(s)
Cell Adhesion Molecules/metabolism , Mammary Glands, Animal/growth & development , Receptors, Prolactin/metabolism , Signal Transduction , Animals , Cell Adhesion , Cell Adhesion Molecules/analysis , Cell Communication , Female , HEK293 Cells , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/ultrastructure , Mice, Inbred C57BL , Nectins , Prolactin/metabolism
10.
Nat Methods ; 12(8): 743-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147917

ABSTRACT

We have developed a multitarget super-resolution microscopy technique called image reconstruction by integrating exchangeable single-molecule localization (IRIS). IRIS uses protein fragment-based probes that directly associate with and dissociate from their targets over durations on the order of tens of milliseconds. By integrating single-molecule localization and sequential labeling, IRIS enables unprecedented labeling density along multiple cellular structures. IRIS can be used to discern the area-specific proximity between cytoskeletal components and focal adhesions within a single cell.


Subject(s)
Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Actins/chemistry , Animals , Cytoskeleton/metabolism , Expressed Sequence Tags , Focal Adhesions/metabolism , Green Fluorescent Proteins/chemistry , Humans , Image Processing, Computer-Assisted/methods , Mice , Microtubules/chemistry , Oxygen/chemistry , Peptides/chemistry , Plasmids/metabolism , Rats , Xenopus laevis
11.
Nat Cell Biol ; 15(4): 395-405, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455479

ABSTRACT

Physical force evokes rearrangement of the actin cytoskeleton. Signalling pathways such as tyrosine kinases, stretch-activated Ca(2+) channels and Rho GTPases are involved in force sensing. However, how signals are transduced to actin assembly remains obscure. Here we show mechanosensitive actin polymerization by formins (formin homology proteins). Cells overexpressing mDia1 increased the amount of F-actin on release of cell tension. Fluorescence single-molecule speckle microscopy revealed rapid induction of processive actin assembly by mDia1 on cell cortex deformation. mDia1 lacking the Rho-binding domain and other formins exhibited mechanosensitive actin nucleation, suggesting Rho-independent activation. Mechanosensitive actin nucleation by mDia1 required neither Ca(2+) nor kinase signalling. Overexpressing LIM kinase abrogated the induction of processive mDia1. Furthermore, s-FDAPplus (sequential fluorescence decay after photoactivation) analysis revealed a rapid actin monomer increase on cell cortex deformation. Our direct visualization of the molecular behaviour reveals a mechanosensitive actin filament regeneration mechanism in which G-actin released by actin remodelling plays a pivotal role.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Fetal Proteins/metabolism , Mechanotransduction, Cellular/physiology , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Formins , Homeostasis , Humans , Immunoenzyme Techniques , Mice , Phosphorylation , Spectrometry, Fluorescence , Xenopus laevis
12.
J Signal Transduct ; 2012: 414913, 2012.
Article in English | MEDLINE | ID: mdl-22675626

ABSTRACT

Abl is a nonreceptor tyrosine kinase and plays an essential role in the modeling and remodeling of F-actin by transducing extracellular signals. Abl and its paralog, Arg, are unique among the tyrosine kinase family in that they contain an unusual extended C-terminal half consisting of multiple functional domains. This structural characteristic may underlie the role of Abl as a mediator of upstream signals to downstream signaling machineries involved in actin dynamics. Indeed, a group of SH3-containing accessory proteins, or adaptor proteins, have been identified that bind to a proline-rich domain of the C-terminal portion of Abl and modulate its kinase activity, substrate recognition, and intracellular localization. Moreover, the existence of signaling cascade and biological outcomes unique to each adaptor protein has been demonstrated. In this paper, we summarize functional roles and mechanisms of adaptor proteins in Abl-regulated actin dynamics, mainly focusing on a family of adaptor proteins, Abi. The mechanism of Abl's activation and downstream signaling mediated by Abi is described in comparison with those by another adaptor protein, Crk.

13.
J Biol Chem ; 287(21): 17493-17502, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22451653

ABSTRACT

Osteoclasts are multinucleated giant cells that reside in osseous tissues and resorb bone. Signaling mediated by receptor activator of nuclear factor (NF)-κB (RANK) and its ligand leads to the nuclear factor of activated T cells 2/c1 (NFAT2 or NFATc1) expression, a critical step in the formation of functional osteoclasts. In addition, adaptor proteins harboring immunoreceptor tyrosine-based activation motifs, such as DNAX-activating protein of 12 kDa (DAP12), play essential roles. In this study, we identified the gene encoding the lectin Siglec-15 as NFAT2-inducible, and we found that the protein product links RANK ligand-RANK-NFAT2 and DAP12 signaling in mouse osteoclasts. Both the recognition of sialylated glycans by the Siglec-15 V-set domain and the association with DAP12 through its Lys-272 are essential for its function. When Siglec-15 expression was knocked down, fewer multinucleated cells developed, and those that did were morphologically contracted with disordered actin-ring structures. These changes were accompanied by significantly reduced bone resorption. Siglec-15 formed complexes with Syk through DAP12 in response to vitronectin. Furthermore, chimeric molecules consisting of the extracellular and transmembrane regions of Siglec-15 with a K272A mutation and the cytoplasmic region of DAP12 significantly restored bone resorption in cells with knocked down Siglec-15 expression. Together, these results suggested that the Siglec-15-DAP12-Syk-signaling cascade plays a critical role in functional osteoclast formation.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Membrane Glycoproteins/biosynthesis , Osteoclasts/metabolism , Receptors, Immunologic/biosynthesis , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , CHO Cells , Cricetinae , Cricetulus , Gene Expression Regulation/physiology , Membrane Glycoproteins/genetics , Mice , Mutation, Missense , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , NIH 3T3 Cells , Osteoclasts/cytology , Protein Structure, Tertiary , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Immunologic/genetics , Sialic Acid Binding Ig-like Lectin 1
14.
Biochem J ; 441(3): 889-99, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22014333

ABSTRACT

Mena [mammalian Ena (Enabled)]/VASP (vasodilator-stimulated phosphoprotein) proteins are the homologues of Drosophila Ena. In Drosophila, Ena is a substrate of the tyrosine kinase DAbl (Drosophila Abl). However, the link between Abl and the Mena/VASP family is not fully understood in mammals. We previously reported that Abi-1 (Abl interactor 1) promotes phosphorylation of Mena and BCAP (B-cell adaptor for phosphoinositide 3-kinase) by bridging the interaction between c-Abl and the substrate. In the present study we have identified VASP, another member of the Mena/VASP family, as an Abi-1-bridged substrate of Abl. VASP is phosphorylated by Abl when Abi-1 is co-expressed. We also found that VASP interacted with Abi-1 both in vitro and in vivo. VASP was tyrosine-phosphorylated in Bcr-Abl-positive leukaemic cells in an Abi-1-dependent manner. Co-expression of c-Abl and Abi-1 or the phosphomimetic Y39D mutation in VASP resulted in less accumulation of VASP at focal adhesions. VASP Y39D had a reduced affinity to the proline-rich region of zyxin. Interestingly, overexpression of both phosphomimetic and unphosphorylated forms of VASP, but not wild-type VASP, impaired adhesion of K562 cells to fibronectin. These results suggest that the phosphorylation and dephosphorylation cycle of VASP by the Abi-1-bridged mechanism regulates association of VASP with focal adhesions, which may regulate adhesion of Bcr-Abl-transformed leukaemic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Focal Adhesions/metabolism , Leukemia, Experimental/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , CHO Cells , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Cytoskeletal Proteins , Focal Adhesions/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Fusion Proteins, bcr-abl/physiology , HEK293 Cells , Humans , K562 Cells , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Mice , Microfilament Proteins/genetics , NIH 3T3 Cells , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/physiology , Tyrosine/metabolism , Xenopus laevis
15.
FEBS Lett ; 585(6): 834-40, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21320496

ABSTRACT

Abi-1 is an adaptor protein for Abelson kinase (c-Abl), and Abi-1 promotes the Abl-mediated phosphorylation of Mammalian Enabled (Mena) by binding both c-Abl and Mena. Here, we identified a new phosphorylation site (Y398) in the SH3 domain of Abi-1, and disruption of Y398, combined with the previously identified phosphorylation site Y213, significantly weakens the binding of Abi-1 to c-Abl. The SH3 domain of Abi-1 and the proline-rich domain of c-Abl are involved in this interaction. Abi-1 phosphorylation at both sites stimulates the phosphorylation of Mena through the activation of c-Abl kinase. The phosphorylation of Abi-1 also plays a role in enhancing the adhesion of Bcr-Abl-transformed leukemic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Mutation , src Homology Domains/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Benzamides , Binding Sites/genetics , Blotting, Western , CHO Cells , Cell Adhesion , Cell Line , Cricetinae , Cricetulus , Cytoskeletal Proteins/metabolism , Fibronectins/metabolism , HEK293 Cells , Humans , Imatinib Mesylate , K562 Cells , Microfilament Proteins/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/analysis , Phosphotyrosine/metabolism , Piperazines/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Tandem Mass Spectrometry , Tyrosine/genetics , Tyrosine/metabolism
16.
FEBS Lett ; 580(27): 6464-70, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17101133

ABSTRACT

Abl interactor (Abi) was identified as an Abl tyrosine kinase-binding protein and subsequently shown to be a component of the macromolecular Abi/WAVE complex, which is a key regulator of Rac-dependent actin polymerization. Previous studies showed that Abi-1 promotes c-Abl-mediated phosphorylation of Mammalian Enabled (Mena) and WAVE2. In addition to Abi-1, mammals possess Abi-2 and NESH (Abi-3). In this study, we compared the three Abi proteins in terms of the promotion of c-Abl-mediated phosphorylation and the formation of Abi/WAVE complex. Although Abi-2, like Abi-1, promoted the c-Abl-mediated phosphorylation of Mena and WAVE2, NESH (Abi-3) had no such effect. This difference was likely due to their binding abilities as to c-Abl. Immunoprecipitation revealed that NESH (Abi-3) is present in the Abi/WAVE complex. Our results suggest that NESH (Abi-3), like Abi-1 and Abi-2, is a component of the Abi/WAVE complex, but likely plays a different role in the regulation of c-Abl.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Multiprotein Complexes/metabolism , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-abl/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation/physiology , Humans , Mice , Microfilament Proteins , Multiprotein Complexes/genetics , Phosphorylation , Proto-Oncogene Proteins c-abl/genetics , Wiskott-Aldrich Syndrome Protein Family/genetics
17.
Biochem Biophys Res Commun ; 350(1): 97-104, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-17005159

ABSTRACT

Osteoclast precursor cells (OPCs) have previously been established from bone marrow cells of SV40 temperature-sensitive T antigen-expressing transgenic mice. Here, we use retrovirus-mediated gene transfer to conditionally immortalize OPCs by expressing temperature-sensitive large T antigen (tsLT) from wild type bone marrow cells. The immortalized OPCs proliferated at the permissive temperature of 33.5 degrees C, but stopped growing at the non-permissive temperature of 39 degrees C. In the presence of receptor activator of NFkappaB ligand (RANKL), the OPCs differentiated into tartrate-resistant acid phosphatase (TRAP)-positive cells and formed multinucleate osteoclasts at 33.5 degrees C. From these OPCs, we cloned two types of cell lines. Both differentiated into TRAP-positive cells, but one formed multinucleate osteoclasts while the other remained unfused in the presence of RANKL. These results indicate that the established cell lines are useful for analyzing mechanisms of differentiation, particularly multinucleate osteoclast formation. Retrovirus-mediated conditional immortalization should be a useful method to immortalize OPCs from primary bone marrow cells.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , Retroviridae/genetics , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Survival , Mice , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/pharmacology , Stem Cells/drug effects , Temperature
18.
J Biotechnol ; 126(4): 463-74, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16766074

ABSTRACT

Conventional stable protein expression systems using mammalian cells include a time-consuming step of antibiotic resistance-based cell cloning. Here, we report a rapid flow cytometry-based method for the collection of retrovirus vector-infected Chinese hamster ovary (CHO) cells that express desired proteins. The vector carries the genes for green fluorescent protein (GFP), as a marker, and glutathione-S-transferase (GST), to express the desired protein as a GST-fusion construct. To render CHO cells susceptible to retrovirus infection, they were forced to express EcoR, the receptor for retroviruses. After infection, cells expressing desired proteins were collected by flow cytometry as a GFP-positive population, and the desired proteins were purified by glutathione affinity chromatography. This method reduces the time required between infection of cells and purification of a desired protein from several months to approximately 2 weeks.


Subject(s)
Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Receptors, Virus/metabolism , Animals , CHO Cells , Cell Culture Techniques , Cells, Cultured , Chromatography, Affinity , Cricetinae , Flow Cytometry , Gene Expression , Genetic Markers , Genetic Vectors , Glutathione Transferase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Time Factors
19.
FEBS Lett ; 579(14): 2986-90, 2005 Jun 06.
Article in English | MEDLINE | ID: mdl-15893754

ABSTRACT

In previous work we showed that Abl interactor 1 (Abi-1), by linking enzyme and substrate, promotes the phosphorylation of Mammalian Enabled (Mena) by c-Abl. To determine whether this mechanism extends to other c-Abl substrates, we used the yeast two-hybrid system to search for proteins that interact with Abi-1. By screening a human leukocyte cDNA library, we identified BCAP (B-cell adaptor for phosphoinositide 3-kinase) as another Abi-1-interacting protein. Binding experiments revealed that the SH3 domain of Abi-1 and the C-terminal polyproline structure of BCAP are involved in interactions between the two. In cultured cells, Abi-1 promoted phosphorylation of BCAP not only by c-Abl but also by v-Abl. The phosphorylation sites of BCAP by c-Abl were mapped to five tyrosine residues in the C-terminal region that are well conserved in mammals. These results show that Abi-1 promotes Abl-mediated BCAP phosphorylation and suggest that Abi-1 in general coordinates kinase-substrate interactions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Cell Line , Humans , Oncogene Proteins v-abl/metabolism , Phosphorylation , Phosphotyrosine/genetics , Phosphotyrosine/metabolism , Protein Binding , Proto-Oncogene Proteins c-abl/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...