Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Int Immunol ; 35(7): 327-338, 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37052267

ABSTRACT

For cellular or tissue transplantation using induced pluripotent stem cells (iPSCs), from the viewpoint of time and economic cost, the use of allogeneic ones is being considered. Immune regulation is one of the key issues in successful allogeneic transplantation. To reduce the risk of rejection, several attempts have been reported to eliminate effects of the major histocompatibility complex (MHC) on the iPSC-derived grafts. On the other hand, we have shown that minor antigen-induced rejection is not negligible even when the MHC's impact is mitigated. In organ transplantation, it is known that donor-specific transfusion (DST) can specifically control immune responses to the donor. However, whether DST could control the immune response in iPSC-based transplantation was not clarified. In this study, using a mouse skin transplantation model, we demonstrate that infusion of donor splenocytes can promote allograft tolerance in the MHC-matched but minor antigen-mismatched conditions. When narrowing down the cell types, we found that infusion of isolated splenic B cells was sufficient to control rejection. As a mechanism, the administration of donor B cells induced unresponsiveness but not deletion in recipient T cells, suggesting that the tolerance was induced in the periphery. The donor B cell transfusion induced allogeneic iPSC engraftment. These results suggest for the first time a possibility that DST using donor B cells could induce tolerance against allogeneic iPSC-derived grafts.


Subject(s)
Induced Pluripotent Stem Cells , Transplantation Tolerance , Graft Survival , Immune Tolerance , Major Histocompatibility Complex , Adoptive Transfer , Graft Rejection
2.
J Biol Chem ; 298(2): 101579, 2022 02.
Article in English | MEDLINE | ID: mdl-35031323

ABSTRACT

Rho family small GTPases (Rho) regulate various cell motility processes by spatiotemporally controlling the actin cytoskeleton. Some Rho-specific guanine nucleotide exchange factors (RhoGEFs) are regulated via tyrosine phosphorylation by Src family tyrosine kinase (SFK). We also previously reported that PLEKHG2, a RhoGEF for the GTPases Rac1 and Cdc42, is tyrosine-phosphorylated by SRC. However, the details of the mechanisms by which SFK regulates RhoGEFs are not well understood. In this study, we found for the first time that PLEKHG1, which has very high homology to the Dbl and pleckstrin homology domains of PLEKHG2, activates Cdc42 following activation by FYN, a member of the SFK family. We also show that this activation of PLEKHG1 by FYN requires interaction between these two proteins and FYN-induced tyrosine phosphorylation of PLEKHG1. We also found that the region containing the Src homology 3 and Src homology 2 domains of FYN is required for this interaction. Finally, we demonstrated that tyrosine phosphorylation of Tyr-720 and Tyr-801 in PLEKHG1 is important for the activation of PLEKHG1. These results suggest that FYN is a regulator of PLEKHG1 and may regulate cell morphology through Rho signaling via the interaction with and tyrosine phosphorylation of PLEKHG1.


Subject(s)
Rho Guanine Nucleotide Exchange Factors , rho GTP-Binding Proteins , src-Family Kinases , Phosphorylation , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Tyrosine/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
J Cell Sci ; 133(15)2020 08 11.
Article in English | MEDLINE | ID: mdl-32661090

ABSTRACT

CENP-B binds to CENP-B boxes on centromeric satellite DNAs (known as alphoid DNA in humans). CENP-B maintains kinetochore function through interactions with CENP-A nucleosomes and CENP-C. CENP-B binding to transfected alphoid DNA can induce de novo CENP-A assembly, functional centromere and kinetochore formation, and subsequent human artificial chromosome (HAC) formation. Furthermore, CENP-B also facilitates H3K9 (histone H3 lysine 9) trimethylation on alphoid DNA, mediated by Suv39h1, at ectopic alphoid DNA integration sites. Excessive heterochromatin invasion into centromere chromatin suppresses CENP-A assembly. It is unclear how CENP-B controls such different chromatin states. Here, we show that the CENP-B acidic domain recruits histone chaperones and many chromatin modifiers, including the H3K36 methylase ASH1L, as well as the heterochromatin components Suv39h1 and HP1 (HP1α, ß and γ, also known as CBX5, CBX1 and CBX3, respectively). ASH1L facilitates the formation of open chromatin competent for CENP-A assembly on alphoid DNA. These results indicate that CENP-B is a nexus for histone modifiers that alternatively promote or suppress CENP-A assembly by mutually exclusive mechanisms. Besides the DNA-binding domain, the CENP-B acidic domain also facilitates CENP-A assembly de novo on transfected alphoid DNA. CENP-B therefore balances CENP-A assembly and heterochromatin formation on satellite DNA.


Subject(s)
Chromatin , Heterochromatin , Autoantigens/genetics , Centromere , Centromere Protein A/genetics , Chromatin/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Epigenesis, Genetic , Heterochromatin/genetics , Humans
4.
Mol Cell Biochem ; 459(1-2): 83-93, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31089935

ABSTRACT

It is well known that Rho family small GTPases (Rho GTPase) has a role of molecular switch in intracellular signal transduction. The switch cycle between GTP-bound and GDP-bound state of Rho GTPase regulates various cell responses such as gene transcription, cytoskeletal rearrangements, and vesicular trafficking. Rho GTPase-specific guanine nucleotide exchange factors (RhoGEFs) are regulated by various extracellular stimuli and activates Rho GTPase such as RhoA, Rac1, and Cdc42. The molecular mechanisms that regulate RhoGEFs are poorly understood. Our studies reveal that Dbl's big sister (DBS), a RhoGEF for Cdc42 and RhoA, is phosphorylated at least on tyrosine residues at 479, 660, 727, and 926 upon stimulation by SRC signaling and that the phosphorylation at Tyr-660 is particularly critical for the serum response factor (SRF)-dependent transcriptional activation of DBS by Ephrin type-B receptor 2 (EPHB2)/SRC signaling. In addition, our studies also reveal that the phosphorylation of Tyr-479 and Tyr-660 on DBS leads to the actin cytoskeletal reorganization by EPHB2/SRC signaling. These findings are thought to be useful for understanding pathological conditions related to DBS such as cancer and non-syndromic autism in future.


Subject(s)
Receptor, EphB2/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , HEK293 Cells , Humans , Receptor, EphB2/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , cdc42 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , src-Family Kinases/genetics
5.
Cell Signal ; 61: 93-107, 2019 09.
Article in English | MEDLINE | ID: mdl-31100317

ABSTRACT

The Rho family small GTPases mediate cell responses through actin cytoskeletal rearrangement. We previously reported that PLEKHG2, a Rho-specific guanine nucleotide exchange factor, is regulated via interaction with several proteins. We found that PLEKHG2 interacted with non-receptor tyrosine kinase ABL1, but the cellular function remains unclear. Here, we show that the interaction between PLEKHG2 and ABL1 attenuated the PLEKHG2-induced serum response element-dependent gene transcription in a tyrosine phosphorylation-independent manner. PLEKHG2 and ABL1 were co-localized and accumulated within cells co-expressing PLEKHG2 and ABL1. The cellular fractionation analysis suggested that the accumulation involved actin cytoskeletal reorganization. We also revealed that the co-expression of PLEKHG2 with ABL1, but not BCR-ABL, suppressed cell growth and synergistically enhanced NF-κB-dependent gene transcription. The cell growth suppression was canceled by co-expression with IκBα, a member of the NF-κB inhibitor protein family. This study suggests that the interaction between PLEKHG2 and ABL1 suppresses cell growth through intracellular protein accumulation via the NF-κB signaling pathway.


Subject(s)
Cell Proliferation/genetics , Guanine Nucleotide Exchange Factors/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Fusion Proteins, bcr-abl/metabolism , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation/genetics , Protein Aggregates/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-abl/genetics , Serum Response Element/genetics , Transcription, Genetic/genetics , Transfection
6.
Small GTPases ; 10(5): 361-366, 2019 09.
Article in English | MEDLINE | ID: mdl-28489964

ABSTRACT

PLEKHG2 is a Gßγ- and Gαs-dependent guanine nucleotide exchange factor for Rac1 and Cdc42 small GTPases and has been shown to mediate signaling pathways such as those for actin cytoskeletal reorganization and serum response element (SRE)-dependent gene transcription. We have shown that the four-and-a-half LIM domains (FHL) 1 acts as a positive regulator of PLEKHG2. Here, we evaluated the other FHL family members and found that the FHL1A specifically regulate the PLEKHG2 activity. Moreover, FHL1A further enhanced Gßγ- and PLEKHG2-induced SRE-dependent gene transcription, whereas FHL1A partially restored the attenuated PLEKHG2-induced SRE-dependent gene transcription by Gαs. Our results suggest that FHL1A specifically interacts with PLEKHG2 to regulate a function of PLEKHG2 that is modified by the interaction of Gßγ and Gαs.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Serum Response Element , Transcription, Genetic , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/genetics , Protein Domains , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Brain Res ; 1307: 53-62, 2010 Jan 11.
Article in English | MEDLINE | ID: mdl-19853587

ABSTRACT

Protein 4.1 (band 4.1 or 4.1R) was originally identified as an abundant protein of the human erythrocyte, in which it stabilizes the spectrin/actin cytoskeleton. Subsequently, several new family members, 4.1N, 4.1G and 4.1B, have been identified, which are expressed in many cell types, in particular at cell-cell junctions. We previously reported that 4.1R and 4.1N are expressed in the inner ear hair cells with specific localization patterns, and that 4.1R forms a complex with the membrane-associated guanylate kinase (MAGUK) protein p55 and two deafness gene products, myosin XV and whirlin. To determine the functions of the other family members, 4.1G and 4.1B, we observed their expression patterns in developing stereocilia in mice inner ear hair cells. 4.1G is expressed in the basal tapers of the stereocilia bundle in early postnatal stages. 4.1B was specifically and constantly expressed in the stereocilia tips during postnatal development. Additionally, we found that 4.1B is ablated in the hair cells of both myosin XV and whirlin mutant mice at all stages in hair cell development. These results suggest that 4.1 family members play important roles in the development and maintenance of the inner ear hair cells, and that 4.1B may be a member of the myosin XV-whirlin complex that is important for stereocilia maturation.


Subject(s)
Cytoskeletal Proteins/metabolism , Ear, Inner/cytology , Ear, Inner/growth & development , Gene Expression Regulation, Developmental/physiology , Hair Cells, Auditory, Inner/metabolism , Membrane Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Microfilament Proteins , Mutation/genetics , Myosins/genetics
8.
Methods Mol Biol ; 577: 25-39, 2009.
Article in English | MEDLINE | ID: mdl-19718506

ABSTRACT

Expression-ready cDNA clones, where the open reading frame (ORF) of the gene of interest is placed under the control of an appropriate promoter, are critical for functional characterization of the gene products. To create a resource of human gene products, we attempted to systematically convert original cDNA clones to expression-ready forms for recombinant proteins. For this purpose, we adopted a rare-cutting restriction enzyme-based system, the Flexi cloning system, to construct ORF clones. Taking advantage of the fully sequenced cDNA clones we accumulated to date, a number of sets of Flexi ORF clones in a 96-well format have been prepared. In this section, two methods for the preparation of Flexi ORF clones in a 96-well format are described. A protocol for transferring ORFs between Flexi vectors in a 96-well format is also described. We believe that the resultant clone set could be successfully used as a versatile reagent for functional characterization of human proteins.


Subject(s)
Cloning, Molecular/methods , High-Throughput Screening Assays/methods , Open Reading Frames , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , DNA Primers/genetics , Escherichia coli/genetics , Genetic Vectors , High-Throughput Screening Assays/instrumentation , Humans , Molecular Biology/instrumentation , Molecular Biology/methods , Polymerase Chain Reaction , Proteomics/instrumentation , Proteomics/methods
9.
Biochem Biophys Res Commun ; 388(4): 689-94, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19695231

ABSTRACT

The Krüppel-associated box-containing zinc finger gene family (KRAB-ZNF) is one of the largest gene families of transcriptional factors in the human genome. Although the functions of most of these genes remain to be determined, it is known that KRAB-mediated transcriptional repression requires a direct interaction with the KAP1 co-repressor. By mammalian one- or two-hybrid experiments in HEK293 cells, we compared transcriptional repression activities of 61 human KRAB-ZNFs. The results showed that six SCAN-KRAB-containing ZNFs are KAP1-independent transcriptional repressors whose SCAN-KRAB domain is unable to associate with KAP1 despite retaining transcriptional repression activity. Transcriptional repression activities of the SCAN-KRAB domain of KAP1-independent KRAB-ZNFs are not influenced by depletion of endogenous KAP1 levels by small interfering RNA. Although the mechanism by which KAP1-independent KRAB-ZNFs repress transcriptional activity remains to be elucidated, it appears that there may be a pathway for transcriptional repression that does not involve KAP1. These results provide new insight into the functions of the members of the KRAB-ZNF family.


Subject(s)
Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Transcription, Genetic , Zinc Fingers/genetics , Cell Line , Humans , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28
10.
Biochim Biophys Acta ; 1793(3): 506-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19321127

ABSTRACT

Protein 4.1B is a membrane skeletal protein expressed in various organs, and is associated with tumor suppressor in lung cancer-1 (TSLC1) in vitro. Although involvement of 4.1B in the intercellular junctions and tumor-suppression was suggested, some controversial results posed questions to the general tumor-suppressive function of 4.1B and its relation to TSLC1 in vivo. In this study, the expression of 4.1B and its interaction with TSLC1 were examined in rodent adrenal gland, and the involvement of 4.1B in tumorigenesis and the effect of 4.1B deficiency on TSLC1 distribution were also investigated using rodent pheochromocytoma and 4.1B-knockout mice. Although plasmalemmal immunolocalization of 4.1B was shown in chromaffin cells of rodent adrenal medulla, expression of 4.1B was maintained in developed pheochromocytoma, and morphological abnormality or pheochromocytoma generation could not be found in 4.1B-deficient mice. Furthermore, molecular interaction and colocalization of 4.1B and TSLC1 were observed in mouse adrenal gland, but the immunolocalization of TSLC1 along chromaffin cell membranes was not affected in the 4.1B-deficient mice. These results suggest that the function of 4.1B as tumor suppressor might significantly differ among organs and species, and that plasmalemmal retention of TSLC1 would be maintained by molecules other than 4.1B interacting in rodent chromaffin cells.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenal Medulla/metabolism , Immunoglobulins/metabolism , Membrane Proteins/metabolism , Pheochromocytoma/metabolism , Tumor Suppressor Proteins/metabolism , Adrenal Gland Neoplasms/pathology , Animals , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Immunoglobulins/analysis , In Situ Hybridization , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins , Microscopy, Electron , Pheochromocytoma/pathology , Tumor Suppressor Proteins/analysis
11.
Genes Cells ; 13(5): 483-95, 2008 May.
Article in English | MEDLINE | ID: mdl-18429820

ABSTRACT

Myosin VI is involved in a wide range of endocytic and exocytic membrane trafficking pathways; clathrin-mediated endocytosis, intracellular transport of clathrin-coated and -uncoated vesicles, AP-1B-dependent basolateral sorting in polarized epithelial cells and secretion from the Golgi complex to the cell surface. In this study, using a yeast two-hybrid screen, we identified brain-enriched kinase/lemur tyrosine kinase 2 (BREK/LMTK2), a transmembrane serine/threonine kinase with previously unknown cellular functions, as a myosin VI-interacting protein. Several binding experiments confirmed the interaction of myosin VI with BREK in vivo and in vitro. Immunocytochemical analyses revealed that BREK localizes to cytoplasmic membrane vesicles and to perinuclear recycling endosomes. Notably, cells in which BREK was depleted by siRNA were still able to internalize transferrin molecules and to transport them to early endosomes, but were unable to transport them to perinuclear recycling endosomes. Our results show that BREK is critical for the transition of endocytosed membrane vesicles from early endosomes to recycling endosomes and also suggest an involvement of myosin VI in this pathway.


Subject(s)
Endosomes/metabolism , Membrane Proteins/metabolism , Myosin Heavy Chains/metabolism , Protein Serine-Threonine Kinases/metabolism , Transport Vesicles/metabolism , Animals , Cell Line , Cytoplasm/chemistry , Cytoplasm/metabolism , Down-Regulation , Humans , Membrane Proteins/analysis , Membrane Proteins/genetics , Myosin Heavy Chains/chemistry , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Transferrin/metabolism , Two-Hybrid System Techniques
12.
DNA Res ; 15(3): 137-49, 2008 Jun 30.
Article in English | MEDLINE | ID: mdl-18316326

ABSTRACT

In this study, we established new systematic protocols for the preparation of cDNA clones, conventionally termed open reading frame (ORF) clones, suitable for characterization of their gene products by adopting a restriction-enzyme-assisted cloning method using the Flexi cloning system. The system has following advantages: (1) preparation of ORF clones and their transfer into other vectors can be achieved efficiently and at lower cost; (2) the system provides a seamless connection to the versatile HaloTag labeling system, in which a single fusion tag can be used for various proteomic analyses; and (3) the resultant ORF clones show higher expression levels both in vitro and in vivo. With this system, we prepared ORF clones encoding 1,929 human genes and characterized the HaloTag-fusion proteins of its subset that are expressed in vitro or in mammalian cells. Results thus obtained have demonstrated that our Flexi ORF clones are efficient for the production of HaloTag-fusion proteins that can provide a new versatile set for a variety of functional analyses of human genes.


Subject(s)
Cloning, Molecular/methods , Open Reading Frames/genetics , Proteome/analysis , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Models, Biological , Molecular Sequence Data , Protein Biosynthesis , Proteins/genetics , Proteins/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
J Histochem Cytochem ; 55(12): 1199-206, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17712176

ABSTRACT

Our recent studies demonstrated the localization of protein 4.1B, a member of the 4.1 skeletal membrane proteins, to the basolateral membranes of the S1-S2 renal proximal tubules. In the present studies, we investigated the presence of binding partners that could form a molecular complex with the 4.1B protein. Immunohistochemistry revealed the localization of p55, a membrane-associated guanylate kinase, and the sodium bicarbonate cotransporter1 (NBC1), to the basolateral membrane domain of S1-S2 in mouse renal proximal tubules. Using immunoprecipitation of kidney lysates with anti-p55 antibody, a positive band was blotted with anti-4.1B antibody. GST fusion proteins including the NBC1 and 4.1B regions were confirmed to bind with each other by electrophoresis after mixing. Both NBC1- and 4.1B-specific bands were detected in renal protein mixtures immunoprecipated by either anti-4.1B- or NBC1-specific antibodies. It is likely that NBC1, 4.1B, and p55 form a molecular complex in the basolateral membrane of the kidney S1-S2 proximal tubules. We propose that the 4.1B-containing membrane skeleton may play a role in regulating the Na(+) and HCO(3)(-) reabsorption in S1-S2 proximal tubules.


Subject(s)
Guanylate Kinases/metabolism , Kidney Tubules, Proximal/metabolism , Membrane Proteins/metabolism , Sodium-Bicarbonate Symporters/metabolism , Animals , Animals, Newborn , Immunohistochemistry , Immunoprecipitation , Kidney Tubules, Proximal/growth & development , Mice , Mice, Inbred C57BL , Microfilament Proteins , Microscopy, Immunoelectron
14.
J Neurosci Res ; 84(3): 568-77, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16752423

ABSTRACT

The membrane-associated cytoskeletal proteins, including protein 4.1 family, play important roles in membrane integrity, protein targeting, and signal transduction. Although protein 4.1G (4.1G) is expressed ubiquitously in mammalian tissues, it can have very discrete distributions within cells. The present study investigated the expression and distributions of 4.1G in rodent sciatic nerve. Northern and Western blot analysis detected abundant 4.1G mRNA and protein in rat sciatic nerve extracts. Immunohistochemical staining with a 4.1G-specific antibody and double immunolabeling with E-cadherin, betaIV spectrin, and connexin 32 detected 4.1G in paranodal loops, Schmidt-Lanterman incisures, and periaxonal, mesaxonal, and abaxonal membranes of rodent sciatic nerve. Immunoelectron microscopy confirmed the immunodistribution of 4.1G in Schwann cells. In developing mouse sciatic nerves, 4.1G was diffusely distributed in immature Schwann cells and gradually localized at paranodes, incisures, and periaxonal and mesaxonal membranes during their maturation. These data support the concept that 4.1G plays an important role in the membrane expansion and specialization that occurs during formation and maintenance of myelin internodes in the peripheral nervous system.


Subject(s)
Axons/metabolism , Cytoskeletal Proteins/metabolism , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , Schwann Cells/metabolism , Animals , Axons/ultrastructure , Cadherins/metabolism , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Connexins/metabolism , Cytoskeletal Proteins/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/metabolism , Peripheral Nerves/ultrastructure , RNA, Messenger/metabolism , Rats , Rats, Wistar , Schwann Cells/ultrastructure , Spectrin/metabolism , Gap Junction beta-1 Protein
15.
Genome Res ; 16(1): 132-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16344567

ABSTRACT

We developed a new microscopic platform for the real-time analysis of molecular interactions by combining microbead-tagging techniques with total internal reflection fluorescent microscopy (TIRFM). The optical manipulation of probe microbeads, followed by photo immobilization on a solid surface, enabled us to generate arrays with extremely high density (>100 microbeads in a 25 microm x 25 microm area), and TIRFM made it possible to monitor the binding reactions of fluorescently labeled targets onto probe microbeads without removal of free targets. We demonstrated the high performance of this platform through analyses of interactions between antigen and antibody and between small compounds and proteins. Then, recombinant protein levels in total cellular lysates of Escherichia coli were quantified from the association kinetics using antibody-immobilized microbead arrays, which served as a model for a protein-profiling array. Furthermore, in combination with in vitro synthesis-coupled protein labeling, we could kinematically analyze the interaction of nuclear factor kappaB (p50) with DNA. These results demonstrated that this platform enabled us to: (1) monitor binding processes of fluorescently labeled targets to multiple probes in real-time without removal of free targets, (2) determine concentrations of free targets only from the association kinetics at an early phase, and (3) greatly reduce the required volume of the target solution, in principle to subnanoliter, for molecular interaction analysis. The unique features of this microbead-based microarray system open the way to explore molecular interactions with a wide range of affinities in extremely small volumes of target solutions, such as extracts from single cells.


Subject(s)
Biosensing Techniques , Microspheres , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Biosensing Techniques/methods , Microscopy, Fluorescence/methods , Oligonucleotide Array Sequence Analysis/methods , Protein Array Analysis/methods , Protein Binding , Sensitivity and Specificity
16.
Histochem Cell Biol ; 124(6): 477-86, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16184385

ABSTRACT

Although it was reported that protein 4.1 G, a cytoskeletal protein characterized by its general expression in the body, interacts with some signal transduction molecules in the central nervous system (CNS), its distribution and significance in vivo remained to be elucidated. In the present study, we have identified 4.1 G-positive cells in the rodent CNS, and demonstrated its immunolocalization in the developing mouse CNS. In the rodent CNS, 4.1 G was colocalized with markers for microglia, such as CD45, OX-42 and ionized calcium-binding adapter molecule 1 (Iba1), but not with markers for neuronal or other glial cells. Additionally, colocalization of 4.1 G and A1 adenosine receptor was observed in the mouse cerebrum. In a mixed glial culture, most OX-42-positive microglia were positive for 4.1 G, and 4.1 G isoforms of the same molecular weight as in the rat brain were expressed in cultured microglia, where 4.1 G mRNA was detected by RT-PCR. In the developing mouse cerebral cortex, 4.1 G was detected in immature microglia, which were positive for Iba1. These results indicate that 4.1 G in the CNS is mainly distributed in microglia in vivo. Considering the interactions between 4.1 G and the signal transduction molecules, putative roles have been proposed for 4.1 G in microglial functions in the CNS.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Microglia/cytology , Animals , Animals, Newborn , Cells, Cultured , Central Nervous System/cytology , Cerebral Cortex/cytology , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microglia/metabolism , Protein Isoforms/biosynthesis , Rats , Rats, Wistar , Sensitivity and Specificity , Signal Transduction/physiology
17.
Histochem Cell Biol ; 124(3-4): 303-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16041627

ABSTRACT

Protein 4.1 families have recently been established as potential organizers of an adherens system. In the adult mouse testis, protein 4.1G (4.1G) localized as a line pattern in both basal and adluminal compartments of the seminiferous tubules, attaching regions of germ cells and Sertoli cells. By double staining for 4.1G and F-actin, their localizations were shown to be different, indicating that 4.1G was localized in a region other than the basal and apical ectoplasmic specializations, which formed the Sertoli-Sertoli cell junction and Sertoli-spermatid junction, respectively. By electron microscopy, immunoreactive products were seen exclusively on the cell membranes of Sertoli cells, attaching to the various differentiating germ cells. The immunolocalization of cadherin was identical to that of 4.1G, supporting the idea that 4.1G may be functionally interconnected with adhesion molecules. In an experimental mouse model of cadmium treatment, in which tight and adherens junctions of seminiferous tubules were disrupted, the 4.1G immunostaining in the seminiferous tubules was dramatically decreased. These results indicate that 4.1G may have a basic adhesive function between Sertoli cells and germ cells from the side of Sertoli cells.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Seminiferous Tubules/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Seminiferous Tubules/ultrastructure , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure
18.
Anat Sci Int ; 80(2): 61-70, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15960311

ABSTRACT

The polarized architecture of epithelial cells is a fundamental determinant of cell structures and functions. Both formation and orientation of proper epithelial polarity are needed for cell-cell or cell-matrix adhesion, signal transduction and cytoskeletal interactions of multimolecular complexes at apical, lateral and basal cell membranes. These cell membrane domains are usually segregated by some junctional complexes. Recent molecular genetic studies on the anchor structure between myelin sheaths and axons have indicated the specific molecular organization for polarization of axolemma and the myelin sheaths at paranodes, termed 'septate-like junctions'. It was also speculated that other mammalian organs may use a similar junctional system. The protein 4.1 B was originally found to be localized in paranodes and juxtaparanodes of myelinated nerve fibers. Our recent immunohistochemical studies on protein 4.1B have indicated its significance for the cell-cell and/or cell-matrix adhesion in various rodent organs. The protein 4.1 family of proteins have been supposed to possess variable molecular domains relating to cell adhesion, ion balance, receptor responses and signal transduction. Therefore, more precise studies on the molecular structure and the functional domains of protein 4.1B, as well as on its changes under physiological and pathological conditions, may provide a clue for organogenesis in various mammalian organs.


Subject(s)
Cell Membrane Structures/metabolism , Epithelial Cells/metabolism , Membrane Proteins/metabolism , Viscera/metabolism , Animals , Cell Adhesion/physiology , Cell Membrane Structures/ultrastructure , Cell Polarity/physiology , Epithelial Cells/ultrastructure , Humans , Microfilament Proteins , Organogenesis/physiology , Viscera/ultrastructure
19.
Biochemistry ; 44(16): 6190-6, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15835906

ABSTRACT

Myosin XVIII is the recently identified 18th class of myosins, and its members are composed of a unique N-terminal domain, a motor domain with an unusual sequence around the ATPase site, one IQ motif, a segmented coiled-coil region for dimerization, and a C-terminal globular tail. To gain insight into the functions of this unique myosin, we characterized its human homologue, MYO18A, focusing on the functional roles of the characteristic N-terminal domain that contains a PDZ module known to mediate protein-protein interaction. GFP-tagged full-length and C-terminally truncated MYO18A molecules that were expressed in HeLa cells exhibited colocalization with actin filaments. Chemical cross-linking of these molecules showed that they form stable dimers as expected from their putative coiled-coil tails. Cosedimentation of the various types of truncated MYO18A constructs with actin filaments indicated the presence of an ATP-insensitive actin-binding site in the N-terminal domain. Further studies on truncated constructs of the N-terminal domain indicated that this actin-binding site is located outside the PDZ module, but within the middle region of this domain, which does not show any homology with the known actin-binding motifs. These results imply that this dimeric myosin might stably cross-link actin filaments by two ATP-insensitive actin-binding sites at the N-terminal domains for higher-order organization of the actin cytoskeleton.


Subject(s)
Actins/metabolism , Myosins/chemistry , Myosins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , DNA/genetics , Dimerization , HeLa Cells , Humans , In Vitro Techniques , Molecular Sequence Data , Myosins/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Subcellular Fractions/metabolism
20.
DNA Res ; 12(4): 257-67, 2005.
Article in English | MEDLINE | ID: mdl-16769688

ABSTRACT

Although we have so far identified and sequenced >2000 human long cDNAs, known as KIAA cDNAs, half of them have yet to be functionally annotated. Expression-ready cDNA clones derived from these genes, where the open reading frame (ORF) of the gene of interest is placed under the control of an appropriate promoter, are critical for functional characterization of these gene products. In this study, we attempted to systematically convert original cDNA clones to expression-ready forms for native and fusion proteins. For this purpose, we developed a new method for ORF cloning based on a homologous recombination in Escherichia coli to avoid laborious manipulations and artificial introduction of mutations in ORF. Using 1589 putative full-length ORFs (from 1002 KIAA genes, 119 human known genes and 468 mouse genes) with an average size of 2.8 kb, we successfully prepared expression plasmids for 1463 native proteins and for 1343 fusion proteins by this method. The resultant expression-ready clones were examined using an in vitro transcription/translation system followed by SDS-polyacrylamide gel electrophoresis and by transient expression of GFP-fusion proteins in human embryonic kidney (HEK) 293 cells. This set of expression-ready clones of long cDNAs encoding large proteins would open a new route to experimentally analyze their functions on a proteomic scale, since unavailability of expression-ready clones for mammalian large proteins has been a major obstacle to the functional analysis of these cDNAs.


Subject(s)
DNA, Complementary/metabolism , Open Reading Frames , Recombinant Fusion Proteins/metabolism , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/genetics , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...