Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Rep ; 25(1): 144-167, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177906

ABSTRACT

The tight junction (TJ) in epithelial cells is formed by integral membrane proteins and cytoplasmic scaffolding proteins. The former contains the claudin family proteins with four transmembrane segments, while the latter includes Par3, a PDZ domain-containing adaptor that organizes TJ formation. Here we show the single membrane-spanning protein TMEM25 localizes to TJs in epithelial cells and binds to Par3 via a PDZ-mediated interaction with its C-terminal cytoplasmic tail. TJ development during epithelial cell polarization is accelerated by depletion of TMEM25, and delayed by overexpression of TMEM25 but not by that of a C-terminally deleted protein, indicating a regulatory role of TMEM25. TMEM25 associates via its N-terminal extracellular domain with claudin-1 and claudin-2 to suppress their cis- and trans-oligomerizations, both of which participate in TJ strand formation. Furthermore, Par3 attenuates TMEM25-claudin association via binding to TMEM25, implying its ability to affect claudin oligomerization. Thus, the TJ protein TMEM25 appears to negatively regulate claudin assembly in TJ formation, which regulation is modulated by its interaction with Par3.


Subject(s)
Claudins , Tight Junctions , Tight Junctions/metabolism , Claudins/genetics , Claudins/metabolism , Carrier Proteins/metabolism , Epithelial Cells , Claudin-1/genetics , Claudin-1/metabolism
2.
Genes Cells ; 29(1): 63-72, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37985134

ABSTRACT

The hydrogen peroxide (H2 O2 )-producing NADPH oxidase Nox4, forming a heterodimer with p22phox , is expressed in a variety of cells including those in the heart to mediate adaptive responses to cellular stresses such as hypoxia. Since Nox4 is constitutively active, H2 O2 production is controlled by its protein abundance. Hypoxia-induced Nox4 expression is observed in various types of cells and generally thought to be regulated at the transcriptional level. Here we show that hypoxia upregulates the Nox4 protein level and Nox4-catalyzed H2 O2 production without increasing the Nox4 mRNA in rat H9c2 cardiomyocytes. In these cells, the Nox4 protein is stabilized under hypoxic conditions in a manner dependent on the presence of p22phox . Cell treatment with the proteasome inhibitor MG132 results in a marked decrease of the Nox4 protein under both normoxic and hypoxic conditions, indicating that the proteasome pathway does not play a major role in Nox4 degradation. The decrease is partially restored by the autophagy inhibitor 3-methyladenine. Furthermore, the Nox4 protein level is upregulated by the lysosome inhibitors bafilomycin A1 and chloroquine. Thus, in cardiomyocytes, Nox4 appears to be degraded via an autophagy-related pathway, and its suppression by hypoxia likely stabilizes Nox4, leading to upregulation of Nox4-catalyzed H2 O2 production.


Subject(s)
Myocytes, Cardiac , Oxidoreductases , Rats , Animals , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Hypoxia , Autophagy , Reactive Oxygen Species/metabolism
3.
J Biol Chem ; 298(10): 102475, 2022 10.
Article in English | MEDLINE | ID: mdl-36089063

ABSTRACT

The adhesion family of G protein-coupled receptors (GPCRs) is defined by an N-terminal large extracellular region that contains various adhesion-related domains and a highly-conserved GPCR-autoproteolysis-inducing (GAIN) domain, the latter of which is located immediately before a canonical seven-transmembrane domain. These receptors are expressed widely and involved in various functions including development, angiogenesis, synapse formation, and tumorigenesis. GPR125 (ADGRA3), an orphan adhesion GPCR, has been shown to modulate planar cell polarity in gastrulating zebrafish, but its biochemical properties and role in mammalian cells have remained largely unknown. Here, we show that human GPR125 likely undergoes cis-autoproteolysis when expressed in canine kidney epithelial MDCK cells and human embryonic kidney HEK293 cells. The cleavage appears to occur at an atypical GPCR proteolysis site within the GAIN domain during an early stage of receptor biosynthesis. The products, i.e., the N-terminal and C-terminal fragments, seem to remain associated after self-proteolysis, as observed in other adhesion GPCRs. Furthermore, in polarized MDCK cells, GPR125 is exclusively recruited to the basolateral domain of the plasma membrane. The recruitment likely requires the C-terminal PDZ-domain-binding motif of GPR125 and its interaction with the cell polarity protein Dlg1. Knockdown of GPR125 as well as that of Dlg1 results in formation of aberrant cysts with multiple lumens in Matrigel 3D culture of MDCK cells. Consistent with the multilumen phenotype, mitotic spindles are incorrectly oriented during cystogenesis in GPR125-KO MDCK cells. Thus, the basolateral protein GPR125, an autocleavable adhesion GPCR, appears to play a crucial role in apicobasal polarization in epithelial cells.


Subject(s)
Receptors, G-Protein-Coupled , Zebrafish , Animals , Dogs , Humans , Cell Adhesion , Cell Membrane/metabolism , Cell Polarity , Discs Large Homolog 1 Protein/metabolism , HEK293 Cells , Mammals/metabolism , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Zebrafish/metabolism , Cell Line , Gene Knockdown Techniques , Amino Acid Motifs
4.
J Biol Chem ; 297(6): 101354, 2021 12.
Article in English | MEDLINE | ID: mdl-34717957

ABSTRACT

Hepatocytes differ from columnar epithelial cells by their multipolar organization, which follows the initial formation of central lumen-sharing clusters of polarized cells as observed during liver development and regeneration. The molecular mechanism for hepatocyte polarity establishment, however, has been comparatively less studied than those for other epithelial cell types. Here, we show that the tight junction protein Par3 organizes hepatocyte polarization via cooperating with the small GTPase Cdc42 to target atypical protein kinase C (aPKC) to a cortical site near the center of cell-cell contacts. In 3D Matrigel culture of human hepatocytic HepG2 cells, which mimics a process of liver development and regeneration, depletion of Par3, Cdc42, or aPKC results in an impaired establishment of apicobasolateral polarity and a loss of subsequent apical lumen formation. The aPKC activity is also required for bile canalicular (apical) elongation in mouse primary hepatocytes. The lateral membrane-associated proteins Lgl1 and Lgl2, major substrates of aPKC, seem to be dispensable for hepatocyte polarity establishment because Lgl-depleted HepG2 cells are able to form a single apical lumen in 3D culture. On the other hand, Lgl depletion leads to lateral invasion of aPKC, and overexpression of Lgl1 or Lgl2 prevents apical lumen formation, indicating that they maintain proper lateral integrity. Thus, hepatocyte polarity establishment and apical lumen formation are organized by Par3, Cdc42, and aPKC; Par3 cooperates with Cdc42 to recruit aPKC, which plays a crucial role in apical membrane development and regulation of the lateral maintainer Lgl.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , Cell Cycle Proteins/analysis , Cytoskeletal Proteins/analysis , Hepatocytes/cytology , Isoenzymes/analysis , Protein Kinase C/analysis , cdc42 GTP-Binding Protein/analysis , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Polarity , Cells, Cultured , Cytoskeletal Proteins/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Isoenzymes/metabolism , Male , Mice , Mice, Inbred ICR , Protein Kinase C/metabolism , cdc42 GTP-Binding Protein/metabolism
5.
Genes Cells ; 23(6): 480-493, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29718541

ABSTRACT

Transmembrane glycoproteins, synthesized at the endoplasmic reticulum (ER), generally reach the Golgi apparatus in COPII-coated vesicles en route to the cell surface. Here, we show that the bona fide nonglycoprotein Nox5, a transmembrane superoxide-producing NADPH oxidase, is transported to the cell surface in a manner resistant to co-expression of Sar1 (H79G), a GTP-fixed mutant of the small GTPase Sar1, which blocks COPII vesicle fission from the ER. In contrast, Sar1 (H79G) effectively inhibits ER-to-Golgi transport of glycoproteins including the Nox5-related oxidase Nox2. The trafficking of Nox2, but not that of Nox5, is highly sensitive to over-expression of syntaxin 5 (Stx5), a t-SNARE required for COPII ER-to-Golgi transport. Thus, Nox2 and Nox5 mainly traffic via the Sar1/Stx5-dependent and -independent pathways, respectively. Both participate in Nox1 trafficking, as Nox1 advances to the cell surface in two differentially N-glycosylated forms, one complex and one high mannose, in a Sar1/Stx5-dependent and -independent manner, respectively. Nox2 and Nox5 also can use both pathways: a glycosylation-defective mutant Nox2 is weakly recruited to the plasma membrane in a less Sar1-dependent manner; N-glycosylated Nox5 mutants reach the cell surface in part as the complex form Sar1-dependently, albeit mainly as the high-mannose form in a Sar1-independent manner.


Subject(s)
Cell Membrane/metabolism , Monomeric GTP-Binding Proteins/metabolism , NADPH Oxidase 5/metabolism , Superoxides/metabolism , Amino Acid Sequence , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , HeLa Cells , Humans , Mutation , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Protein Transport , Sequence Homology
6.
Sci Rep ; 7(1): 17402, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234109

ABSTRACT

Interleukin (IL)-17-producing T helper (Th17) cells are crucial for host defense against extracellular microbes and pathogenesis of autoimmune diseases. Here we show that the AP-1 transcription factor JunB is required for Th17 cell development. Junb-deficient CD4+ T cells are able to develop in vitro into various helper T subsets except Th17. The RNA-seq transcriptome analysis reveals that JunB is crucial for the Th17-specific gene expression program. Junb-deficient mice are completely resistant to experimental autoimmune encephalomyelitis, a Th17-mediated inflammatory disease, and naive T helper cells from such mice fail to differentiate into Th17 cells. JunB appears to activate Th17 signature genes by forming a heterodimer with BATF, another AP-1 factor essential for Th17 differentiation. The mechanism whereby JunB controls Th17 cell development likely involves activation of the genes for the Th17 lineage-specifying orphan receptors RORγt and RORα and reduced expression of Foxp3, a transcription factor known to antagonize RORγt function.


Subject(s)
Cell Differentiation/physiology , Th17 Cells/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Transcription Factors/genetics
7.
J Biol Chem ; 291(39): 20739-52, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27489104

ABSTRACT

The nuclear protein IκBζ, comprising the N-terminal trans-activation domain and the C-terminal ankyrin repeat (ANK) domain composed of seven ANK motifs, activates transcription of a subset of nuclear factor-κB (NF-κB)-dependent innate immune genes such as Lcn2 encoding the antibacterial protein lipocalin-2. Lcn2 activation requires formation of a complex containing IκBζ and NF-κB p50, a transcription factor that harbors the DNA-binding Rel homology region but lacks a trans-activation domain, on the promoter with the canonical NF-κB-binding site (κB site) and its downstream cytosine-rich element. Here we show that IκBζ productively interacts with p50 via Asp-451 in the N terminus of ANK1, a residue that is evolutionarily conserved among IκBζ and the related nuclear IκB proteins Bcl-3 and IκBNS Threonine substitution for Asp-451 abrogates direct association with the κB-site-binding protein p50, complex formation with the Lcn2 promoter DNA, and activation of Lcn2 transcription. The basic residues Lys-717 and Lys-719 in the C-terminal region of ANK7 contribute to IκBζ binding to the Lcn2 promoter, probably via interaction with the cytosine-rich element required for Lcn2 activation; glutamate substitution for both lysines results in a loss of transcriptionally active complex formation without affecting direct contact of IκBζ with p50. Both termini of the ANK domain in Bcl-3 and IκBNS function in a manner similar to that of IκBζ to interact with promoter DNA, indicating a common mechanism in which the nuclear IκBs form a regulatory complex with NF-κB and promoter DNA via the invariant aspartate in ANK1 and the conserved basic residues in ANK7.


Subject(s)
I-kappa B Proteins/metabolism , Lipocalin-2/biosynthesis , NF-kappa B p50 Subunit/metabolism , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins/metabolism , Transcription, Genetic/physiology , Amino Acid Motifs , Animals , B-Cell Lymphoma 3 Protein , HEK293 Cells , Humans , I-kappa B Proteins/genetics , Lipocalin-2/genetics , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Genes Cells ; 19(8): 620-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24948478

ABSTRACT

The nuclear protein IκBζ activates transcription of a subset of NF-κB-dependent innate immune genes such as Lcn2 encoding the antibacterial protein lipocalin-2. IκBζ functions as a coactivator via its interaction with NF-κB p50, which contains a DNA-binding Rel-homology domain but lacks a transcriptional activation domain. However cis-regulatory elements involved in IκBζ function have remained unknown. Here, we show that, although IκBζ by itself is unable to associate with the Lcn2 promoter, IκBζ interacts with the promoter via p50 binding to the NF-κB-binding site (κB site) and the interaction also requires the pyrimidine-rich site (CCCCTC) that localizes seven bases downstream of the κB site. The pyrimidine-rich site is also essential for IκBζ-mediated activation of the Lcn2 gene. Introduction of both sites into an IκBζ-independent gene culminates in IκBζ-p50-DNA complex formation and transcriptional activation. Furthermore, spacing between the two sites is crucial for both IκBζ-DNA interaction and IκBζ-mediated gene activation. Thus, the pyrimidine-rich IκBζ-responsive site plays an essential role in productive interaction of IκBζ with the p50-DNA complex.


Subject(s)
Acute-Phase Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lipocalins/genetics , NF-kappa B p50 Subunit/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Response Elements , Acute-Phase Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , DNA/metabolism , Lipocalin-2 , Lipocalins/metabolism , Mice, Knockout , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Transcriptional Activation
9.
Biochim Biophys Acta ; 1760(9): 1364-72, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16860483

ABSTRACT

We found that a mouse homolog of human DNA polymerase delta interacting protein 38, referred to as Mitogenin I in this paper, and mitochondrial single-stranded DNA-binding protein (mtSSB), identified as upregulated genes in the heart of mice with juvenile visceral steatosis, play a role in the regulation of mitochondrial morphology. We demonstrated that overexpression of Mitogenin I or mtSSB increased elongated or fragmented mitochondria in mouse C2C12 myoblast cells, respectively. On the other hand, the silencing of Mitogenin I or mtSSB by RNA interference led to an increase in fragmented or elongated mitochondria in the cells, respectively, suggesting that Mitogenin I and mtSSB are involved in the processes of mitochondrial fusion and fission, respectively. In addition, we showed that the silencing of Mitogenin I resulted in an increase in the number of trypan blue-positive cells and the silencing of mtSSB resulted in an enhancement of the sensitivity of the cells to apoptotic stimulation by etoposide. The present results demonstrated that these proteins play a role in cell survival.


Subject(s)
DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Survival , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Silencing , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Sequence Data , Nuclear Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...