Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Biochem Biophys Res Commun ; 628: 91-97, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36084556

ABSTRACT

The adaptor protein GAREM has two subtypes. Each is involved in Erk activation signaling downstream of the cell growth factor receptor in cultured cells. Regarding their role in individual animals, we have previously reported that mice deficient in GAREM2, which is highly expressed in the brain, exhibit emotional changes. In this paper, we report an amino acid substitution mutation (K291R) in GAREM1, in a patient with idiopathic short stature, which indicates that the mutant exhibits dominant-negative properties. The GAREM K291R mutant did not promote Erk activation in EGF-stimulated cultured cells. Similar features were also observed in cells in which GAREM1 expression was suppressed by genome editing; along with Erk, phosphorylation of S6 kinase and 4EBP1, whose activation is necessary for cell proliferation and biological growth, were inhibited Furthermore, we generated mice deficient in GAREM1 and showed that the mutant mice are lighter in weight. Overall, the results of this paper suggest that GAREM1 is required for normal growth and for maintaing average body size in humans and mice.


Subject(s)
Body Weight , Dwarfism , GRB2 Adaptor Protein , Adaptor Proteins, Signal Transducing , Animals , Body Weight/genetics , Cell Cycle Proteins , Cell Line , Dwarfism/genetics , Epidermal Growth Factor/metabolism , GRB2 Adaptor Protein/metabolism , Humans , MAP Kinase Signaling System , Mice , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism
2.
Cell Mol Biol Lett ; 26(1): 16, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33931009

ABSTRACT

BACKGROUND: In mammals, there are two subtypes of Grb2-associated regulator of Erk/MAPK (GAREM), an adaptor protein that functions downstream of the cell growth factor receptor. GAREM1 is ubiquitously expressed, whereas GAREM2 is mainly expressed in the brain. However, the precise mechanism of the translocation of each GAREM subtype in growth factor-stimulated cells is still unclear. METHODS: In this study, immunofluorescence staining with specific antibodies against each GAREM subtype and time-lapse analysis using GFP fusion proteins were used to analyze the subcellular localization of each GAREM subtype in a cell growth stimulus-dependent manner. We also biochemically analyzed the correlation between its subcellular localization and tyrosine phosphorylation of GAREM2. RESULTS: We found that endogenously and exogenously expressed GAREM2 specifically aggregated and formed granules in NGF-stimulated PC-12 cells and in EGF-stimulated COS-7 cells. Based on the observed subcellular localizations of chimeric GAREM1 and GAREM2 proteins, a glycine-rich region, which is present only in GAREM2, is required for the observed granule formation. This region also regulates the degree of EGF-stimulation-dependent tyrosine phosphorylation of GAREM2. CONCLUSIONS: Our results, showing that aggregation of GAREM2 in response to EGF stimulation is dependent on a glycine-rich region, suggest that GAREM2 aggregation may be involved in neurodegenerative diseases.


Subject(s)
Epidermal Growth Factor/pharmacology , Protein Aggregates/drug effects , Animals , Apoptosis/drug effects , COS Cells , Chlorocebus aethiops , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Phosphorylation/drug effects , Protein Domains/genetics , Time-Lapse Imaging
3.
Mol Brain ; 12(1): 94, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31718706

ABSTRACT

Grb2-associated regulator of Erk/MAPK (GAREM), is an adaptor protein related to the several cell growth factor receptor-signaling. The GAREM family has two subtypes, GAREM1 and GAREM2, both encoded in the human and mouse genome. Recent genome-wide research identified GAREM2 as a candidate of neurodegenerative diseases. Here, we use knockout (KO) mice to show the role of GAREM2, that is highly expressed in the brain. According to the comprehensive behavioral battery, they exhibited less anxiety both in elevated plus maze and open field tests, mildly increased social approaching behavior in the reciprocal social interaction test, and longer latency to immobility in the tail suspension test as compared to wild-type (WT). Additionally, the extension of neurites in the primary cultured neurons was suppressed in ones derived from GAREM2 KO mice. Furthermore, we also identified Intersectin, as a binding partner of GAREM2 in this study. Intersectin is also a multi-domain adaptor protein that regulates endocytosis and cell signaling, which can potentially alter the subcellular localization of GAREM2. The important molecules, such as the neurotrophin receptor and Erk family, that are involved in the signaling pathway of the neural cell growth in the mouse brain, have been reported to participate in emotional behavior. As GAREM plays a role in the cellular growth factor receptor signaling pathway, GAREM2 may have a common role related to the transduction of Erk signaling in the higher brain functions.


Subject(s)
Behavior, Animal , Brain/metabolism , GRB2 Adaptor Protein/deficiency , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Anxiety/pathology , Cell Line , Exploratory Behavior , Female , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Maze Learning , Mice, Knockout , Neuronal Outgrowth , Neurons/metabolism , Reaction Time , Social Behavior
4.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 285-295, 2019 02.
Article in English | MEDLINE | ID: mdl-30458214

ABSTRACT

WDR54 is a member of the WD40 repeat (WDR) domain-containing protein family that was recently identified as a novel oncogene in colorectal cancer. However, the molecular mechanism of WDR54 and its functional association with other molecules related to tumor cell growth are unknown. Here, we show that WDR54 can be cross-linked by the action of transglutaminase (TG) 2, which enhances the activation of EGF receptor-mediated signaling pathway. The most carboxyl-terminal WD domain was required for cross-linking. In addition, lysine 280 in WDR54, also in this WD domain, was an important residue for both cross-linking and ubiquitination. Cross-linked WDR54 was found in vesicles aggregated at the plasma membrane. The activated EGF receptor was co-localized with this vesicle, and the internalization of the EGF receptor into the cytosol was sustained. As a result, Erk activity in response to EGF stimulation was enhanced. Furthermore, the growth of the cells lacking WDR54 expression generated by genome editing was delayed compared with that in wild-type cells. Because TG2 is also has been proposed to activate the EGF receptor-signaling and proliferation of tumor cells, WDR54 might have a functional relationship with the EGF receptor and TG2. Our study on the mechanism of biological function of WDR54 may provide rationale for the design and development of a cancer drug based on inhibiting the post-translational modification of this oncogene product.


Subject(s)
Arabidopsis Proteins/metabolism , GTP-Binding Proteins/metabolism , Transglutaminases/metabolism , Animals , Arabidopsis Proteins/physiology , COS Cells , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/physiology , Chlorocebus aethiops , ErbB Receptors/metabolism , GTP-Binding Proteins/physiology , HEK293 Cells , Humans , Phosphorylation/physiology , Protein Binding , Protein Glutamine gamma Glutamyltransferase 2 , Protein Processing, Post-Translational/physiology , Signal Transduction/physiology , Transglutaminases/genetics , Transglutaminases/physiology , Ubiquitination
5.
Biochem Biophys Res Commun ; 495(1): 1249-1256, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180010

ABSTRACT

Cardiolipin and phosphatidic acid-binding protein (CLPABP) controls the stability of the mRNA harboring an AU-rich element (ARE) in the 3' UTR with the help of the RNA stabilizer, human antigen R (HuR). Although CLPABP is localized on the mitochondrial surface as a large protein-RNA complex, its precise role is not yet known. Recently, CLPABP was identified as an N-myristoylated protein. Here, we demonstrate the effects of N-myristoylation on the functions of CLPABP. In the present study, compared to the wild-type protein that possessed the "MG" motif at the N-terminus for N-myristoylation, the mutant CLPABP protein that lacked N-myristoylation modification site was unstable. Furthermore, the expression of the G/A mutant of CLPABP, which lacked N-myristoylation site, induced morphological alterations in mitochondria. Because pleckstrin homology domain-deleted mutant, which was fused with the N-myristoylation site derived from intact CLPABP, could not colocalize with mitochondria, N-myristoylation of CLPABP was predicted to affect its stability onto the mitochondrial membrane rather than its subcellular localization.


Subject(s)
Lipid Metabolism/physiology , Lipid-Linked Proteins/metabolism , Myristic Acid/metabolism , Protein Prenylation/physiology , Subcellular Fractions/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans
6.
Neurosci Res ; 134: 18-29, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29203264

ABSTRACT

During embryonic development, GABAergic interneurons, a main inhibitory component in the cerebral cortex, migrate tangentially from the ganglionic eminence (GE) to cerebral cortex. After reaching the cerebral cortex, they start to extend their neurites for constructing local neuronal circuits around the neonatal stage. Aberrations in migration or neurite outgrowth are implicated in neurological and psychiatric disorders such as epilepsy, schizophrenia and autism. Previous studies revealed that in the early phase of cortical development the neural population migrates tangentially from the GE in the telencephalon and several genes have been characterized as regulators of migration and specification of GABAergic interneurons. However, much less is known about the molecular mechanisms of GABAergic interneurons-specific maturation at later stages of development. Here, we performed genome-wide screening to identify genes related to the later stage by flow cytometry based-microarray (FACS-array) and identified 247 genes expressed in cortical GABAergic interneurons. Among them, Dgkg, a member of diacylglycerol kinase family, was further analyzed. Correlational analysis revealed that Dgkg is dominantly expressed in somatostatin (SST)-expressing GABAergic interneurons. The functional study of Dgkg using GE neurons indicated alteration in neurite outgrowth of GABAergic neurons. This study shows a new functional role for Dgkg in GABAergic interneurons as well as the identification of other candidate genes for their maturation.


Subject(s)
GABAergic Neurons/physiology , Gene Expression Regulation/physiology , Glutamate Decarboxylase/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Computational Biology , Embryo, Mammalian , Female , Flow Cytometry , Frizzled Receptors/metabolism , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Somatostatin/metabolism , Transfection
7.
J Hum Genet ; 63(3): 297-307, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29273731

ABSTRACT

PR interval is the period from the onset of P wave to the start of the QRS complex on electrocardiograms. A recent genomewide association study (GWAS) suggested that GAREM1 was linked to the PR interval on electrocardiograms. This study was designed to validate this correlation using additional subjects and examined the function of Garem1 in a mouse model. We analyzed the association of rs17744182, a variant in the GAREM1 locus, with the PR interval in 5646 subjects who were recruited from 2 Korean replication sets, Yangpyeong (n = 2471) and Yonsei (n = 3175), and noted a significant genomewide association by meta-analysis (P = 2.39 × 10-8). To confirm the function of Garem1 in mice, Garem1 siRNA was injected into mouse tail veins to reduce the expression of Garem1. Garem1 transcript levels declined by 53% in the atrium of the heart (P = 0.029), and Garem1-siRNA injected mice experienced a significant decrease in PR interval (43.27 ms vs. 44.89 ms in control, P = 0.007). We analyzed the expression pattern of Garem1 in the heart by immunohistology and observed specific expression of Garem1 in intracardiac ganglia. Garem1 was expressed in most neurons of the ganglion, including cholinergic and adrenergic cells. We have provided evidence that GAREM1 is involved in the PR interval of ECGs. These findings increase our understanding of the regulatory signals of heart rhythm through intracardiac ganglia of the autonomic nervous system and can be used to guide the development of a therapeutic target for heart conditions, such as atrial fibrillation.


Subject(s)
Electrocardiography , GRB2 Adaptor Protein/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Heart Conduction System , Adult , Aged , Alleles , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Cell Line , Disease Models, Animal , Female , GRB2 Adaptor Protein/metabolism , Gene Expression , Gene Silencing , Genetic Variation , Genotype , Heart Atria/cytology , Heart Atria/metabolism , Heart Atria/physiopathology , Humans , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide , RNA, Small Interfering/genetics
8.
Biochim Biophys Acta ; 1861(11): 1816-1827, 2016 11.
Article in English | MEDLINE | ID: mdl-27616329

ABSTRACT

Cardiolipin and phosphatidic acid-binding protein (CLPABP) is a pleckstrin homology domain-containing protein and is localized on the surface of mitochondria of cultured cells as a large protein-RNA complex. To analyze the physiological functions of CLPABP, we established and characterized a CLPABP knockout (KO) mouse. Although expression levels of CLPABP transcripts in the developmental organs were high, CLPABP KO mice were normal at birth and grew normally when young. However, old male mice presented a fatty phenotype, similar to that seen in metabolic syndrome, in parallel with elevated male- and age-dependent CLPABP gene expression. One of the reasons for this obesity in CLPABP KO mice is dependence on increases in leptin concentration in plasma. The leptin transcripts were also upregulated in the adipose tissue of KO mice compared with wild-type (WT) mice. To understand the difference in levels of the transcriptional product, we focused on the effect of CLPABP on the stability of mRNA involving an AU-rich element (ARE) in its 3'UTR dependence on the RNA stabilizer, human antigen R (HuR), which is one of the CLPABP-binding proteins. Increase in stability of ARE-containing mRNAs of leptin by HuR was antagonized by the expression of CLPABP in cultured cells. Depletion of CLPABP disturbed the normal subcellular localization of HuR to stress granules, and overexpression of CLPABP induced instability of leptin mRNA by inhibiting HuR function. Consequently, leptin levels in old male mice might be regulated by CLPABP expression, which might lead to body weight control.


Subject(s)
AU Rich Elements/genetics , Aging/genetics , ELAV Proteins/metabolism , Leptin/genetics , Lipid-Linked Proteins/metabolism , Obesity/genetics , RNA Stability/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Gene Deletion , Gene Expression Regulation , Leptin/metabolism , Lipid-Linked Proteins/genetics , Male , Metabolome , Mice, Knockout , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Subcellular Fractions/metabolism , Transcription, Genetic
9.
Biochem Biophys Res Commun ; 469(3): 377-83, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26657846

ABSTRACT

The CLOCK-interacting protein, Circadian (CIPC), has been identified as an additional negative-feedback regulator of the circadian clock. However, recent study on CIPC knockout mice has shown that CIPC is not critically required for basic circadian clock function, suggesting other unknown biological roles for CIPC. In this study, we focused on the cell cycle dependent nuclear-cytoplasmic shuttling function of CIPC and on identifying its binding proteins. Lys186 and 187 were identified as the essential amino acid residues within the nuclear localization signal (NLS) of CIPC. We identified CIPC-binding proteins such as the multifunctional enzyme CAD protein (carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotase), which is a key enzyme for de novo pyrimidine synthesis. Compared to control cells, HEK293 cells overexpressing wild-type CIPC showed suppressed cell proliferation and retardation of cell cycle. We also found that PMA-induced Erk activation was inhibited with expression of wild-type CIPC. In contrast, the NLS mutant of CIPC, which reduced the ability of CIPC to translocate into the nucleus, did not exhibit these biological effects. Since CAD and Erk have significant roles in cell proliferation and cell cycle, CIPC may work as a cell cycle regulator by interacting with these binding proteins.


Subject(s)
Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Carrier Proteins/metabolism , Circadian Rhythm/physiology , Dihydroorotase/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation , Feedback, Physiological/physiology , HEK293 Cells , HeLa Cells , Humans
10.
Biochem Biophys Res Commun ; 464(2): 616-21, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26164232

ABSTRACT

GAREM1 (Grb2-associated regulator of Erk/MAPK1) is an adaptor protein that is involved in the epidermal growth factor (EGF) pathway. The nuclear localization of GAREM1 depends on the nuclear localization sequence (NLS), which is located at the N-terminal CABIT (cysteine-containing, all in Themis) domain. Here, we identified 14-3-3ε as a GAREM-binding protein, and its binding site is closely located to the NLS. This 14-3-3 binding site was of the atypical type and independent of GAREM phosphorylation. Moreover, the binding of 14-3-3 had an effect on the nuclear localization of GAREM1. Unexpectedly, we observed that the CABIT domain had intramolecular association with the C-terminal SAM (sterile alpha motif) domain. This association might be inhibited by binding of 14-3-3 at the CABIT domain. Our results demonstrate that the mechanism underlying the nuclear localization of GAREM1 depends on its NLS in the CABIT domain, which is controlled by the binding of 14-3-3 and the C-terminal SAM domain. We suggest that the interplay between 14-3-3, SAM domain and CABIT domain might be responsible for the distribution of GAREM1 in mammalian cells.


Subject(s)
14-3-3 Proteins/metabolism , GRB2 Adaptor Protein/metabolism , Protein Structure, Tertiary , Subcellular Fractions/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , GRB2 Adaptor Protein/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL