Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Article in English | MEDLINE | ID: mdl-38246006

ABSTRACT

Thiopurine is metabolized to 6-thio-(deoxy) guanosine triphosphate (6-thio-(d) GTP), which is then incorporated into DNA or RNA and causes cytotoxicity. Nudix hydrolase 15 (NUDT15) reduces the cytotoxic effects of thiopurine by converting 6-thio-(d) GTP to 6-thio-(d) guanosine monophosphate (6-thio-(d) GMP). NUDT15 polymorphisms like the Arg139Cys variant are strongly linked to thiopurine-induced severe leukocytopenia and alopecia. Therefore, measurement of NUDT15 enzymatic activity in individual patients can help predict thiopurine tolerability and adjust the dosage. We aimed to develop a quantitative assay for NUDT15 enzymatic activity in human blood samples. Blood samples were collected from donors whose NUDT15 genetic status was determined. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assess the 6-thio-GTP metabolic activity in cell extracts. Because 6-thio-guanosine diphosphate (6-thio-GDP) and 6-thio-GMP were generated upon incubation of 6-thio-GTP with human blood cell extracts, the method detecting 6-thio-GTP, 6-thio-GDP, and 6-thio-GMP was validated. All three metabolites were linearly detected, and the lower limit of quantification (LLOQ) of 6-thio-GTP, 6-thio-GDP, and 6-thio-GMP were 5 µM, 1 µM, and 2 µM, respectively. Matrix effects of human blood cell extracts to detect 6-thio-GTP, 6-thio-GDP, and 6-thio-GMP were 99.0 %, 100.5 %, and 101.4 %, respectively, relative to the signals in the absence of blood cell extracts. The accuracy and precision of the method and the stability of the samples were also assessed. Using this established method, the genotype-dependent differences in NUDT15 activities were successfully determined using cell extracts derived from human blood cells with NUDT15 wild-type (WT) or Arg139Cys variant and 6-thio-GTP (100 µM) as a substrate (18.1, 14.9, and 6.43 µM/h/106 cells for WT, Arg139Cys heterozygous, and homozygous variant, respectively). We developed a method for quantifying intracellular NUDT15 activity in peripheral blood mononuclear cells (PBMCs), which we defined as the conversion of 6-thio-GTP to 6-thio-GMP. Although PBMCs preparation takes some time, its reproducibility in experiments makes it a promising candidate for clinical application. This method can tell the difference between WT and Arg139Cys homozygous blood samples. Even in patients with WT NUDT15, WT samples showed variations in NUDT15 activity, which may correlate with variations in thiopurine dosage.


Subject(s)
Leukocytes, Mononuclear , Nudix Hydrolases , Purines , Sulfhydryl Compounds , Humans , Chromatography, Liquid , Cell Extracts , Leukocytes, Mononuclear/metabolism , Reproducibility of Results , Pyrophosphatases/genetics , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Tandem Mass Spectrometry , Guanosine Triphosphate , Mercaptopurine
2.
Viruses ; 15(6)2023 05 28.
Article in English | MEDLINE | ID: mdl-37376566

ABSTRACT

The cell-cell contact between HIV-1-infected and uninfected cells forms a virological synapse (VS) to allow for efficient HIV-1 transmission. Not only are HIV-1 components polarized and accumulate at cell-cell interfaces, but viral receptors and lipid raft markers are also. To better understand the nature of the HIV-1 VS, detergent-resistant membrane (DRM) fractions were isolated from an infected-uninfected cell coculture and compared to those from non-coculture samples using 2D fluorescence difference gel electrophoresis. Mass spectrometry revealed that ATP-related enzymes (ATP synthase subunit and vacuolar-type proton ATPase), protein translation factors (eukaryotic initiation factor 4A and mitochondrial elongation factor Tu), protein quality-control-related factors (protein disulfide isomerase A3 and 26S protease regulatory subunit), charged multivesicular body protein 4B, and vimentin were recruited to the VS. Membrane flotation centrifugation of the DRM fractions and confocal microscopy confirmed these findings. We further explored how vimentin contributes to the HIV-1 VS and found that vimentin supports HIV-1 transmission through the recruitment of CD4 to the cell-cell interface. Since many of the molecules identified in this study have previously been suggested to be involved in HIV-1 infection, we suggest that a 2D difference gel analysis of DRM-associated proteins may reveal the molecules that play crucial roles in HIV-1 cell-cell transmission.


Subject(s)
Detergents , HIV Infections , Humans , Detergents/pharmacology , Vimentin/metabolism , Proteomics/methods , HIV Infections/metabolism , Adenosine Triphosphate/metabolism , Membrane Microdomains/metabolism
3.
Mol Cancer Res ; 19(5): 921-931, 2021 05.
Article in English | MEDLINE | ID: mdl-33574130

ABSTRACT

Malignant mesothelioma (MM) is an aggressive tumor that typically develops after a long latency following asbestos exposure. Although mechanistic target of rapamycin complex 1 (mTORC1) activation enhances MM cell growth, the mTORC1 inhibitor everolimus has shown limited efficacy in clinical trials of MM patients. We explored the mechanism underlying mTORC1 activation in MM cells and its effects on cell proliferation and progression. Analysis of the expression profiles of 87 MMs from The Cancer Genome Atlas revealed that 40 samples (46%) displayed altered expression of RPTOR (mTORC1 component) and genes immediately upstream that activate mTORC1. Among them, we focused on RHEB and RHEBL1, which encode direct activators of mTORC1. Exogenous RHEBL1 expression enhanced MM cell growth, indicating that RHEB-mTORC1 signaling acts as a pro-oncogenic cascade. We investigated molecules that directly activate RHEBs, identifying SmgGDS as a novel RHEB-binding protein. SmgGDS knockdown reduced mTORC1 activation and inhibited the proliferation of MM cells with mTORC1 activation. Interestingly, SmgGDS displayed high binding affinity with inactive GDP-bound RHEBL1, and its knockdown reduced cytosolic RHEBL1 without affecting its activation. These findings suggest that SmgGDS retains GDP-bound RHEBs in the cytosol, whereas GTP-bound RHEBs are localized on intracellular membranes to promote mTORC1 activation. We revealed a novel role for SmgGDS in the RHEB-mTORC1 pathway and its potential as a therapeutic target in MM with aberrant mTORC1 activation. IMPLICATIONS: Our data showing that SmgGDS regulates RHEB localization to activate mTORC1 indicate that SmgGDS can be used as a new therapeutic target for MM exhibiting mTORC1 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesothelioma, Malignant/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Animals , Cell Proliferation/physiology , Female , HEK293 Cells , HeLa Cells , Humans , Mesothelioma, Malignant/pathology , Mice , Mice, Nude
4.
Biochem Biophys Res Commun ; 522(3): 676-683, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31785816

ABSTRACT

The Arf (ADP-ribosylation factor) GAPs (GTPase-activating proteins) regulate membrane trafficking and actin cytoskeleton. The molecular mechanism of how Arf GAPs regulate actin cytoskeleton remains to be elucidated. We identified AGAP1, a subtype of Arf GAP, as a binding protein of FilGAP, a Rac-specific GAP, in mammalian cells. AGAP1 binds to C-terminus of FilGAP whereas FilGAP binds to N-terminus of AGAP1 containing GLD domain. FilGAP co-localized with AGAP1 at intracellular vesicles and targeting of FilGAP at the vesicles requires its interaction with AGAP1. Consistently, depletion of endogenous AGAP1 induced the accumulation of endogenous FilGAP into paxillin-positive focal adhesions and actin cytoskeletal structures. Knockdown of endogenous AGAP1 suppressed cell spreading on collagen and the suppression was released by depletion of endogenous FilGAP. Moreover, depletion of AGAP1 in MDA-MB-231 cells promoted cell invasion in extracellular matrices and depletion of FilGAP blocked the invasion. Taken together, the present study suggests that AGAP1 may regulate subcellular localization of FilGAP and control cell migration and invasion through interaction with FilGAP.


Subject(s)
GTPase-Activating Proteins/metabolism , Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , GTPase-Activating Proteins/analysis , HEK293 Cells , Humans , Neoplasm Invasiveness/pathology , Neoplasms/pathology
5.
Cell Death Dis ; 10(10): 746, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582731

ABSTRACT

A GGGGCC hexanucleotide repeat expansion in the C9ORF72 gene has been identified as the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. The repeat expansion undergoes unconventional translation to produce five dipeptide repeat proteins (DPRs). Although DPRs are thought to be neurotoxic, the molecular mechanism underlying the DPR-caused neurotoxicity has not been fully elucidated. The current study shows that poly-proline-arginine (poly-PR), the most toxic DPR in vitro, binds to and up-regulates nuclear paraspeckle assembly transcript 1 (NEAT1) that plays an essential role as a scaffold non-coding RNA during the paraspeckle formation. The CRISPR-assisted up-regulation of endogenous NEAT1 causes neurotoxicity. We also show that the poly-PR modulates the function of several paraspeckle-localizing heterogeneous nuclear ribonucleoproteins. Furthermore, dysregulated expression of TAR DNA-binding protein 43 (TDP-43) up-regulates NEAT1 expression and induces neurotoxicity. These results suggest that the increase in the paraspeckle formation may be involved in the poly-PR- and TDP-43-mediated neurotoxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Dipeptides/chemistry , Frontotemporal Dementia/metabolism , Inclusion Bodies/metabolism , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Inclusion Bodies/drug effects , Mice, Inbred ICR , Neurotoxins/toxicity , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Up-Regulation/drug effects
6.
Lab Invest ; 99(6): 793-806, 2019 06.
Article in English | MEDLINE | ID: mdl-30710119

ABSTRACT

Although inhibition of epidermal growth factor receptor (EGFR)-mediated cell signaling by the EGFR tyrosine kinase inhibitor gefitinib is highly effective against advanced non-small cell lung cancer, this drug might promote severe acute interstitial pneumonia. We previously reported that molecular hydrogen (H2) acts as a therapeutic and preventive anti-oxidant. Here, we show that treatment with H2 effectively protects the lungs of mice from severe damage caused by oral administration of gefitinib after intraperitoneal injection of naphthalene, the toxicity of which is related to oxidative stress. Drinking H2-rich water ad libitum mitigated naphthalene/gefitinib-induced weight loss and significantly improved survival, which was associated with a decrease in lung inflammation and inflammatory cytokines in the bronchoalveolar lavage fluid. Naphthalene decreased glutathione in the lung, increased malondialdehyde in the plasma, and increased 4-hydroxy-2-nonenal production in airway cells, all of which were mitigated by H2-rich water, indicating that the H2-rich water reverses cellular damage to the bronchial wall caused by oxidative stress. Finally, treatment with H2 did not interfere with the anti-tumor effects of gefitinib on a lung cancer cell line in vitro or on tumor-bearing mice in vivo. These results indicate that H2-rich water has the potential to improve quality of life during gefitinib therapy by mitigating lung injury without impairing anti-tumor activity.


Subject(s)
Acute Lung Injury/prevention & control , Antineoplastic Agents/adverse effects , Gefitinib/adverse effects , Hydrogen/therapeutic use , Lung Neoplasms/drug therapy , Acute Lung Injury/chemically induced , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Hydrogen/pharmacology , Lung/drug effects , Mice, Inbred C57BL , Naphthalenes , Oxidative Stress/drug effects , Random Allocation
7.
Cell Death Dis ; 9(10): 975, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250194

ABSTRACT

A GGGGCC repeat expansion in the C9ORF72 gene has been identified as the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. The repeat expansion undergoes unconventional translation to produce dipeptide repeat (DPR) proteins. Although it has been reported that DPR proteins cause neurotoxicity, the underlying mechanism has not been fully elucidated. In this study, we have first confirmed that proline-arginine repeat protein (poly-PR) reduces levels of ribosomal RNA and causes neurotoxicity and found that the poly-PR-induced neurotoxicity is repressed by the acceleration of ribosomal RNA synthesis. These results suggest that the poly-PR-induced inhibition of ribosome biogenesis contributes to the poly-PR-induced neurotoxicity. We have further identified DEAD-box RNA helicases as poly-PR-binding proteins, the functions of which are inhibited by poly-PR. The enforced reduction in the expression of DEAD-box RNA helicases causes impairment of ribosome biogenesis and neuronal cell death. These results together suggest that poly-PR causes neurotoxicity by inhibiting the DEAD-box RNA helicase-mediated ribosome biogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Arginine/metabolism , C9orf72 Protein/genetics , DEAD-box RNA Helicases/metabolism , Dipeptides/genetics , Frontotemporal Dementia/metabolism , Microsatellite Repeats/physiology , Proline/metabolism , Ribosomes/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Survival , Frontotemporal Dementia/genetics , HEK293 Cells , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred ICR/embryology , Neurons/metabolism , RNA, Ribosomal/metabolism
8.
Anticancer Res ; 37(9): 4789-4798, 2017 09.
Article in English | MEDLINE | ID: mdl-28870897

ABSTRACT

BACKGROUND: Curcumin, a major polyphenol of the spice turmeric, acts as a potent chemopreventive and chemotherapeutic agent in several cancer types, including colon cancer. Although various proteins have been shown to be affected by curcumin, how curcumin exerts its anticancer activity is not fully understood. MATERIALS AND METHODS: Phosphoproteomic analyses were performed using SW480 and SW620 human colon cancer cells to identify curcumin-affected signaling pathways. RESULTS: Curcumin inhibited the growth of the two cell lines in a dose-dependent manner. Thirty-nine curcumin-regulated phosphoproteins were identified, five of which are involved in cancer signaling pathways. Detailed analyses revealed that the mTORC1 and p53 signaling pathways are main targets of curcumin. CONCLUSION: Our results provide insight into the molecular mechanisms of the anticancer activities of curcumin and future molecular targets for its clinical application.


Subject(s)
Colonic Neoplasms/metabolism , Curcumin/pharmacology , Phosphoproteins/metabolism , Proteomics/methods , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , Humans
9.
Sci Rep ; 7: 46174, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28393893

ABSTRACT

Ectodomain shedding (shedding) is a post-translational modification, which liberates the extracellular domain of membrane proteins through juxtamembrane processing executed mainly by the ADAM (a disintegrin and metalloprotease) family of metalloproteases. Because shedding alters characteristics of cells in a rapid and irreversible manner, it should be strictly regulated. However, the molecular mechanisms determining membrane protein susceptibility to shedding (shedding susceptibility) are largely unknown. Here we report that alternative splicing can give rise to both shedding-susceptible and shedding-resistant CADM1 (cell adhesion molecule 1) variant proteins. We further show that O-glycans adjacent to the shedding cleavage site interfere with CADM1 shedding, and the only 33-bp alternative exon confers shedding susceptibility to CADM1 by inserting five non-glycosylatable amino acids between interfering O-glycans and the shedding cleavage site. These results demonstrate that shedding susceptibility of membrane protein can be determined at two different levels of its biosynthesis pathway, alternative splicing and O-glycosylation.


Subject(s)
Alternative Splicing/genetics , Cell Adhesion Molecule-1/chemistry , Cell Adhesion Molecule-1/genetics , ADAM17 Protein/metabolism , Alternative Splicing/drug effects , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Adhesion Molecule-1/metabolism , Exons/genetics , Glycosylation/drug effects , Isotope Labeling , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Models, Biological , Proteomics , RAW 264.7 Cells , Receptors, Immunologic/metabolism , Threonine/genetics
10.
Sci Rep ; 6: 33742, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27645772

ABSTRACT

Leber congenital amaurosis (LCA) is a hereditary early-onset retinal dystrophy that is accompanied by severe macular degeneration. In this study, novel compound heterozygous mutations were identified as LCA-causative in chaperonin-containing TCP-1, subunit 2 (CCT2), a gene that encodes the molecular chaperone protein, CCTß. The zebrafish mutants of CCTß are known to exhibit the eye phenotype while its mutation and association with human disease have been unknown. The CCT proteins (CCT α-θ) forms ring complex for its chaperon function. The LCA mutants of CCTß, T400P and R516H, are biochemically instable and the affinity for the adjacent subunit, CCTγ, was affected distinctly in both mutants. The patient-derived induced pluripotent stem cells (iPSCs), carrying these CCTß mutants, were less proliferative than the control iPSCs. Decreased proliferation under Cct2 knockdown in 661W cells was significantly rescued by wild-type CCTß expression. However, the expression of T400P and R516H didn't exhibit the significant effect. In mouse retina, both CCTß and CCTγ are expressed in the retinal ganglion cells and connecting cilium of photoreceptor cells. The Cct2 knockdown decreased its major client protein, transducing ß1 (Gß1). Here we report the novel LCA mutations in CCTß and the impact of chaperon disability by these mutations in cellular biology.


Subject(s)
Cell Proliferation/genetics , Chaperonin Containing TCP-1 , Induced Pluripotent Stem Cells , Leber Congenital Amaurosis , Mutation , Animals , Chaperonin Containing TCP-1/genetics , Chaperonin Containing TCP-1/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Protein Stability , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
Genes Cells ; 21(6): 579-93, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27059097

ABSTRACT

The serine/threonine kinase mTOR forms two distinct complexes, mTORC1 and mTORC2, and controls a number of biological processes, including proliferation, survival and autophagy. Although the function of mTORC1 has been extensively studied, the mTORC2 signaling pathway largely remains to be elucidated. Here, we have shown that mTORC2 phosphorylates filamin A, an actin cross-linking protein, at serine 2152 (S2152) both in vivo and in living cells. Treatment of HeLa cells with Torin1 (an mTORC1/mTORC2 inhibitor), but not rapamycin (an mTORC1 inhibitor), suppressed the phosphorylation of filamin A, which decreased the binding of filamin A with ß7-integrin cytoplasmic tail. Torin1 also inhibited focal adhesion formation and cell migration in A7 filamin A-replete melanoma cells but not in M2 filamin A-deficient cells, suggesting a pivotal role for mTORC2 in filamin A function. Finally, reduced focal adhesion formation in M2 cells was significantly rescued by expressing wild type but not S2152A nonphosphorylatable mutant of filamin A. Taken together, our results indicate that mTORC2 regulates filamin A-dependent focal adhesions and cell migration.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Filamins/metabolism , Focal Adhesions , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein
12.
Mitochondrion ; 28: 79-87, 2016 05.
Article in English | MEDLINE | ID: mdl-27090168

ABSTRACT

We applied stimulated emission depletion (STED) imaging with subdiffraction resolution to submitochondrial structures in mitochondria. Their shapes depend on both a cell's type and its physiological state. Staining with a cationic fluorescent dye, tetramethylrhodamine methyl ester (TMRM), unveiled intriguing details of lamellar structure, consisting of rapidly changeable, curtain-like formations. The TMRM-positive structure colocalized with neither proteins in the matrix nor on the outer membrane, but partially localized with the nucleoid. Suppression of a component in the mitochondrial contact site disrupted the lamellar TMRM-positive structure. Uncoupling of the oxidative phosphorylation system released TMRM from the inner membrane without any alteration in the matrix structure. STED images further showed that complexes of the electron transport chain are located on the surface of TMRM-positive structures. The approach presented here provides novel insights into the in vivo nature of submitochondrial structures, and can be used for further functional investigations of these complex structures.


Subject(s)
Microscopy/methods , Mitochondria/chemistry , Mitochondria/ultrastructure , Staining and Labeling/methods , Humans , Rhodamines/metabolism
13.
PLoS One ; 11(1): e0146593, 2016.
Article in English | MEDLINE | ID: mdl-26751795

ABSTRACT

FilGAP is a Rac-specific GTPase-activating protein (GAP) that suppresses lamellae formation. In this study, we have identified RBM10 (RNA Binding Motif domain protein 10) as a FilGAP-interacting protein. Although RBM10 is mostly localized in the nuclei in human melanoma A7 cells, forced expression of Src family tyrosine kinase Fyn induced translocation of RBM10 from nucleus into cell peripheries where RBM10 and FilGAP are co-localized. The translocation of RBM10 from nucleus appears to require catalytic activity of Fyn since kinase-negative Fyn mutant failed to induce translocation of RBM10 in A7 cells. When human breast carcinoma MDA-MB-231 cells are spreading on collagen-coated coverslips, endogenous FilGAP and RBM10 were localized at the cell periphery with tyrosine-phosphorylated proteins. RBM10 appears to be responsible for targeting FilGAP at the cell periphery because depletion of RBM10 by siRNA abrogated peripheral localization of FilGAP during cell spreading. Association of RBM10 with FilGAP may stimulate RacGAP activity of FilGAP. First, forced expression of RBM10 suppressed FilGAP-mediated cell spreading on collagen. Conversely, depletion of endogenous RBM10 by siRNA abolished FilGAP-mediated suppression of cell spreading on collagen. Second, FilGAP suppressed formation of membrane ruffles induced by Fyn and instead produced spiky cell protrusions at the cell periphery. This protrusive structure was also induced by depletion of Rac, suggesting that the formation of protrusions may be due to suppression of Rac by FilGAP. We found that depletion of RBM10 markedly reduced the formation of protrusions in cells transfected with Fyn and FilGAP. Finally, depletion of RBM10 blocked FilGAP-mediated suppression of ruffle formation induced by EGF. Taken together, these results suggest that Src family tyrosine kinase signaling may regulate FilGAP through association with RBM10.


Subject(s)
Breast Neoplasms/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/metabolism , Signal Transduction , src-Family Kinases/metabolism , Catalysis , Cell Line, Tumor , Cell Nucleus/metabolism , Collagen/chemistry , DNA, Complementary/metabolism , Female , HEK293 Cells , Humans , Microscopy, Fluorescence , Plasmids/metabolism , RNA, Small Interfering/metabolism , Tyrosine/chemistry
14.
Eur J Hum Genet ; 24(3): 408-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26059841

ABSTRACT

Loss-of-function mutations in filamin A (FLNA) cause an X-linked dominant disorder with multiple organ involvement. Affected females present with periventricular nodular heterotopia (PVNH), cardiovascular complications, thrombocytopenia and Ehlers-Danlos syndrome. These mutations are typically lethal to males, and rare male survivors suffer from failure to thrive, PVNH, and severe cardiovascular and gastrointestinal complications. Here we report two surviving male siblings with a loss-of-function mutation in FLNA. They presented with multiple complications, including valvulopathy, intestinal malrotation and chronic intestinal pseudo-obstruction (CIPO). However, these siblings had atypical clinical courses, such as a lack of PVNH and a spontaneous improvement of CIPO. Trio-based whole-exome sequencing revealed a 4-bp deletion in exon 40 that was predicted to cause a lethal premature protein truncation. However, molecular investigations revealed that the mutation induced in-frame skipping of the mutated exon, which led to the translation of a mutant FLNA missing an internal region of 41 amino acids. Functional analyses of the mutant protein suggested that its binding affinity to integrin, as well as its capacity to induce focal adhesions, were comparable to those of the wild-type protein. These results suggested that exon skipping of FLNA partially restored its protein function, which could contribute to amelioration of the siblings' clinical courses. This study expands the diversity of the phenotypes associated with loss-of-function mutations in FLNA.


Subject(s)
Exons/genetics , Filamins/genetics , Filamins/metabolism , Mutation/genetics , Adult , Blood Cells/metabolism , Child , Child, Preschool , Female , Fluorescent Antibody Technique , HEK293 Cells , Humans , Infant , Infant, Newborn , Male , Pedigree , Phenotype , Young Adult
15.
PLoS One ; 10(5): e0128189, 2015.
Article in English | MEDLINE | ID: mdl-26010099

ABSTRACT

Kawasaki disease (KD), an acute vasculitis that preferentially affects coronary arteries, is still the leading cause of acquired heart disease in children. Although the involvement of immune system malfunction in the onset of KD is suggested, its etiology still remains to be clarified. We investigated autoantibodies in KD patients, which are frequently found in sera from patients with autoimmune diseases, vasculitides and arteritides. We performed two-dimensional western blotting and LC-MS/MS to analyze the antigens of autoantibodies, detected two protein spots with 4 out of 24 sera from KD patients but not with 6 control sera, and identified the antigens as 4-trimethylaminobutyraldehyde dehydrogenase (TMABA-DH). A slot blot analysis with TMABA-DH as an antigen also revealed higher reactivities of patients' sera than control sera (positive rates: 18/43 vs 3/41). Using an enzyme-linked immunosorbent assay (ELISA), we found that the reactivity of anti-TMABA-DH antibodies in sera from KD patients was significantly higher than that in sera from age-matched controls. The optimal cut-off value of 0.043 had a sensitivity of 83.7% and a specificity of 80.0% in detecting KD patients (positive rates: 37/43 for KD patients, 9/41 for controls). Immunohistochemistry performed on thin sections of rat heart revealed that TMABA-DH colocalized with myosin light chains in cardiac myocytes. Patient sera with high reactivity gave similar immunostaining pattern. These results suggest that the detection of anti-TMABA-DH autoantibody could be a potential strategy for a diagnosis of KD.


Subject(s)
Aldehyde Oxidoreductases/immunology , Autoantibodies/immunology , Autoantigens/immunology , Mucocutaneous Lymph Node Syndrome/immunology , Myocytes, Cardiac/immunology , Aldehyde Oxidoreductases/blood , Animals , Autoantibodies/blood , Autoantigens/blood , Child , Child, Preschool , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Infant , Male , Mucocutaneous Lymph Node Syndrome/blood , Mucocutaneous Lymph Node Syndrome/diagnosis , Myocytes, Cardiac/metabolism , Myosin Light Chains/immunology , Myosin Light Chains/metabolism , Rats
16.
Anticancer Res ; 35(2): 881-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25667470

ABSTRACT

BACKGROUND/AIM: Accumulating evidence shows that various types of cancers induce a specific immune response resulting in the production of antibodies against self-components (autoantibodies). The aim of the present study was to identify antigens for autoantibodies in sera from patients with ovarian cancer, especially clear cell carcinoma (CCC), as novel diagnostic markers for the disease. MATERIALS AND METHODS: The reactivity of individual sera from patients was examined by two-dimensional (2-D) immunoblotting using lysates of CCC cell lines, ES-2 and RMG-1, as antigens to identify autoantigens. ELISA was established to quantitatively measure autoantibody titer of patients' sera. RESULTS: Autoantibodies against RhoGDI were induced in sera of ovarian cancer patients. Elevated levels of autoantibodies against heterogeneous nuclear ribonucleoprotein L (hnRNPL) and a mitochondrial protein, dihydrolipoamide dehydrogenase (DLD), were detected in patients with CCC. CONCLUSION: Autoantibodies against RhoGDI and hnRNPL and DLD may serve as novel diagnostic markers for ovarian cancer and CCC, respectively.


Subject(s)
Autoantibodies/blood , Biomarkers, Tumor/blood , Ovarian Neoplasms/immunology , Proteomics , Autoantibodies/genetics , Cloning, Molecular , Electrophoresis, Gel, Two-Dimensional , Enzyme-Linked Immunosorbent Assay , Female , Humans , Middle Aged , Ovarian Neoplasms/blood
17.
Cytoskeleton (Hoboken) ; 72(3): 146-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25712270

ABSTRACT

The function of kidney podocytes is closely associated with actin cytoskeleton. Rho family small GTPase RhoA promotes stress fiber assembly through Rho-associated protein kinase (ROCK)-dependent myosin II phosphorylation and plays an important role in maintenance of actin stress fibers of podocytes. However, little is known how stress fiber assembly is regulated in podocytes. Here, we found that afadin, an actin filament-binding protein, is required for RhoA/ROCK-dependent formation of actin stress fibers in rat podocyte C7 cells. We show that depletion of afadin in C7 cells induced loss of actin stress fibers. Conversely, forced expression of afadin increased the formation of actin stress fibers. Depletion of afadin inactivated RhoA and reduced the phosphorylation of myosin II. Moreover, the DIL domain of afadin appears to be responsible for actin stress fiber formation. Thus, afadin mediates RhoA/ROCK signaling and contributes to the formation of actin stress fibers in podocyte cells.


Subject(s)
Kidney/cytology , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Stress Fibers/metabolism , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Actins/metabolism , Animals , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Podocytes/pathology , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Temperature , rho GTP-Binding Proteins/metabolism
18.
Hum Mol Genet ; 24(6): 1513-27, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25378556

ABSTRACT

Dysregulation of transactive response DNA-binding protein-43 (TDP-43) is thought to be linked to the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). TDP-43 normally localizes in the nucleus but its main localization shifts to the cytoplasm in most affected cells of ALS and FTLD patients. It is not yet known whether nuclear or cytoplasmic TDP-43 is responsible for TDP-43-induced neurotoxicity. In this study, we show that nuclear TDP-43 causes TDP-43 neurotoxicity. DNA/RNA-binding and dimerization of TDP-43 are both essential for TDP-43-induced cell death. Moreover, endogenous heterogeneous nuclear ribonucleoprotein-U (hnRNP-U) binds to TDP-43 and knocking-down of hnRNP-U induces neurotoxicity, whereas overexpression of hnRNP-U or hnRNP-A2 inhibits TDP-43-induced neurotoxicity. In addition, hnRNP-U inhibits TDP-43-mediated alterations in splicing of POLDIP3 mRNA. Altogether, these results suggest that nuclear TDP-43 becomes neurotoxic by escaping from the inhibitory regulation by hnRNPs.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Neurons/metabolism , Animals , Chlorocebus aethiops , DNA-Binding Proteins/toxicity , Humans , Mice , Neurons/drug effects , Nuclear Proteins/genetics , Protein Transport , RNA Splicing , RNA-Binding Proteins/genetics
19.
J Biol Chem ; 290(2): 1096-105, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25422319

ABSTRACT

Rheb small GTPases, which consist of Rheb1 and Rheb2 (also known as RhebL1) in mammalian cells, are unique members of the Ras superfamily and play central roles in regulating protein synthesis and cell growth by activating mTOR. To gain further insight into the function of Rheb, we carried out a search for Rheb-binding proteins and found that Rheb binds to CAD protein (carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotase), a multifunctional enzyme required for the de novo synthesis of pyrimidine nucleotides. CAD binding is more pronounced with Rheb2 than with Rheb1. Rheb binds CAD in a GTP- and effector domain-dependent manner. The region of CAD where Rheb binds is located at the C-terminal region of the carbamoyl-phosphate synthetase domain and not in the dihydroorotase and aspartate transcarbamoylase domains. Rheb stimulated carbamoyl-phosphate synthetase activity of CAD in vitro. In addition, an elevated level of intracellular UTP pyrimidine nucleotide was observed in Tsc2-deficient cells, which was attenuated by knocking down of Rheb. Immunostaining analysis showed that expression of Rheb leads to increased accumulation of CAD on lysosomes. Both a farnesyltransferase inhibitor that blocks membrane association of Rheb and knockdown of Rheb mislocalized CAD. These results establish CAD as a downstream effector of Rheb and suggest a possible role of Rheb in regulating de novo pyrimidine nucleotide synthesis.


Subject(s)
Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Pyrimidine Nucleosides/biosynthesis , ras Proteins/metabolism , Animals , Cell Proliferation/genetics , Humans , Lysosomes/metabolism , Lysosomes/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/genetics , Multiprotein Complexes/metabolism , Neuropeptides/genetics , Protein Binding , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases/metabolism , ras Proteins/genetics
20.
Biochem Biophys Res Commun ; 453(1): 117-23, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25261720

ABSTRACT

Aralin from Aralia elata is a newly identified type II ribosome- inactivating protein, which preferentially induces apoptosis in cancer cells. In this study, we identified that the aralin receptor is a 110-kDa high-density lipoprotein-binding protein (HDLBP), which functions as a HDL receptor. The sensitivities of tumor cell lines to aralin were dependent on the expression levels of the 110-kDa HDLBP and its forced expression in aralin-resistant Huh7 cells conferred aralin sensitivity. HDLBP-knockdown HeLa cells showed a significant aralin resistance in vitro and in vivo. Conversely, ectopic expression of the 150-kDa HDLBP resulted in increased aralin sensitivity in vivo, accompanying enhanced expression of the 110-kDa HDLBP. Thus, these results showed that the 110-kDa HDLBP in lipid rafts acted as an aralin receptor and that its expression levels determined aralin sensitivity, suggesting that aralin could be a promising anticancer drug for HDLBP-overexpressing tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , RNA-Binding Proteins/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Aralia/chemistry , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Hep G2 Cells , Humans , Lipoproteins, HDL/antagonists & inhibitors , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Membrane Microdomains/metabolism , Mice, Nude , Molecular Weight , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Receptors, Lipoprotein/antagonists & inhibitors , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/pharmacokinetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...