Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4798, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37558718

ABSTRACT

UBA1 is the primary E1 ubiquitin-activating enzyme responsible for generation of activated ubiquitin required for ubiquitination, a process that regulates stability and function of numerous proteins. Decreased or insufficient ubiquitination can cause or drive aging and many diseases. Therefore, a small-molecule enhancing UBA1 activity could have broad therapeutic potential. Here we report that auranofin, a drug approved for the treatment of rheumatoid arthritis, is a potent UBA1 activity enhancer. Auranofin binds to the UBA1's ubiquitin fold domain and conjugates to Cys1039 residue. The binding enhances UBA1 interactions with at least 20 different E2 ubiquitin-conjugating enzymes, facilitating ubiquitin charging to E2 and increasing the activities of seven representative E3s in vitro. Auranofin promotes ubiquitination and degradation of misfolded ER proteins during ER-associated degradation in cells at low nanomolar concentrations. It also facilitates outer mitochondrial membrane-associated degradation. These findings suggest that auranofin can serve as a much-needed tool for UBA1 research and therapeutic exploration.


Subject(s)
Ubiquitin-Conjugating Enzymes , Ubiquitin , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Auranofin/pharmacology , Ubiquitination , Ubiquitin-Activating Enzymes/metabolism
2.
Mol Biol Cell ; 33(13): ar120, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36074076

ABSTRACT

RNF5 E3 ubiquitin ligase has multiple biological roles and has been linked to the development of severe diseases such as cystic fibrosis, acute myeloid leukemia, and certain viral infections, emphasizing the importance of discovering small-molecule RNF5 modulators for research and drug development. The present study describes the synthesis of a new benzo[b]thiophene derivative, FX12, that acts as a selective small-molecule inhibitor and degrader of RNF5. We initially identified the previously reported STAT3 inhibitor, Stattic, as an inhibitor of dislocation of misfolded proteins from the endoplasmic reticulum (ER) lumen to the cytosol in ER-associated degradation. A concise structure-activity relationship campaign (SAR) around the Stattic chemotype led to the synthesis of FX12, which has diminished activity in inhibition of STAT3 activation and retains dislocation inhibitory activity. FX12 binds to RNF5 and inhibits its E3 activity in vitro as well as promoting proteasomal degradation of RNF5 in cells. RNF5 as a molecular target for FX12 was supported by the facts that FX12 requires RNF5 to inhibit dislocation and negatively regulates RNF5 function. Thus, this study developed a small-molecule inhibitor and degrader of the RNF5 ubiquitin ligase, providing a chemical biology tool for RNF5 research and therapeutic development.


Subject(s)
DNA-Binding Proteins , Ubiquitin , Cyclic S-Oxides , DNA-Binding Proteins/metabolism , Thiophenes/pharmacology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Hum Mol Genet ; 31(15): 2639-2654, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35333353

ABSTRACT

XBP1 variant 1 (Xv1) is the most abundant XBP1 variant and is highly enriched across cancer types but nearly none in normal tissues. Its expression is associated with poor patients' survival and is specifically required for survival of malignant cells, but the underlying mechanism is not known. Here we report that Xv1 upregulates the polyglutamylase tubulin tyrosine ligase-like 6 (TTLL6) and promotes mitosis of cancer cells. Like the canonical XBP1, Xv1 mRNA undergoes unconventional splicing by IRE1α under endoplasmic reticulum stress, but it is also constitutively spliced by IRE1ß. The spliced Xv1 mRNA encodes the active form of Xv1 protein (Xv1s). RNA sequencing in HeLa cells revealed that Xv1s overexpression regulates expression of genes that are not involved in the canonical unfolded protein response, including TTLL6 as a highly upregulated gene. Gel shift assay and chromatin immunoprecipitation revealed that Xv1s bind to the TTLL6 promoter region. Knockdown of TTLL6 caused death of cancer cells but not benign and normal cells, similar to the effects of knocking down Xv1. Moreover, overexpression of TTLL6 partially rescued BT474 cells from apoptosis induced by either TTLL6 or Xv1 knockdown, supporting TTLL6 as an essential downstream effector of Xv1 in regulating cancer cell survival. TTLL6 is localized in the mitotic spindle of cancer cells. Xv1 or TTLL6 knockdown resulted in decreased spindle polyglutamylation and interpolar spindle, as well as congression failure, mitotic arrest and cell death. These findings suggest that Xv1 is essential for cancer cell mitosis, which is mediated, at least in part, by increasing TTLL6 expression.


Subject(s)
Endoribonucleases , Neoplasms , Endoplasmic Reticulum Stress , Endoribonucleases/genetics , Endoribonucleases/metabolism , HeLa Cells , Humans , Mitosis , Neoplasms/genetics , Peptide Synthases/genetics , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , Up-Regulation , X-Box Binding Protein 1/genetics
4.
Biochem Biophys Res Commun ; 562: 69-75, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34038755

ABSTRACT

XBP1 is a basic leucine zipper (bZIP) transcription factor and a key mediator of the endoplasmic reticulum (ER) stress-activated unfolded protein response (UPR). XBP1-mediated transcription facilitates cell adaptation to ER stress and also promotes tumor progression, while suppressing anti-tumor immunity. Here we report a novel XBP1 variant, namely XBP1 variant 1 (XBP1v1, Xv1 for short), that is specifically required for survival of cancer cells. Xv1 contains a cryptic first exon that is conserved only in humans and great apes. Comparing to XBP1, Xv1 encodes a protein with a different N-terminal sequence containing 25 amino acids. Analysis of RNAseq database reveals that Xv1 is broadly expressed across cancer types but almost none in normal tissues. Elevated Xv1 expression is associated with poor survival of patients with several types of cancer. Knockdown of Xv1 induces death of multiple cancer cell lines but has little effect on non-cancerous cells in vitro. Moreover, knockdown of Xv1 also inhibits growth of a xenograft breast tumor in mice. Together, our results indicate that Xv1 is essential for survival of cancer cells.


Subject(s)
Genetic Variation , Neoplasms/genetics , Neoplasms/pathology , X-Box Binding Protein 1/genetics , Animals , Cell Line, Tumor , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
5.
Antiviral Res ; 171: 104590, 2019 11.
Article in English | MEDLINE | ID: mdl-31421166

ABSTRACT

Two major flaviviruses, dengue virus (DENV) and Zika virus (ZIKV), cause severe health and economic burdens worldwide. Recently, genome-wide screenings have uncovered the importance of regulators of the Hrd1 ubiquitin ligase-mediated endoplasmic reticulum (ER)-associated degradation (ERAD) pathway for flavivirus replication in host cells. Here we report the identification of the compound Bardoxolone methyl (CDDO-me) as a potent inhibitor of the Hrd1 ubiquitin ligase-mediated ERAD, which possesses a broad-spectrum activity against both DENV and ZIKV. Cellular thermal shift assay (CETSA) suggested that CDDO-me binds to grp94, a key component of the Hrd1 pathway, at a low nanomolar concentration, whereas interaction was not detected with its paralog Hsp90. CDDO-me and the grp94 inhibitor PU-WS13 substantially suppressed DENV2 replication and the cytopathic effects caused by DENV and ZIKV infection. The antiviral activities of both compounds were demonstrated for all four DENV serotypes and four ZIKV strains in multiple human cell lines. This study defines grp94 as a crucial host factor for flavivirus replication and identified CDDO-me as a potent small molecule inhibitor of flavivirus infection. Inhibition of grp94 may contribute to the antiviral activity of CDDO-me. Further investigation of grp94 inhibitors may lead to a new class of broad-spectrum anti-flaviviral medications.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Membrane Glycoproteins/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , Zika Virus Infection/virology , Zika Virus/drug effects , Cell Survival , Dengue/drug therapy , Dengue/metabolism , Humans , Oleanolic Acid/pharmacology , Virus Replication/drug effects , Zika Virus Infection/drug therapy , Zika Virus Infection/metabolism
6.
Sci Rep ; 9(1): 10901, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358863

ABSTRACT

Infection with flaviviruses, such as dengue virus (DENV) and the recently re-emerging Zika virus (ZIKV), represents an increasing global risk. Targeting essential host elements required for flavivirus replication represents an attractive approach for the discovery of antiviral agents. Previous studies have identified several components of the Hrd1 ubiquitin ligase-mediated endoplasmic reticulum (ER)-associated degradation (ERAD) pathway, a cellular protein quality control process, as host factors crucial for DENV and ZIKV replication. Here, we report that CP26, a small molecule inhibitor of protein dislocation from the ER lumen to the cytosol, which is an essential step for ERAD, has broad-spectrum anti-flavivirus activity. CP26 targets the Hrd1 complex, inhibits ERAD, and induces ER stress. Ricin and cholera toxins are known to hijack the protein dislocation machinery to reach the cytosol, where they exert their cytotoxic effects. CP26 selectively inhibits the activity of cholera toxin but not that of ricin. CP26 exhibits a significant inhibitory activity against both DENV and ZIKV, providing substantial protection to the host cells against virus-induced cell death. This study identified a novel dislocation inhibitor, CP26, that shows potent anti-DENV and anti-ZIKV activity in cells. Furthermore, this study provides the first example of the targeting of host ER dislocation with small molecules to combat flavivirus infection.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Virus Replication/drug effects , Zika Virus/drug effects , Animals , Chlorocebus aethiops , Dengue/drug therapy , Dengue Virus/physiology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum-Associated Degradation/drug effects , HeLa Cells , Host Microbial Interactions/drug effects , Humans , Ubiquitin-Protein Ligases/metabolism , Vero Cells , Zika Virus/physiology , Zika Virus Infection/drug therapy
7.
Nat Commun ; 8(1): 1472, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29133892

ABSTRACT

Endoplasmic-reticulum-associated degradation (ERAD) is an important protein quality control system which maintains protein homeostasis. Constituents of the ERAD complex and its role in neurodegeneration are not yet fully understood. Here, using proteomic and FRET analyses, we demonstrate that the ER protein membralin is an ERAD component, which mediates degradation of ER luminal and membrane substrates. Interestingly, we identify nicastrin, a key component of the γ-secretase complex, as a membralin binding protein and membralin-associated ERAD substrate. We demonstrate a reduction of membralin mRNA and protein levels in Alzheimer's disease (AD) brain, the latter of which inversely correlates with nicastrin abundance. Furthermore, membralin deficiency enhances γ-secretase activity and neuronal degeneration. In a mouse AD model, downregulating membralin results in ß-amyloid pathology, neuronal death, and exacerbates synaptic/memory deficits. Our results identify membralin as an ERAD component and demonstrate a critical role for ERAD in AD pathogenesis.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Cognitive Dysfunction/pathology , Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Memory/physiology , Nerve Tissue Proteins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/physiology , Cell Line , Female , HEK293 Cells , Humans , Male , Membrane Glycoproteins/metabolism , Membrane Proteins , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Protein Binding , Protein Folding , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics
8.
Elife ; 62017 05 02.
Article in English | MEDLINE | ID: mdl-28463106

ABSTRACT

Over 170 different mutations in the gene encoding SOD1 all cause amyotrophic lateral sclerosis (ALS). Available studies have been primarily focused on the mechanisms underlying mutant SOD1 cytotoxicity. How cells defend against the cytotoxicity remains largely unknown. Here, we show that misfolding of ALS-linked SOD1 mutants and wild-type (wt) SOD1 exposes a normally buried nuclear export signal (NES)-like sequence. The nuclear export carrier protein CRM1 recognizes this NES-like sequence and exports misfolded SOD1 to the cytoplasm. Antibodies against the NES-like sequence recognize misfolded SOD1, but not native wt SOD1 both in vitro and in vivo. Disruption of the NES consensus sequence relocalizes mutant SOD1 to the nucleus, resulting in higher toxicity in cells, and severer impairments in locomotion, egg-laying, and survival in Caenorhabditis elegans. Our data suggest that SOD1 mutants are removed from the nucleus by CRM1 as a defense mechanism against proteotoxicity of misfolded SOD1 in the nucleus.


Subject(s)
Active Transport, Cell Nucleus , Karyopherins/metabolism , Protein Folding , Receptors, Cytoplasmic and Nuclear/metabolism , Superoxide Dismutase-1/metabolism , Superoxide Dismutase-1/toxicity , Amino Acid Motifs , Animals , Caenorhabditis elegans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/toxicity , Protein Binding , Protein Sorting Signals , Superoxide Dismutase-1/chemistry , Exportin 1 Protein
9.
Mol Cancer Ther ; 16(4): 717-728, 2017 04.
Article in English | MEDLINE | ID: mdl-27903750

ABSTRACT

Although proteasome inhibitors such as bortezomib had significant therapeutic effects in multiple myeloma and mantel cell lymphoma, they exhibited minimal clinical activity as a monotherapy for solid tumors, including colorectal cancer. We found in this study that proteasome inhibition induced a remarkable nuclear exportation of ubiquitinated proteins. Inhibition of CRM1, the nuclear export carrier protein, hampered protein export and synergistically enhanced the cytotoxic action of bortezomib on colon cancer cells containing wild-type p53, which underwent G2-M cell-cycle block and apoptosis. Further analysis indicated that tumor suppressor p53 was one of the proteins exported from nuclei upon proteasome inhibition, and in the presence of CRM1 inhibitor KPT330, nuclear p53, and expression of its target genes were increased markedly. Moreover, knockdown of p53 significantly reduced the synergistic cytotoxic action of bortezomib and KPT330 on p53+/+ HCT116 cells. In mice, KPT330 markedly augmented the antitumor action of bortezomib against HCT116 xenografts as well as patient-derived xenografts that harbored functional p53. These results indicate that nuclear p53 is a major mediator in the synergistic antitumor effect of bortezomib and KPT330, and provides a rationale for the use of proteasome inhibitor together with nuclear export blocker in the treatment of colorectal cancer. It is conceivable that targeting nuclear exportation may serve as a novel strategy to overcome resistance and raise chemotherapeutic efficacy, especially for the drugs that activate the p53 system. Mol Cancer Ther; 16(4); 717-28. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Bortezomib/administration & dosage , Cell Nucleus/metabolism , Colorectal Neoplasms/drug therapy , Proteasome Inhibitors/administration & dosage , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Drug Synergism , HCT116 Cells , HeLa Cells , Humans , Hydrazines/administration & dosage , Hydrazines/pharmacology , Mice , Proteasome Inhibitors/pharmacology , Triazoles/administration & dosage , Triazoles/pharmacology , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
10.
Nat Plants ; 2: 16094, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27322605

ABSTRACT

When membrane proteins and secretory proteins are misfolded or incompletely folded, they are retained in the endoplasmic reticulum (ER) for further folding or degradation. The HMG-COA reductase degradation 1 (HRD1) and degradation of alpha2 10 (DOA10) complexes are two major components involved in the ER-associated protein degradation (ERAD) system in eukaryotic organisms(1-4). However, the relationship between these two complexes is largely unknown, especially in higher eukaryotes. Here, we report that the plant ubiquitin-conjugating enzyme 32 (UBC32), an ER-bound E2 working in the DOA10 complex, is maintained at low levels under standard conditions by proteasome-dependent degradation mediated by the HRD1 complex, the other E3 complex involved in ERAD. Loss of this negative regulation under ER stress increases capacity for degradation of misfolded proteins retained in the ER. Consistently, UBE2J1, the homologue of UBC32 in mammals, was also identified to be targeted by HRD1 for degradation. Taken together, these results suggest that the regulation of UBC32 (or UBE2J1) by the HRD1 complex is conserved between plants and mammals.


Subject(s)
Acyl Coenzyme A/metabolism , Arabidopsis Proteins/genetics , Arabidopsis/physiology , Endoplasmic Reticulum-Associated Degradation , Oxidoreductases/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
11.
Biochim Biophys Acta ; 1853(12): 3279-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26417682

ABSTRACT

The amount of transcription factor OCT4 is strictly regulated. A tight regulation of OCT4 levels is crucial for mammalian embryonic development and oncogenesis. However, the mechanisms underlying regulation of OCT4 protein expression and nuclear distribution are largely unknown. Here, we report that DPF2, a plant homeodomain (PHD) finger protein, is upregulated during H9 cell differentiation induced by retinoic acid. Endogenous interaction between DPF2 and OCT4 in P19 cells was revealed by an immunoprecipitation assay. GST-pull down assay proved that OCT4 protein in H9 cells and recombinant OCT4 can precipitate with DPF2 in vitro. In vitro ubiquitination assay demonstrated DPF2 might serve as an E3 ligase. Knock down of dpf2 using siRNA increased OCT4 protein level and stability in P19 cells. DPF2 siRNAs also up-regulates OCT4 but not NANOG in H9 cells. However, RA fails to downregulates OCT4 protein level in cells infected by lenitviruses containing DPF2 siRNA. Moreover, overexpression of both DPF2 and OCT4 in 293 cells proved the DPF2-OCT4 interaction. DPF2 but not PHD2 mutant DPF2 enhanced ubiquitination and degradation of OCT4 in 293 cells co-expressed DPF2 and OCT4. Both wild type DPF2 and PHD2 mutant DPF2 redistributes nuclear OCT4 without affecting DPF2-OCT4 interaction. Further analysis indicated that DPF2 decreases monomeric and mono-ubiquitinated OCT4, assembles poly-ubiquitin chains on OCT4 mainly through Ub-K48 linkage. These findings contribute to an understanding of how OCT4 protein level and nuclear distribution is regulated by its associated protein.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/physiology , Octamer Transcription Factor-3/metabolism , Cell Differentiation/drug effects , Cell Line , DNA-Binding Proteins/genetics , Humans , Protein Binding , Transcription Factors , Tretinoin/pharmacology , Ubiquitination
12.
Biochem Biophys Res Commun ; 458(2): 424-8, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25660456

ABSTRACT

Misfolded proteins or orphan subunits of protein complexes are removed from the endoplasmic reticulum (ER) by ER-associated degradation (ERAD). ERAD requires dislocation, also known as retrotranslocation, of those unwanted proteins from the ER lumen to the cytosol for destruction by the proteasomes. Over one hundred ERAD component proteins have been identified but their role in dislocation remain poorly understood. Here we assessed the requirement of ERAD components for dislocation of NHK in live cells using our recently developed dislocation-induced reconstituted GFP (drGFP) assay. RNAi revealed that 12 out of 21 ERAD components examined are required for efficient dislocation of NHK among which Hrd1, Sel1L, GRP94 and p97/VCP are critically required. In addition, knockdown of 7 of the 21 components enhanced NHK dislocation. This study uncovers a complex functional network of proteins required for NHK dislocation.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , alpha 1-Antitrypsin/metabolism , HeLa Cells , Hong Kong , Humans
13.
Data Brief ; 5: 599-604, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26958616

ABSTRACT

DPF2, also named ubi-d4/requiem (REQU), interacts with a protein complex containing OCT4. This paper provides data in support of the research article entitled "DPF2 regulates OCT4 protein level and nuclear distribution". The highlights include: (1) Denature-immunoprecipitation assay revealed ubiquitination of OCT4 in pluripotent H9 cells, which was enhancedby MG132, a proteasome inhibitor. (2) Well colocalization of ectopic OCT4 and FLAG-Ub was found in HeLa cells, which was also increased by MG132. (3) MG132 treatment decreased DPF2 cytoplasmic expression in vivo. These data give insights into how proteasome inhibition contributes to studying ubiquitnation of OCT4.

14.
PLoS One ; 8(9): e73482, 2013.
Article in English | MEDLINE | ID: mdl-24039955

ABSTRACT

Proteins can be modified with eight homogenous ubiquitin chains linked by an isopeptide bond between the C-terminus of one ubiquitin and an amine from one of the seven lysines or the N-terminal methionine of the next ubiquitin. These topologically distinct ubiquitin chains signal for many essential cellular functions, such as protein degradation, cell cycle progression, DNA repair, and signal transduction. The lysine 48 (K48)-linked ubiquitin chain is one of the most abundant chains and a major proteasome-targeting signal in cells. Despite recent advancements in imaging linkage-specific polyubiquitin chains, no tool is available for imaging K48 chains in live cells. Here we report on a ubiquitination-induced fluorescence complementation (UiFC) assay for detecting K48 ubiquitin chains in vitro and in live cells. For this assay, two nonfluorescent fragments of a fluorescent protein were fused to the ubiquitin-interacting motifs (UIMs) of epsin1 protein. Upon simultaneous binding to a ubiquitin chain, the nonfluorescent fragments of the two fusion proteins are brought in close proximity to reconstitute fluorescence. When used in vitro, UiFC preferentially detected K48 ubiquitin chains with excellent signal-to-noise ratio. Time-lapse imaging revealed that UiFC is capable of monitoring increases in polyubiquitination induced by treatment with proteasome inhibitor, by agents that induce stress, and during mitophagy in live cells.


Subject(s)
Ubiquitin/analysis , Cell Survival , Fluorescence , HeLa Cells , Humans , Microscopy, Fluorescence , Ubiquitin/metabolism , Ubiquitination
15.
Mol Biol Cell ; 24(22): 3511-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24068325

ABSTRACT

Smyd1b is a member of the Smyd family that is specifically expressed in skeletal and cardiac muscles. Smyd1b plays a key role in thick filament assembly during myofibrillogenesis in skeletal muscles of zebrafish embryos. To better characterize Smyd1b function and its mechanism of action in myofibrillogenesis, we analyzed the effects of smyd1b knockdown on myofibrillogenesis in skeletal and cardiac muscles of zebrafish embryos. The results show that knockdown of smyd1b causes significant disruption of myofibril organization in both skeletal and cardiac muscles of zebrafish embryos. Microarray and quantitative reverse transcription-PCR analyses show that knockdown of smyd1b up-regulates heat shock protein 90 (hsp90) and unc45b gene expression. Biochemical analysis reveals that Smyd1b can be coimmunoprecipitated with heat shock protein 90 α-1 and Unc45b, two myosin chaperones expressed in muscle cells. Consistent with its potential function in myosin folding and assembly, knockdown of smyd1b significantly reduces myosin protein accumulation without affecting mRNA expression. This likely results from increased myosin degradation involving unc45b overexpression. Together these data support the idea that Smyd1b may work together with myosin chaperones to control myosin folding, degradation, and assembly into sarcomeres during myofibrillogenesis.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myosins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Histone-Lysine N-Methyltransferase/deficiency , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Muscle Development/genetics , Muscle Proteins , Muscle, Skeletal/growth & development , Muscle, Skeletal/ultrastructure , Myocardium/ultrastructure , Myosins/chemistry , Myosins/metabolism , Protein Binding , Protein Folding , Protein Stability , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
16.
Biochem Biophys Res Commun ; 439(1): 154-9, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23939048

ABSTRACT

The neural differentiation of human embryonic stem cells (ESCs) is a potential tool for elucidating the key mechanisms involved in human neurogenesis. Nestin and ß-III-tubulin, which are cytoskeleton proteins, are marker proteins of neural stem cells (NSCs) and neurons, respectively. However, the expression patterns of nestin and ß-III-tubulin in neural derivatives from human ESCs remain unclear. In this study, we found that neural progenitor cells (NPCs) derived from H9 cells express high levels of nestin and musashi-1. In contrast, ß-III-tubulin was weakly expressed in a few NPCs. Moreover, in these cells, nestin formed filament networks, whereas ß-III-tubulin was distributed randomly as small particles. As the differentiation proceeded, the nestin filament networks and the ß-III-tubulin particles were found in both the cell soma and the cellular processes. Moreover, the colocalization of nestin and ß-III-tubulin was found mainly in the cell processes and neurite-like structures and not in the cell soma. These results may aid our understanding of the expression patterns of nestin and ß-III-tubulin during the neural differentiation of H9 cells.


Subject(s)
Cell Differentiation , Cytoskeleton/metabolism , Embryonic Stem Cells/cytology , Neurons/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Intermediate Filament Proteins/metabolism , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Nestin , Neurogenesis , RNA-Binding Proteins/metabolism , Tubulin/metabolism
17.
J Biol Chem ; 287(33): 28057-66, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22722934

ABSTRACT

Misfolded proteins in the endoplasmic reticulum (ER) are dislocated to the cytosol to be degraded by the proteasomes. Various plant and bacterial toxins and certain viruses hijack this dislocation pathway to exert their toxicity or to infect cells. In this study, we report a dislocation-dependent reconstituted GFP (drGFP) assay that allows, for the first time, imaging proteins dislocated from the ER lumen to the cytosol in living cells. Our results indicate that both luminal and membrane-spanning ER proteins can be fully dislocated from the ER to the cytosol. By combining the drGFP assay with RNAi or chemical inhibitors of proteins in the Hrd1 ubiquitin ligase complex, we demonstrate that the Sel1L, Hrd1, p97/VCP, and importin ß proteins are required for the dislocation of misfolded luminal α-1 antitrypsin. The strategy described in this work is broadly applicable to the study of other types of transmembrane transport of proteins and likely also of viruses and toxins in living cells.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha 1-Antitrypsin/metabolism , beta Karyopherins/metabolism , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Cytosol/metabolism , Endoplasmic Reticulum/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Intracellular Membranes/metabolism , Microscopy, Fluorescence , Protein Transport/physiology , Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein , alpha 1-Antitrypsin/genetics , beta Karyopherins/genetics
18.
Biochem Biophys Res Commun ; 417(2): 738-43, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22197812

ABSTRACT

Endoplasmic reticulum (ER) stress occurs during early embryonic development. The aim of this study is to determine whether ER stress occurs during human embryonic stem cell differentiation induced by retinoic acid (RA). H9 human embryonic stem cells were subjected to RA treatment for up to 29days to induce differentiation. HEK293 cells were treated with RA as a control. The results demonstrate that several ER stress-responsive genes are differentially regulated in H9 and HEK293 cells in response to 5days of RA treatment. GRP78/Bip was upregulated in H9 cells but downregulated in HEK293 cells. eIF2α was downregulated in H9 cells but not in HEK293 cells. Phosphorylation of eIF2α was downregulated in H9 cells but upregulated in HEK293 cells. XBP-1 was downregulated immediately after RA treatment in H9 cells, but its downregulation was much slower in HEK293 cells. Additionally, two ER-resident E3 ubiquitin ligases, gp78 and Hrd1, were both upregulated in H9 cells following 5 days of exposure to RA. Moreover, the protein Bcl2 was undetectable in H9 cells and H9-derived cells but was expressed in HEK293 cells, and it expression in the two types of cells was unaltered by RA treatment. In H9 cells treated with RA for 29 days, GRP78/Bip, XBP-1 and Bcl2 were all upregulated. These results suggest that ER stress is involved in H9 cell differentiation induced by RA.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Endoplasmic Reticulum Stress/physiology , Tretinoin/physiology , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Phosphorylation , Receptors, Autocrine Motility Factor/biosynthesis , Tretinoin/pharmacology , Ubiquitin-Protein Ligases/biosynthesis
19.
PLoS One ; 6(8): e24478, 2011.
Article in English | MEDLINE | ID: mdl-21909394

ABSTRACT

The small p97/VCP-interacting protein (SVIP) functions as an inhibitor of the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway. Here we show that overexpression of SVIP in HeLa cells leads to localization of p97/VCP at the plasma membrane, intracellular foci and juxtanuclear vacuoles. The p97/VCP-positive vacuolar structures colocalized or associated with LC3 and lamp1, suggesting that SVIP may regulate autophagy. In support of this possibility, knockdown of SVIP diminished, whereas overexpression of SVIP enhanced LC3 lipidation. Surprisingly, knockdown of SVIP reduced the levels of p62 protein at least partially through downregulation of its mRNA, which was accompanied by a decrease in starvation-induced formation of p62 bodies. Overexpression of SVIP, on the other hand, increased the levels of p62 protein and enhanced starvation-activated autophagy as well as promoted sequestration of polyubiquitinated proteins and p62 in autophagosomes. These results suggest that SVIP plays a regulatory role in p97 subcellular localization and is a novel regulator of autophagy.


Subject(s)
Adenosine Triphosphatases/metabolism , Autophagy , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Intracellular Membranes/metabolism , Lysosomes/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Line , Gene Knockdown Techniques , Green Fluorescent Proteins , Humans , Lysosomal Membrane Proteins/metabolism , Membrane Proteins , Mice , Microtubule-Associated Proteins/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Phagosomes/metabolism , Phosphate-Binding Proteins , Protein Transport , Recombinant Fusion Proteins/metabolism , Sequestosome-1 Protein , Spinal Cord/cytology , Spinal Cord/metabolism , Ubiquitination , Vacuoles/metabolism , Valosin Containing Protein
20.
J Biol Chem ; 286(39): 33921-30, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21832065

ABSTRACT

The mechanism by which misfolded proteins in the endoplasmic reticulum (ER) are retrotranslocated to the cytosol for proteasomal degradation is still poorly understood. Here, we show that importin ß, a well established nucleocytoplasmic transport protein, interacts with components of the retrotranslocation complex and promotes ER-associated degradation (ERAD). Knockdown of importin ß specifically inhibited the degradation of misfolded ERAD substrates but did not affect turnover of non-ERAD proteasome substrates. Genetic studies and in vitro reconstitution assays demonstrate that importin ß is critically required for ubiquitination of mutant α1-antitrypsin, a luminal ERAD substrate. Furthermore, we show that importin ß cooperates with Ran GTPase to promote ubiquitination and proteasomal degradation of mutant α1-antitrypsin. These results establish an unanticipated role for importin ß in ER protein quality control.


Subject(s)
Endoplasmic Reticulum/metabolism , Mutation , Ubiquitination/physiology , Unfolded Protein Response/physiology , alpha 1-Antitrypsin/metabolism , beta Karyopherins/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/genetics , HEK293 Cells , HeLa Cells , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Transport/physiology , alpha 1-Antitrypsin/genetics , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...