scispace - formally typeset
Search or ask a question
Topic

MG132

About: MG132 is a research topic. Over the lifetime, 1499 publications have been published within this topic receiving 56589 citations. The topic is also known as: MG132 & Z-Leu-leu-leu-al.


Papers
More filters
Journal ArticleDOI
TL;DR: A novel mechanism for proteasome inhibitor-mediated radiosensitization in which c-Myc down-regulation is involved is revealed in the tumor tissues followed by combinational treatment of MG132 and IR.
Abstract: In this study, the effect of MG132 (carbobenzoxyl-leucinyl-leucinyl-leucinal-H) at a low dose on radiotherapeutic efficacy and its accurate mechanism of radiosensitization were investigated in human non-small cell lung cancer. The effect of MG132 on ionizing radiation (IR)-induced cytotoxicity, cell proliferation and survival of A549 cells was evaluated. The protein expression modulated by MG132 and IR were inspected by Western blot analysis. To determine in vivo radiotherapeutic efficacy, tumor growth delay was analyzed in a A549 tumor-bearing xenograft mouse model after single or repeated treatment of MG132 and/or IR. Induction of apoptosis and change of c-Myc expression in the tumor tissue was explored by histological analysis. MG132 at a non-toxic dose enhanced the radiation-induced cytotoxicity of A549 cells, accompanying a significant decrease of c-Myc expression. Suppression of c-Myc expression by small interfering RNA (siRNA) displayed enhancement of radiosensitivity similarly to MG132 treatment. Tumor growth in the xenograft mice was markedly delayed by systemic administration of MG132 combined with IR. In vivo down-regulation of c-Myc and increased induction of apoptosis were simultaneously observed in the tumor tissues followed by combinational treatment of MG132 and IR. The results reveal a novel mechanism for proteasome inhibitor-mediated radiosensitization in which c-Myc down-regulation is involved.

24 citations

Journal Article
TL;DR: A dose-dependent, opposite effect of proteasome inhibition on tau processing is demonstrated, as well as the participation of prote asome- dependent, ubiquitination-independent mechanisms in tau degradation and aggregation, and the promotion of tau aggregation by caspase-mediated tAU degradation/truncation.
Abstract: Tauopathies are characterized by accumulation of filamentous tau aggregates. These aggregates can be recapitulated in transfectant M1C overproducing wild-type human brain tau 4R0N via the tetracycline off (TetOff) inducible expression mechanism. To determine the contribution of proteasomes to tau degradation and aggregation, we exposed M1C cells to epoxomicin (Epx; 2-50 nM) or MG132 (0.5 microM) on the 3(rd) or 4(th) day of a 5-day TetOff induction and demonstrated a reduction of proteasomal activity. Cultures treated with 2 nM Exp showed accumulation of full-length tau without affecting ubiquitin and beta-catenin immunoblotting profiles. In contrast, cells treated with 10, 50 nM Epx or MG132 displayed changes in ubiquitin or beta-catenin immunoblotting profiles and extensive tau degradation/truncation. The increase of tau degradation/truncation was accompanied with accumulation of oligomers and sarkosyl-insoluble aggregates of tau, augmented thioflavin-binding and activation of caspases and calpains. Truncated, oligomeric and sarkosyl-insoluble tau derivatives appeared with caspase-specific cleavage and their production was diminished when pretreated with a pan-caspase inhibitor. The results demonstrate (i) a dose-dependent, opposite effect of proteasome inhibition on tau processing, (ii) the participation of proteasome-dependent, ubiquitination-independent mechanisms in tau degradation and aggregation, and (iii) the promotion of tau aggregation by caspase-mediated tau degradation/truncation.

24 citations

Journal ArticleDOI
TL;DR: This work identified 498 proteins to be significantly co-modified by SUMO and ubiquitin upon MG132 treatment, revealing a highly complex interconnected network of crosstalk among different PTMs, including many enzymatic components of PTM machinery, involved in SUMOylation and ubiquitylation but also phosphorylation, methylation and acetylation.

24 citations

Journal ArticleDOI
TL;DR: Investigation of how these two systems communicate and coordinate with each other in RPE cells for eliminating intracellular misfolded and damaged proteins indicates that the UPP and the ALP are interrelated and that dysfunction of the ALP would also result in dysfunction ofThe U PP and severely compromise the capacity of eliminating mis folded and other forms of damaged proteins.
Abstract: Background: The accumulation of damaged or misfolded proteins in retinal pigment epithelial (RPE) cells was considered a contributing factor for RPE dysfunction in age-related macular degeneration (AMD). The ubiquitinproteasome pathway (UPP) and the autophagy-lysosome pathway (ALP) are the two major proteolytic systems for clearance of misfolded or damaged proteins. Objective: The aim is to investigate how these two systems communicate and coordinate with each other in RPE cells for eliminating intracellular misfolded and damaged proteins. Methods: Cultured ARPE-19 cells were treated with proteasome inhibitor MG132 and lysosomotropic agent chloroquine (CQ), respectively. The levels and cellular distributions of ubiquitinated proteins, LC3-I, LC3-II, LAMP1 and p62 were analyzed by Western blotting and immunofluorescence. Proteasome activity was determined using Suc-LLVY-AMC as a substrate. Results: The level of ubiquitinated protein aggregations was significantly increased after the treatment of MG132 in RPE cells. The levels of LC3-I, LC3-II and LAMP1 increased in MG132 treated cells. The levels of γ-tubulin and p62 also increased in MG132 treated cells, suggesting that inhibition of the UPP up-regulates autophagy-lysosome pathway. Inhibition of lysosomal activity with CQ also increased the levels of high mass ubiquitin conjugates, LC3-II and p62. In addition, proteasome activity was compromised upon prolonged lysosomal inhibition. Conclusions: These data indicate that the UPP and the ALP are interrelated and that dysfunction of the ALP would also result in dysfunction of the UPP and severely compromise the capacity of eliminating misfolded and other forms of damaged proteins.

24 citations

Journal Article
TL;DR: Externally ASB10 protein expression in human trabecular meshwork (HTM) cells is investigated to provide the first clues to the biologic function of this protein, and evidence suggests that ASB 10 may play a role in ubiquitin-mediated degradation pathways in TM cells.
Abstract: Purpose: Ankyrin repeat and suppressor of cytokine signaling (SOCS) box containing protein-10 (ASB10) was recently identified as a gene that causes primary open-angle glaucoma. Here, we investigated endogenous ASB10 protein expres sion in human trabecular meshwork (HTM) cells to provide the first clues to the biologic function of this protein. Methods: Primary HTM cells were cultured and immunostained with anti-ASB10 and various biomarkers of the ubiquitin-mediated proteasomal and autophagy-lysosomal degradation pathways. Cells were imaged with confocal and high-resolution structured illumination microscopy. Colocalization was quantified using Imaris Bitplane software, which generated a Pearson’s correlation coefficient value. Coimmunoprecipitation of ASB10-transfected cells was performed. Results: Immunofluorescence and confocal analysis showed that ASB10 was localized in intracellular structures in HTM cells. Two populations were observed: small, spherical vesicles and larger, less abundant structures. In the ASB10silenced cells, the number of large structures was significantly decreased. ASB10 partially colocalized with biomarkers of the ubiquitin-mediated proteasomal pathway including ubiquitin and the α4 subunit of the 20S proteasome. However, ASB10 itself was not ubiquitinated. ASB10 also colocalized with numerous biomarkers of specific autophagic structures: aggresomes (histone deacetylase 6 [HDAC6] and heat shock protein 70 [HSP70]), autophagosomes (light chain 3 [LC3] and p62), amphisomes (Rab7), and lysosomes (lysosomal-associated membrane protein 1 [LAMP1]). Pearson coeffi cients indicated strong colocalization of large ASB10-stained structures with the α4 subunit of the 20S proteasome, K48 and K63-linked ubiquitin antibodies, p62, HSP70, and HDAC6 (Pearson’s range, 0.59–0.82). Coimmunoprecipitation assays showed a positive interaction of ASB10 with HSP70 and with the α4 subunit of the 20S proteasome. Super-resolution structured illumination confocal microscopy suggested that the smaller ASB10-stained vesicles aggregated into the larger structures, which resembled aggresome-like induced structures. Treatment of HTM cells with an autophagy activator (MG132) or inhibitors (wortmannin, bafilomycin A1) significantly increased and decreased the number of small ASB10-stained vesicles, respectively. No discernible differences in the colocalization of large ASB10-stained structures with ubiquitin or HDAC6 were observed between dermal fibroblasts derived from a normal individual and a patient with primary open-angle glaucoma carrying a synonymous ASB10 mutation. Conclusions: Our evidence suggests that ASB10 may play a role in ubiquitin-mediated degradation pathways in TM cells.

24 citations


Network Information
Related Topics (5)
Signal transduction
122.6K papers, 8.2M citations
90% related
Cell culture
133.3K papers, 5.3M citations
90% related
Gene expression
113.3K papers, 5.5M citations
88% related
Transcription factor
82.8K papers, 5.4M citations
88% related
Regulation of gene expression
85.4K papers, 5.8M citations
87% related
Performance
Metrics
No. of papers in the topic in previous years
YearPapers
202386
202270
202157
202059
201962
201848