Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Cancer Drug Resist ; 7: 14, 2024.
Article in English | MEDLINE | ID: mdl-38835349

ABSTRACT

Human epidermal growth factor receptor 3 (HER3), which is part of the HER family, is aberrantly expressed in various human cancers. Since HER3 only has weak tyrosine kinase activity, when HER3 ligand neuregulin 1 (NRG1) or neuregulin 2 (NRG2) appears, activated HER3 contributes to cancer development and drug resistance by forming heterodimers with other receptors, mainly including epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2). Inhibition of HER3 and its downstream signaling, including PI3K/AKT, MEK/MAPK, JAK/STAT, and Src kinase, is believed to be necessary to conquer drug resistance and improve treatment efficiency. Until now, despite multiple anti-HER3 antibodies undergoing preclinical and clinical studies, none of the HER3-targeted therapies are licensed for utilization in clinical cancer treatment because of their safety and efficacy. Therefore, the development of HER3-targeted drugs possessing safety, tolerability, and sensitivity is crucial for clinical cancer treatment. This review summarizes the progress of the mechanism of HER3 in drug resistance, the HER3-targeted therapies that are conducted in preclinical and clinical trials, and some emerging molecules that could be used as future designed drugs for HER3, aiming to provide insights for future research and development of anticancer drugs targeting HER3.

2.
Heliyon ; 10(8): e29567, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38681656

ABSTRACT

XIAP, or the X-linked Inhibitor of Apoptosis Protein, is the most extensively studied member within the IAP gene family. It possesses the capability to impede apoptosis through direct inhibition of caspase activity. Various kinds of cancers overexpress XIAP to enable cancer cells to avoid apoptosis. Consequently, the inhibition of XIAP holds significant clinical implications for the development of anti-tumor medications and the treatment of cancer. In this study, sterigmatocystin, a natural compound obtained from the genus asperigillus, was demonstrated to be able to induce apoptotic and autophagic cell death in liver cancer cells. Mechanistically, sterigmatocystin induces apoptosis by downregulation of XIAP expression. Additionally, sterigmatocystin treatment induces cell cycle arrest, blocks cell proliferation, and slows down colony formation in liver cancer cells. Importantly, sterigmatocystin exhibits a remarkable therapeutic effect in a nude mice model. Our findings revealed a novel mechanism through which sterigmatocystin induces apoptotic and autophagic cell death of liver cancer cells by suppressing XIAP expression, this offers a promising therapeutic approach for treating liver cancer patients.

3.
Article in English | MEDLINE | ID: mdl-37256809

ABSTRACT

Graph convolutional network (GCN) with the powerful capacity to explore graph-structural data has gained noticeable success in recent years. Nonetheless, most of the existing GCN-based models suffer from the notorious over-smoothing issue, owing to which shallow networks are extensively adopted. This may be problematic for complex graph datasets because a deeper GCN should be beneficial to propagating information across remote neighbors. Recent works have devoted effort to addressing over-smoothing problems, including establishing residual connection structure or fusing predictions from multilayer models. Because of the indistinguishable embeddings from deep layers, it is reasonable to generate more reliable predictions before conducting the combination of outputs from various layers. In light of this, we propose an alternating graph-regularized neural network (AGNN) composed of graph convolutional layer (GCL) and graph embedding layer (GEL). GEL is derived from the graph-regularized optimization containing Laplacian embedding term, which can alleviate the over-smoothing problem by periodic projection from the low-order feature space onto the high-order space. With more distinguishable features of distinct layers, an improved Adaboost strategy is utilized to aggregate outputs from each layer, which explores integrated embeddings of multi-hop neighbors. The proposed model is evaluated via a large number of experiments including performance comparison with some multilayer or multi-order graph neural networks, which reveals the superior performance improvement of AGNN compared with the state-of-the-art models.

4.
J Biol Chem ; 299(6): 104723, 2023 06.
Article in English | MEDLINE | ID: mdl-37075843

ABSTRACT

Dysregulation of autophagy has been implicated in the development of many diseases, including cancer. Here, we revealed a novel function of the E3 ubiquitin ligase HRD1 in non-small cell lung carcinoma (NSCLC) metastasis by regulating autophagy. Mechanistically, HRD1 inhibits autophagy by promoting ATG3 ubiquitination and degradation. Additionally, a pro-migratory and invasive factor, MIEN1 (migration and invasion enhancer 1), was found to be autophagically degraded upon HRD1 deficiency. Importantly, expression of both HRD1 and MIEN1 are upregulated and positively correlated in lung tumors. Based on these results, we proposed a novel mechanism of HRD1 function that the degradation of ATG3 protein by HRD1 leads to autophagy inhibition and MIEN1 release, thus promoting NSCLC metastasis. Therefore, our findings provided new insights into the role of HRD1 in NSCLC metastasis and new therapeutic targets for lung cancer treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Ubiquitination , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Autophagy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
5.
J Mol Cell Biol ; 15(2)2023 06 13.
Article in English | MEDLINE | ID: mdl-36806855

ABSTRACT

Tea domain transcription factor 4 (TEAD4) plays a pivotal role in tissue development and homeostasis by interacting with Yes-associated protein (YAP) in response to Hippo signaling inactivation. TEAD4 and YAP can also cooperate with transforming growth factor-ß (TGF-ß)-activated Smad proteins to regulate gene transcription. Yet, it remains unclear whether TEAD4 plays a YAP-independent role in TGF-ß signaling. Here, we unveil a novel tumor suppressive function of TEAD4 in liver cancer via mitigating TGF-ß signaling. Ectopic TEAD4 inhibited TGF-ß-induced signal transduction, Smad transcriptional activity, and target gene transcription, consequently suppressing hepatocellular carcinoma cell proliferation and migration in vitro and xenograft tumor growth in mice. Consistently, depletion of endogenous TEAD4 by siRNAs enhanced TGF-ß signaling in cancer cells. Mechanistically, TEAD4 associates with receptor-regulated Smads (Smad2/3) and Smad4 in the nucleus, thereby impairing the binding of Smad2/3 to the histone acetyltransferase p300. Intriguingly, these negative effects of TEAD4 on TGF-ß/Smad signaling are independent of YAP, as impairing the TEAD4-YAP interaction through point mutagenesis or depletion of YAP and/or its paralog TAZ has little effect. Together, these results unravel a novel function of TEAD4 in fine tuning TGF-ß signaling and liver cancer progression in a YAP-independent manner.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , TEA Domain Transcription Factors , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , YAP-Signaling Proteins
6.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36499382

ABSTRACT

The advent of precision medicine has brought light to the treatment of non-small cell lung cancer (NSCLC), expanding the options for patients with advanced NSCLC by targeting therapy through genetic and epigenetic cues. Tumor driver genes in NSCLC patients have been uncovered one by one, including epidermal growth factor receptor (EGFR), mesenchymal lymphoma kinase (ALK), and receptor tyrosine kinase ROS proto-oncogene 1 (ROS1) mutants. Antibodies and inhibitors that target the critical gene-mediated signaling pathways that regulate tumor growth and development are anticipated to increase patient survival and quality of life. Targeted drugs continue to emerge, with as many as two dozen approved by the FDA, and chemotherapy and targeted therapy have significantly improved patient prognosis. However, resistance due to cancer drivers' genetic alterations has given rise to significant challenges in treating patients with metastatic NSCLC. Here, we summarized the main targeted therapeutic sites of NSCLC drugs and discussed their resistance mechanisms, aiming to provide new ideas for follow-up research and clues for the improvement of targeted drugs.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Protein-Tyrosine Kinases/genetics , Quality of Life , Proto-Oncogene Proteins/genetics , Mutation , Drug Resistance , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
8.
Cell Mol Life Sci ; 79(1): 70, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35018513

ABSTRACT

The histone methyltransferase SETD3 plays critical roles in various biological events, and its dysregulation is often associated with human diseases including cancer. However, the underlying regulatory mechanism remains elusive. Here, we reported that ubiquitin-specific peptidase 27 (USP27) promotes tumor cell growth by specifically interacting with SETD3, negatively regulating its ubiquitination, and enhancing its stability. Inhibition of USP27 expression led to the downregulation of SETD3 protein level, the blockade of the cell proliferation and tumorigenesis of hepatocellular carcinoma (HCC) cells. In addition, we found that USP27 and SETD3 expression is positively correlated in HCC tissues. Notably, higher expression of USP27 and SETD3 predicts a worse survival in HCC patients. Collectively, these data elucidated that a USP27-dependent mechanism controls SETD3 protein levels and facilitates its oncogenic role in liver tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , Histone Methyltransferases/metabolism , Liver Neoplasms/pathology , Ubiquitin-Specific Proteases/metabolism , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , HEK293 Cells , Histone Methyltransferases/genetics , Humans , Liver Neoplasms/mortality , Ubiquitin-Specific Proteases/genetics , Ubiquitination/physiology
9.
Int J Mol Sci ; 22(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34769343

ABSTRACT

MicroRNAs (miRNAs) are non-coding single-stranded RNA molecules encoded by endogenous genes with ~22 nucleotides which are involved in the regulation of post-transcriptional gene expression. Ubiquitination and deubiquitination are common post-translational modifications in eukaryotic cells and important pathways in regulating protein degradation and signal transduction, in which E3 ubiquitin ligases and deubiquitinases (DUBs) play a decisive role. MiRNA and ubiquitination are involved in the regulation of most biological processes, including autophagy. Furthermore, in recent years, the direct interaction between miRNA and E3 ubiquitin ligases or deubiquitinases has attracted much attention, and the cross-talk between miRNA and ubiquitination system has been proved to play key regulatory roles in a variety of diseases. In this review, we summarized the advances in autophagy regulation by crosstalk between miRNA and E3 ubiquitin ligases or deubiquitinases.


Subject(s)
Autophagy , MicroRNAs/genetics , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Humans , Proteolysis , Signal Transduction , Ubiquitin-Protein Ligases/genetics
10.
Biomedicines ; 9(9)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34572328

ABSTRACT

The overexpression of adenosine triphosphate (ATP)-binding cassette (ABC) subfamily B member 1 (ABCB1; P-glycoprotein; MDR1) in some types of cancer cells is one of the mechanisms responsible for the development of multidrug resistance (MDR), which leads to the failure of chemotherapy. Therefore, it is important to inhibit the activity or reduce the expression level of ABCB1 to maintain an effective intracellular level of chemotherapeutic drugs. In this study, we found that rutaecarpine, a bioactive alkaloid isolated from Evodia Rutaecarpa, has the capacity to reverse ABCB1-mediated MDR. Our data indicated that the reversal effect of rutaecarpine was related to the attenuation of the protein level of ABCB1. Mechanistically, we demonstrated that ABCB1 is a newly discovered substrate of E3 ubiquitin ligase membrane-associated RING-CH 8 (MARCH8). MARCH8 can interact with ABCB1 and promote its ubiquitination and degradation. In short, rutaecarpine increased the degradation of ABCB1 protein by upregulating the protein level of MARCH8, thereby antagonizing ABCB1-mediated MDR. Notably, the treatment of rutaecarpine combined with other anticancer drugs exhibits a therapeutic effect on transplanted tumors. Therefore, our study provides a potential chemotherapeutic strategy of co-administrating rutaecarpine with other conventional chemotherapeutic agents to overcome MDR and improve therapeutic effect.

11.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070303

ABSTRACT

Colorectal cancer (CRC) is the third leading malignant tumor in the world, which has high morbidity and mortality. In this study we found that trichodermic acid (TDA), a secondary metabolite isolated from the plant endophytic fungus Penicillium ochrochloronthe with a variety of biological and pharmacological activities, exhibited the antitumor effects on colorectal cancer cells in vitro and in vivo. Our results showed that TDA inhibited the proliferation of colon cancer cells in a dose-dependent manner. TDA induces sustained endoplasmic reticulum stress, which triggers apoptosis through IRE1α/XBP1 and PERK/ATF4/CHOP pathways. In addition, we found that TDA mediated endoplasmic reticulum stress also induces autophagy as a protective mechanism. Moreover, combined treatment of TDA with autophagy inhibitors significantly enhanced its anticancer effect. In conclusion, our results indicated that TDA can induce ER stress and autophagy mediated apoptosis, suggesting that targeting ER stress and autophagy may be an effective strategy for the treatment of CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Colonic Neoplasms , Endoplasmic Reticulum Stress/drug effects , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Mice , Xenograft Model Antitumor Assays
12.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34072267

ABSTRACT

The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell-cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), the activities of which are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins and CDK inhibitors (CKIs). Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell-cycle process via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor proteins and oncoproteins or, inversely, abnormally high accumulation results in cell proliferation deregulation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the UPS machinery regulation of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell-cycle transition, cancer treatment, and the development of anti-cancer drugs.


Subject(s)
Cell Cycle , Neoplasms/metabolism , Animals , Cell Cycle/genetics , Cell Cycle Checkpoints , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Disease Progression , Disease Susceptibility , Gene Expression Regulation , Humans , Neoplasms/etiology , Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
13.
Int J Mol Sci ; 22(11)2021 May 29.
Article in English | MEDLINE | ID: mdl-34072333

ABSTRACT

Mitophagy plays a pro-survival or pro-death role that is cellular-context- and stress-condition-dependent. In this study, we revealed that cyclovirobuxine D (CVB-D), a natural compound derived from Buxus microphylla, was able to provoke mitophagy in lung cancer cells. CVB-D-induced mitophagy potentiates apoptosis by promoting mitochondrial dysfunction. Mechanistically, CVB-D initiates mitophagy by enhancing the expression of the mitophagy receptor BNIP3 and strengthening its interaction with LC3 to provoke mitophagy. Our results further showed that p65, a transcriptional suppressor of BNIP3, is downregulated upon CVB-D treatment. The ectopic expression of p65 inhibits BNIP3 expression, while its knockdown significantly abolishes its transcriptional repression on BNIP3 upon CVB-D treatment. Importantly, nude mice bearing subcutaneous xenograft tumors presented retarded growth upon CVB-D treatment. Overall, we demonstrated that CVB-D treatment can provoke mitophagy and further revealed that the p65/BNIP3/LC3 axis is one potential mechanism involved in CVB-D-induced mitophagy in lung cancer cells, thus providing an effective antitumor therapeutic strategy for the treatment of lung cancer patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Mitophagy/drug effects , Signal Transduction/drug effects , Animals , Biomarkers , Cell Cycle Checkpoints , Cell Line, Tumor , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Immunophenotyping , Lung Neoplasms , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Xenograft Model Antitumor Assays
14.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546413

ABSTRACT

The endoplasmic reticulum (ER) is a highly dynamic organelle in eukaryotic cells, which is essential for synthesis, processing, sorting of protein and lipid metabolism. However, the cells activate a defense mechanism called endoplasmic reticulum stress (ER stress) response and initiate unfolded protein response (UPR) as the unfolded proteins exceed the folding capacity of the ER due to the environmental influences or increased protein synthesis. ER stress can mediate many cellular processes, including autophagy, apoptosis and senescence. The ubiquitin-proteasome system (UPS) is involved in the degradation of more than 80% of proteins in the cells. Today, increasing numbers of studies have shown that the two important components of UPS, E3 ubiquitin ligases and deubiquitinases (DUBs), are tightly related to ER stress. In this review, we summarized the regulation of the E3 ubiquitin ligases and DUBs in ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism , Animals , Apoptosis , Biomarkers , Carrier Proteins , Humans , Protein Binding , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response
15.
Cell Rep ; 33(10): 108487, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33296645

ABSTRACT

Ferroptosis is a recently discovered form of programed cell death caused by the metabolically regulated lipid peroxidation and holds promise for cancer treatment, but its regulatory mechanisms remain elusive. In this study, we observe that lactate-rich liver cancer cells exhibit enhanced resistance to the ferroptotic damage induced by common ferroptosis inducers such as Ras-selective lethal small molecule 3 (RSL3) and Erastin and that the monocarboxylate transporter 1 (MCT1)-mediated lactate uptake could promote ATP production in hepatocellular carcinoma (HCC) cells and deactivate the energy sensor AMP-activated protein kinase (AMPK), leading to the upregulation of sterol regulatory element-binding protein 1 (SREBP1) and the downstream stearoyl-coenzyme A (CoA) desaturase-1 (SCD1) to enhance the production of anti-ferroptosis monounsaturated fatty acids. Additionally, blocking the lactate uptake via hydroxycarboxylic acid receptor 1 (HCAR1)/MCT1 inhibition promotes ferroptosis by activating the AMPK to downregulate SCD1, which may synergize with its acyl-coenzyme A synthetase 4 (ACSL4)-promoting effect to amplify the ferroptotic susceptibility. In vitro and in vivo evidence confirms that lactate regulates the ferroptosis of HCC cells and highlights its translational potential as a therapeutic target for ferroptosis-based tumor treatment.


Subject(s)
Ferroptosis/genetics , Monocarboxylic Acid Transporters/metabolism , Receptors, G-Protein-Coupled/metabolism , Symporters/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Coenzyme A Ligases/metabolism , Female , Ferroptosis/physiology , Humans , Lactic Acid/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Monocarboxylic Acid Transporters/genetics , Neoplasms/metabolism , Receptors, G-Protein-Coupled/genetics , Stearoyl-CoA Desaturase/metabolism , Symporters/genetics
16.
iScience ; 23(8): 101351, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32711345

ABSTRACT

Ubiquitin specific protease 39 (USP39), an ortholog of Sad1p in yeast, is essential for spliceosome assembly during pre-mRNA splicing in human. Although it is known that USP39 is upregulated and plays an oncogenic role in hepatocellular carcinoma (HCC), the underlying mechanism remains unknown. The results of this study demonstrated that USP39 can be acetylated by the histone acetyltransferase MYST1, which is required for its proteasome-mediated degradation by Von Hippel-Lindau protein. In HCC cells, USP39 interacts with and is deacetylated by the lysine deacetylase sirtuin 7 (SIRT7). Notably, the deacetylation of USP39 by SIRT7 promotes its stability and thereby accelerates HCC cell proliferation and tumorigenesis in vitro and in vivo. Our data demonstrated a novel mechanism by which SIRT7 modulates the deacetylation of USP39 to promote HCC development, thus providing an effective anti-tumor therapeutic strategy for HCC.

17.
Oncogene ; 39(22): 4450-4464, 2020 05.
Article in English | MEDLINE | ID: mdl-32361710

ABSTRACT

The NAD-dependent deacetylase sirtuin 1 (SIRT1), a member of the mammalian sirtuin family, plays a pivotal role in deacetylating histone and nonhistone proteins. Recently, it has been reported that SIRT1 is upregulated in various kinds of tumors and is associated with cell growth and metastasis. However, the factors and molecular mechanism regulating its cellular levels remain to be clarified. Here, we reported that the E3 ubiquitin ligase SMURF2 interacts with SIRT1 and mediates its ubiquitination and degradation. Depletion of SMURF2 leads to SIRT1 upregulation and induces the tumor formation and growth of colorectal cancer in vitro and in vivo. Furthermore, we show a negative correlation between SIRT1 and SMURF2 expression in human colorectal cancer. Thus, we propose a novel mechanism of colorectal tumorigenesis via SIRT1 regulation by SMURF2, which could potentially give rise to a new strategy for the treatment of colorectal cancer.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Neoplasm Proteins/metabolism , Sirtuin 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Cell Division , Cell Movement , Colorectal Neoplasms/metabolism , Heterografts , Humans , Lung Neoplasms/secondary , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Proteolysis , Recombinant Proteins/metabolism , Ubiquitin/metabolism , Ubiquitination
18.
Mol Cell Biol ; 40(7)2020 03 16.
Article in English | MEDLINE | ID: mdl-31932479

ABSTRACT

The NAD-dependent histone deacetylase sirtuin 2 (SIRT2) plays critical roles in mitosis and cell cycle progression and recently was shown to suppress tumor growth and to be downregulated in several types of cancers. However, the underlying mechanism of SIRT2 downregulation remains unknown. In this study, using bioinformatics, gene expression profiling, protein overexpression approaches, and cell migration assays, we showed that E3 ubiquitin ligase 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase degradation 1 (HRD1) interacts with SIRT2 and promotes its ubiquitination and degradation. Furthermore, we found that HRD1 deficiency induces SIRT2 upregulation and inhibits the growth and tumor formation of lung cancer cells both in vitro and in vivo Of note, we observed that SIRT2 expression is downregulated in human lung cancer and also negatively correlates with HRD1 expression in these cancers. Additionally, we found that patients with lung adenocarcinoma having lower HRD1 or higher SIRT2 expression levels tend to survive longer. On the basis of these results, we propose a mechanism of lung tumorigenesis that involves HRD1-mediated downregulation of SIRT2 and suggest that interventions targeting HRD1 activity could be a potential therapeutic strategy to treat patients with lung cancer.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinogenesis/genetics , Lung Neoplasms/pathology , Sirtuin 2/metabolism , Ubiquitin-Protein Ligases/metabolism , A549 Cells , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation/genetics , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology , Up-Regulation/genetics
19.
Cancer Lett ; 448: 40-51, 2019 04 28.
Article in English | MEDLINE | ID: mdl-30742943

ABSTRACT

Upregulation of special AT-rich sequence-binding protein-1 (SATB1) has been suggested to promote tumor growth and metastasis. However, the factors governing its cellular levels remain unclear. Here, we report that ubiquitin-specific peptidase 47 (USP47), a member of the deubiquitinating enzymes family, interacts with SATB1 and mediates its deubiquitination and stability. USP47 deficiency impairs transcriptional activity of SATB1 target genes and inhibits colon cancer cell proliferation, migration, and tumorigenesis in a mouse model of colon cancer. Furthermore, we identified SMURF2 as an E3 ubiquitin ligase that promotes SATB1 degradation by upregulating its ubiquitination, and its deficiency promotes colon cancer cell proliferation and SATB1 target gene transcription. SMURF2 is negatively regulated by USP47, and USP47 depletion sensitizes colon cancer cells to 5-FU treatment-induced apoptosis. Taken together, our findings provide a ubiquitination-related mechanistic link to USP47, SMURF2, and SATB1 and suggest that USP47 might be targeted for colon cancer treatment when SATB1 is overexpressed.


Subject(s)
Cell Proliferation/physiology , Colonic Neoplasms/metabolism , Matrix Attachment Region Binding Proteins/physiology , Ubiquitin Thiolesterase/physiology , Ubiquitin-Protein Ligases/physiology , Ubiquitination/physiology , Animals , Humans , Mice , Up-Regulation
20.
J Asian Nat Prod Res ; 21(9): 851-858, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30129376

ABSTRACT

Three new 3,4,6-trisubstituted α-pyrone derivatives, namely 6-(2'R-hydroxy-3'E,5'E-diene-1'-heptyl)-4-hydroxy-3-methyl-2H-pyran-2-one (1), 6-(2'S-hydroxy-5'E-ene-1'-heptyl)-4-hydroxy-3-methyl-2H-pyran-2-one (2), and 6-(2'S-hydroxy-1'-heptyl)-4 -hydroxy-3-methyl-2H-pyran-2-one (3), together with one known compound trichodermic acid (4), were isolated from the solid-substrate fermentation culture of Penicillium ochrochloronthe associated the roots of Taxus media. Compounds 1-4 displayed the antimicrobial activity selectively against tested fungal and bacterial strains with minimum inhibitory concentration (MIC) values ranging from 12.5 to 100 µg/ml. Furthermore, we found that only compound 4 exhibited moderate cytotoxicity against five human cancer cells (A549, LN229, MGC, LOVO, and MDA231) with IC50 values of 51.45, 23.43, 39.16, 46.97, and 42.85 µg/ml, respectively.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Penicillium/chemistry , Pyrones/chemistry , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemistry , Antineoplastic Agents/chemistry , Bacteria/drug effects , Cell Line, Tumor , Cell Survival , Fungi/drug effects , Humans , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL