Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Biol Chem ; 299(5): 104675, 2023 05.
Article in English | MEDLINE | ID: mdl-37028761

ABSTRACT

MafA and c-Maf are close members of the Maf transcription factor family and indicators of poor prognosis of multiple myeloma (MM). Our previous study finds that the ubiquitin ligase HERC4 induces c-Maf degradation but stabilizes MafA, and the mechanism is elusive. In the present study, we find that HERC4 interacts with MafA and mediates its K63-linked polyubiquitination at K33. Moreover, HERC4 inhibits MafA phosphorylation and its transcriptional activity triggered by glycogen synthase kinase 3ß (GSK3ß). The K33R MafA variant prevents HERC4 from inhibiting MafA phosphorylation and increases MafA transcriptional activity. Further analyses reveal that MafA can also activate the STAT3 signaling, but it is suppressed by HERC4. Lastly, we demonstrate that lithium chloride, a GSK3ß inhibitor, can upregulate HERC4 and synergizes dexamethasone, a typical anti-MM drug, in inhibiting MM cell proliferation and xenograft growth in nude mice. These findings thus highlight a novel regulation of MafA oncogenic activity in MM and provide the rationale by targeting HERC4/GSK3ß/MafA for the treatment of MM.


Subject(s)
Glycogen Synthase Kinase 3 beta , Maf Transcription Factors, Large , Multiple Myeloma , Polyubiquitin , Ubiquitin-Protein Ligases , Ubiquitination , Animals , Humans , Mice , Cell Proliferation , Dexamethasone/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Lithium Chloride/pharmacology , Maf Transcription Factors, Large/antagonists & inhibitors , Maf Transcription Factors, Large/metabolism , Mice, Nude , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Phosphorylation , Polyubiquitin/metabolism , STAT3 Transcription Factor/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
2.
Acta Pharmacol Sin ; 44(9): 1920-1931, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37055530

ABSTRACT

The cell cycle regulator cyclin D3 (CCND3) is highly expressed in multiple myeloma (MM) and it promotes MM cell proliferation. After a certain phase of cell cycle, CCND3 is rapidly degraded, which is essential for the strict control of MM cell cycle progress and proliferation. In the present study, we investigated the molecular mechanisms regulating CCND3 degradation in MM cells. By utilizing affinity purification-coupled tandem mass spectrometry, we identified the deubiquitinase USP10 interacting with CCND3 in human MM OPM2 and KMS11 cell lines. Furthermore, USP10 specifically prevented CCND3 from K48-linked polyubiquitination and proteasomal degradation, therefore enhancing its activity. We demonstrated that the N-terminal domain (aa. 1-205) of USP10 was dispensable for binding to and deubiquitinating CCND3. Although Thr283 was important for CCND3 activity, it was dispensable for CCND3 ubiquitination and stability modulated by USP10. By stabilizing CCND3, USP10 activated the CCND3/CDK4/6 signaling pathway, phosphorylated Rb, and upregulated CDK4, CDK6 and E2F-1 in OPM2 and KMS11 cells. Consistent with these findings, inhibition of USP10 by Spautin-1 resulted in accumulation of CCND3 with K48-linked polyubiquitination and degradation that synergized with Palbociclib, a CDK4/6 inhibitor, to induce MM cell apoptosis. In nude mice bearing myeloma xenografts with OPM2 and KMS11 cells, combined administration of Spautin-l and Palbociclib almost suppressed tumor growth within 30 days. This study thus identifies USP10 as the first deubiquitinase of CCND3 and also finds that targeting the USP10/CCND3/CDK4/6 axis may be a novel modality for the treatment of myeloma.


Subject(s)
Multiple Myeloma , Mice , Animals , Humans , Cyclin D3 , Multiple Myeloma/metabolism , Mice, Nude , Apoptosis , Deubiquitinating Enzymes , Cell Line, Tumor , Ubiquitin Thiolesterase/metabolism
3.
J Biol Chem ; 298(9): 102314, 2022 09.
Article in English | MEDLINE | ID: mdl-35926709

ABSTRACT

The zinc finger ubiquitin ligase RNF6 has been proposed as a potential therapeutic target in several cancers, but understanding its molecular mechanism of degradation has been elusive. In the present study, we find that RNF6 is degraded via auto-ubiquitination in a manner dependent on its Really Interesting New Gene (RING) domain. We determine that when the RING domain is deleted (ΔRING) or the core cysteine residues in the zinc finger are mutated (C632S/C635S), the WT protein, but not the ΔRING or mutant RNF6 protein, undergoes polyubiquitination. We also identify USP7 as a deubiquitinase of RNF6 by tandem mass spectrometry. We show that USP7 interacts with RNF6 and abolishes its K48-linked polyubiquitination, thereby preventing its degradation. In contrast, we found a USP7-specific inhibitor promotes RNF6 polyubiquitination, degradation, and cell death. Furthermore, we demonstrate the anti-leukemic drug Nilotinib and anti-myeloma drug Panobinostat (LBH589) induce RNF6 K48-linked polyubiquitination and degradation in both multiple myeloma (MM) and leukemia cells. In agreement with our hypothesis on the mode of RNF6 degradation, we show these drugs promote RNF6 auto-ubiquitination in an in vitro ubiquitination system without other E3 ligases. Consistently, reexpression of RNF6 ablates drug-induced MM and leukemia cell apoptosis. Therefore, our results reveal that RNF6 is a RING E3 ligase that undergoes auto-ubiquitination, which could be abolished by USP7 and induced by anti-cancer drugs. We propose that chemical induction of RNF6 auto-ubiquitination and degradation could be a novel strategy for the treatment of hematological malignancies including MM and leukemia.


Subject(s)
Antineoplastic Agents , DNA-Binding Proteins , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Multiple Myeloma , Panobinostat , Ubiquitin-Protein Ligases , Ubiquitination , Zinc Fingers , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cysteine/metabolism , DNA-Binding Proteins/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Multiple Myeloma/drug therapy , Panobinostat/pharmacology , Panobinostat/therapeutic use , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Peptidase 7/metabolism
4.
Microbiol Res ; 256: 126953, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34972023

ABSTRACT

Micropterus salmoides is an economical important species of freshwater-cultured fish, the in-depth knowledge of its immune system is in urgent development to cope with serious infectious diseases. Piscidin is an important antimicrobial peptide (AMP) family existing in almost all teleosts. However, no piscidin has been reported in largemouth bass. In this study, three novel piscidins (MSPiscidin-1, -2, and -3) were firstly identified and characterized from the largemouth bass. The predicted mature peptides of MSPiscidin-1, -2, and -3 (consists of 24, 27, 25 amino acid residues, respectively) all adopted an amphipathic α-helical conformation representative of cationic AMPs that are important for membrane permeabilization and antibacterial activity. MSPiscidin-2 and -3 indeed displayed strong, broad-spectrum, and highly efficient antimicrobial activities in vitro against aquatic pathogens, but MSPiscidin-1 didn't show direct antimicrobial activity. MSPiscidin-2 and -3 killed bacteria mainly by inducing membrane permeabilization, in addition, they also can interact with bacterial genomic DNA, which might influence the DNA replication and transcription. Besides, MSPiscidin-2 and -3 could effectively inhibit the formation of the bacterial biofilm and eliminate the preformed biofilms. In vivo, MSPiscidin-1-3 genes showed an inducible expression pattern in the tested tissues upon Vibrio harveyi infection, which further indicated the key roles of piscidins in innate immunity in largemouth bass. Overall, this study will supplement the understanding of M. salmoides innate immune system and provide candidates for the design of novel peptide antibacterial agents used in aquaculture.


Subject(s)
Antimicrobial Peptides/immunology , Bass , Fish Proteins/immunology , Animals , Bass/immunology , Bass/microbiology , Fresh Water , Immunity, Innate
5.
Acta Pharmacol Sin ; 43(3): 681-691, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33931764

ABSTRACT

The PTEN/AKT/mTOR signaling pathway is frequently dysregulated in non-small cell lung cancer (NSCLC), but the mechanisms are not well-understood. The present study found that the ubiquitin ligase TRIM25 is highly expressed in NSCLC tissues and promotes NSCLC cell survival and tumor growth. Mechanistic studies revealed that TRIM25 binds to PTEN and mediates its K63-linked ubiquitination at K266. This modification prevents the plasma membrane translocation of PTEN and reduces its phosphatase activity therefore accumulating PI(3,4,5)P3. TRIM25 thus activates the AKT/mTOR signaling. Moreover, we found that the antibacterial nitroxoline can activate PTEN by reducing its K63-linked polyubiquitination and sensitizes NSCLC to cisplatin-induced apoptosis. This study thus identified a novel modulation on the PTEN signaling pathway by TRIM25 and provides a potential target for NSCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , DNA-Binding Proteins/metabolism , Lung Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Nitroquinolines/pharmacology , Phosphoric Monoester Hydrolases/physiology , RNA, Small Interfering/metabolism , Ubiquitination/physiology
6.
J Biol Chem ; 297(3): 101088, 2021 09.
Article in English | MEDLINE | ID: mdl-34416231

ABSTRACT

The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) protein is a key player in tumorigenesis of non-small cell lung cancer (NSCLC) and was recently found to be inactivated by tripartite motif containing 25 (TRIM25)-mediated K63-linked polyubiquitination. However, the deubiquitinase (Dub) coordinate TRIM25 in PTEN ubiquitination is still elusive. In the present study, we found that this K63-linked polyubiquitination could be ablated by the ubiquitin-specific protease 10 (USP10) in a screen against a panel of Dubs. We found using coimmununoprecipitation/immunoblotting that USP10 interacted with PTEN and reduced the K63-linked polyubiquitination of PTEN mediated by TRIM25 in NSCLC cells. Moreover, USP10, but not its inactive C424A deubiquitinating mutant or other Dubs, abolished PTEN from K63-linked polyubiquitination mediated by TRIM25. In contrast to TRIM25, USP10 restored PTEN phosphatase activity and reduced the production of the secondary messenger phosphatidylinositol-3,4,5-trisphosphate, thereby inhibiting AKT/mammalian target of rapamycin progrowth signaling transduction in NSCLC cells. Moreover, USP10 was downregulated in NSCLC cell lines and primary tissues, whereas TRIM25 was upregulated. Consistent with its molecular activity, re-expression of USP10 suppressed NSCLC cell proliferation and migration, whereas knockout of USP10 promoted NSCLC cell proliferation and migration. In conclusion, the present study demonstrates that USP10 coordinates TRIM25 to modulate PTEN activity. Specifically, USP10 activates PTEN by preventing its K63-linked polyubiquitination mediated by TRIM25 and suppresses the AKT/mammalian target of rapamycin signaling pathway, thereby inhibiting NSCLC proliferation, indicating that it may be a potential drug target for cancer treatment.


Subject(s)
PTEN Phosphohydrolase/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Deubiquitinating Enzymes/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Signal Transduction/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/physiology , Ubiquitination
7.
Cell Death Dis ; 12(5): 456, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963175

ABSTRACT

Chronic myelogenous leukemia (CML) is a clonal malignancy of hematopoietic stem cells featured with the fusion protein kinase BCR-ABL. To elicit the mechanism underlying BCR-ABL stability, we perform a screen against a panel of deubiquitinating enzymes (DUBs) and find that the ubiquitin-specific protease 7 (USP7) drastically stabilizes the BCR-ABL fusion protein. Further studies show that USP7 interacts with BCR-ABL and blocks its polyubiquitination and degradation. Moreover, USP7 knockdown triggers BCR-ABL degradation and suppresses its downstream signaling transduction. In line with this finding, genetic or chemical inhibition of USP7 leads to BCR-ABL protein degradation, suppresses BCR/ABL signaling, and induces CML cell apoptosis. Furthermore, we find the antimalarial artesunate (ART) significantly inhibits USP7/BCR-ABL interaction, thereby promoting BCR-ABL degradation and inducing CML cell death. This study thus identifies USP7 as a putative Dub of BCR-ABL and provides a rationale in targeting USP7/BCR-ABL for the treatment of CML.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Ubiquitin-Specific Peptidase 7/metabolism , Apoptosis , Cell Proliferation , Humans , Transfection
8.
Cell Commun Signal ; 19(1): 24, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627137

ABSTRACT

BACKGROUND: The oncogenic transcript factor c-Maf is stabilized by the deubiquitinase Otub1 and promotes myeloma cell proliferation and confers to chemoresistance. Inhibition of the Otub1/c-Maf axis is a promising therapeutic target, but there are no inhibitors reported on this specific axis. METHODS: A luciferase assay was applied to screen potential inhibitors of Otub1/c-Maf. Annexin V staining/flow cytometry was applied to evaluate cell apoptosis. Immunoprecipitation was applied to examine protein ubiquitination and interaction. Xenograft models in nude mice were used to evaluate anti-myeloma activity of AVT. RESULTS: Acevaltrate (AVT), isolated from Valeriana glechomifolia, was identified based on a bioactive screen against the Otub1/c-Maf/luciferase system. AVT disrupts the interaction of Otub1/c-Maf thus inhibiting Otub1 activity and leading to c-Maf polyubiquitination and subsequent degradation in proteasomes. Consistently, AVT inhibits c-Maf transcriptional activity and downregulates the expression of its target genes key for myeloma growth and survival. Moreover, AVT displays potent anti-myeloma activity by triggering myeloma cell apoptosis in vitro and impairing myeloma xenograft growth in vivo but presents no marked toxicity. CONCLUSIONS: The natural product AVT inhibits the Otub1/c-Maf axis and displays potent anti-myeloma activity. Given its great safety and efficacy, AVT could be further developed for MM treatment. Video Abstract.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Iridoids/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-maf/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cysteine Endopeptidases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Female , Humans , Iridoids/pharmacology , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism
9.
Blood ; 137(11): 1478-1490, 2021 03 18.
Article in English | MEDLINE | ID: mdl-32842143

ABSTRACT

The oncogenic transcription factor c-Maf has been proposed as an ideal therapeutic target for multiple myeloma (MM), but how to achieve it is still elusive. In the present study, we found the Otub1/c-Maf axis could be a potential target. Otub1, an OTU family deubiquitinase, was found to interact with c-Maf by mass spectrometry. Otub1 abrogates c-Maf K48-linked polyubiquitination, thus preventing its degradation and enhancing its transcriptional activity. Specifically, this deubiquitinating activity depends on its Lys71 and the N terminus but is independent of UBE2O, a known E2 of c-Maf. Otub1 promotes MM cell survival and MM tumor growth. In contrast, silence of Otub1 leads to c-Maf degradation and c-Maf-expressing MM cell apoptosis. Therefore, the Otub1/c-Maf axis could be a therapeutic target of MM. In order to explore this concept, we performed a c-Maf recognition element-driven luciferase-based screen against US Food and Drug Administration-approved drugs and natural products, from which the generic cardiac glycoside lanatoside C (LanC) is found to prevent c-Maf deubiquitination and induces its degradation by disrupting the interaction of Otub1 and c-Maf. Consequently, LanC inhibits c-Maf transcriptional activity, induces c-Maf-expressing MM cell apoptosis, and suppresses MM growth and prolongs overall survival of model mice, but without apparent toxicity. Therefore, the present study identifies Otub1 as a novel deubiquitinase of c-Maf and establishes that the Otub1/c-Maf axis is a potential therapeutic target for MM.


Subject(s)
Antineoplastic Agents/pharmacology , Deubiquitinating Enzymes/metabolism , Multiple Myeloma/drug therapy , Protein Interaction Maps/drug effects , Proto-Oncogene Proteins c-maf/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Discovery , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
10.
Acta Pharmacol Sin ; 42(8): 1338-1346, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33184448

ABSTRACT

Recent studies show that the expression of CCND1, a key factor in cell cycle control, is increased following the progress and deteriotation of glioma and predicts poor outcomes. On the other hand, dysregulated deubiquitinase USP10 also predicts poor prognosis for patients with glioblastoma (GBM). In the present study, we investigated the interplay between CCND1 protein and USP10 in GBM cells. We showed that the expression of CCND1 was significantly higher in both GBM tissues and GBM-derived stem cells. USP10 interacted with CCND1 and prevented its K48- but not K63-linked polyubiquitination in GBM U251 and HS683 cells, which led to increased CCND1 stability. Consistent with the action of USP10 on CCND1, knockdown of USP10 by single-guided RNA downregulated CCND1 and caused GBM cell cycle arrest at the G1 phase and induced GBM cell apoptosis. To implement this finding in the treatment of GBMs, we screened a natural product library and found that acevaltrate (AVT), an active component derived from the herbal plant Valeriana jatamansi Jones was strikingly potent to induce GBM cell apoptosis, which was confirmed by the Annexin V staining and activation of the apoptotic signals. Furthermore, we revealed that AVT concentration-dependently suppressed USP10-mediated deubiquitination on CCND1 therefore inducing CCND1 protein degradation. Collectively, the present study demonstrates that the USP10/CCND1 axis could be a promising therapeutic target for patients with GBMs.


Subject(s)
Cyclin D1/metabolism , Glioblastoma/metabolism , Iridoids/pharmacology , Ubiquitin Thiolesterase/metabolism , Ubiquitination/physiology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/physiology , Glioblastoma/drug therapy , HEK293 Cells , Humans , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination/drug effects
11.
Cell Death Dis ; 11(9): 818, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999280

ABSTRACT

As a deubiqutinase Otub1 stabilizes and promotes the oncogenic activity of the transcription factor c-Maf in multiple myeloma (MM), a malignancy of plasma cells. In the screen for bioactive inhibitors of the Otub1/c-Maf axis for MM treatment, nanchangmycin (Nam), a polyketide antibiotic, was identified to suppress c-Maf activity in the presence of Otub1. By suppressing Otub1, Nam induces c-Maf polyubiquitination and subsequent degradation in proteasomes but does not alter its mRNA level. Consistently, Nam downregulates the expression of CCND2, ARK5, and ITGB7, the downstream genes regulated by c-Maf, and promotes MM cell apoptosis as evidenced by PARP and Caspase-3 cleavage, as well as Annexin V staining. In line with the hypothesis, overexpression of Otub1 partly rescues Nam-induced MM cell apoptosis, and interestingly, when Otub1 is knocked down, Nam-decreased MM cell survival is also partly ablated, suggesting Otub1 is essential for Nam anti-MM activity. Nam also displays potent anti-MM activity synergistically with Doxorubicin or lenalidomide. In the in vivo assays, Nam almost completely suppresses the growth of MM xenografts in nude mice at low dosages but it shows no toxicity. Given its safety and efficacy, Nam has a potential for MM treatment by targeting the Otub1/c-Maf axis.


Subject(s)
Cysteine Endopeptidases/metabolism , Ethers/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-maf/metabolism , Spiro Compounds/therapeutic use , Animals , Cell Line, Tumor , Ethers/pharmacology , Humans , Mice , Spiro Compounds/pharmacology
13.
Biosci Trends ; 14(2): 96-103, 2020 May 21.
Article in English | MEDLINE | ID: mdl-32173687

ABSTRACT

4-anilinoquinazoline-containing inhibitors of the epidermal growth factor receptor (EGFR) are widely used in non-small cell lung cancer patients with mutated EGFR, but they are less effective in multiple myeloma (MM), a fatal malignancy derived from plasma cells. The present study designed a series of novel compounds by conjugating a peroxide bridge to the 4-anilinoquinazoline pharmacophore. Further studies showed that these agents such as 4061 and 4065B displayed potent activity to induce MM cell apoptosis by upregulating pro-apoptotic p53 and Bax while downregulating pro-survival Bcl-2. The mechanistic analysis revealed that both 4061 and 4065B inhibited IGF1-R, AKT and mTOR activation in a concentration dependent manner but had no effects on the expression of their total proteins, suggesting the conjugates of endoperoxide and 4-anilinoquinazoline may exert its anti-myeloma activity by targeting the IGF1-R/AKT/mTOR pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Artemisinins/chemistry , Artemisinins/pharmacology , Artemisinins/therapeutic use , Cell Line, Tumor , Gefitinib/pharmacology , Humans , Multiple Myeloma/pathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
14.
J Biol Chem ; 295(7): 2084-2096, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31822558

ABSTRACT

The Maf proteins, including c-Maf, MafA, and MafB, are critical transcription factors in myelomagenesis. Previous studies demonstrated that Maf proteins are processed by the ubiquitin-proteasome pathway, but the mechanisms remain elusive. This study applied MS to identify MafB ubiquitination-associated proteins and found that the ubiquitin-specific protease USP7 was present in the MafB interactome. Moreover, USP7 also interacted with c-Maf and MafA and blocked their polyubiquitination and degradation. Consistently, knockdown of USP7 resulted in Maf protein degradation along with increased polyubiquitination levels. The action of USP7 thus promoted Maf transcriptional activity as evidenced by luciferase assays and by the up-regulation of the expression of Maf-modulated genes. Furthermore, USP7 was up-regulated in myeloma cells, and it was negatively associated with the survival of myeloma patients. USP7 promoted myeloma cell survival, and when it was inhibited by its specific inhibitor P5091, myeloma cell lines underwent apoptosis. These results therefore demonstrated that USP7 is a deubiquitinase of Maf proteins and promotes MM cell survival in association with Maf stability. Given the significance of USP7 and Maf proteins in myeloma genesis, targeting the USP7/Maf axle is a potential strategy to the precision therapy of MM.


Subject(s)
Maf Transcription Factors, Large/genetics , MafB Transcription Factor/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-maf/genetics , Ubiquitin-Specific Peptidase 7/genetics , Apoptosis/drug effects , Carcinogenesis/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Male , Multiple Myeloma/pathology , Polyubiquitin/genetics , Progression-Free Survival , Proteolysis/drug effects , Thiophenes/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitination/genetics
15.
Acta Pharmacol Sin ; 41(3): 394-403, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31645658

ABSTRACT

RNF6, a RING-type ubiquitin ligase, has been identified as an oncogene in various cancers but its role in multiple myeloma (MM) remains elusive. In the present study we first showed that the expression levels of RNF6 in MM were significantly elevated compared with the bone marrow cells of healthy donors. Overexpression of RNF6 in LP1 and PRMI-8266 MM cell lines promoted cell proliferation, whereas knockdown of RNF6 led to apoptosis of MM cells. Furthermore, we revealed that RNF6, as a ubiquitin ligase, interacted with glucocorticoid receptor (GR) and induced its K63-linked polyubiquitination. Different from current knowledge, RNF6 increased GR stability at both endogenous and exogenous contexts. Such an action greatly promoted GR transcriptional activity, which was confirmed by luciferase assays and by the increased expression levels of prosurvival genes including Bcl-xL and Mcl-1, two typical downstream genes of the GR pathway. Consistent with these findings, ectopic expression of RNF6 in MM cells conferred resistance to dexamethasone, a typical anti-myeloma agent. In conclusion, we demonstrate that RNF6 promotes MM cell proliferation and survival by inducing atypical polyubiquitination to GR, and RNF6 could be a promising therapeutic target for the treatment of MM.


Subject(s)
DNA-Binding Proteins/metabolism , Multiple Myeloma/metabolism , Receptors, Glucocorticoid/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Humans , Molecular Structure , Multiple Myeloma/pathology , Receptors, Glucocorticoid/genetics , Structure-Activity Relationship , Ubiquitination
16.
17.
Acta Pharmacol Sin ; 40(12): 1568-1577, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31197245

ABSTRACT

c-Maf is a critical oncogenic transcription factor that contributes to myelomagenesis. Our previous studies demonstrated that the deubiquitinase USP5 stabilizes c-Maf and promotes myeloma cell proliferation and survival; therefore, the USP5/c-Maf axis could be a potential target for myeloma therapy. As a concept of principle, the present study established a USP5/c-Maf-based luciferase system that was used to screen an FDA-approved drug library. It was found that mebendazole, a typical anthelmintic drug, preferentially induced apoptosis in c-Maf-expressing myeloma cells. Moreover, oral administration of mebendazole delayed the growth of human myeloma xenografts in nude mice but did not show overt toxicity. Further studies showed that the selective antimyeloma activity of mebendazole was associated with the inhibition of the USP5/c-Maf axis. Mebendazole downregulated USP5 expression and disrupted the interaction between USP5 and c-Maf, thus leading to increased levels of c-Maf ubiquitination and subsequent c-Maf degradation. Mebendazole inhibited c-Maf transcriptional activity, as confirmed by both luciferase assays and expression measurements of c-Maf downstream genes. In summary, this study identified mebendazole as a USP5/c-Maf inhibitor that could be developed as a novel antimyeloma agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Mebendazole/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-maf/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyanoacrylates/therapeutic use , Drug Repositioning , Drug Synergism , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/metabolism , Proof of Concept Study , Protein Binding/drug effects , Proto-Oncogene Proteins c-maf/chemistry , Pyridines/therapeutic use , Ubiquitin-Specific Proteases/chemistry , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
18.
J Biol Chem ; 294(12): 4572-4582, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30718275

ABSTRACT

Chemoresistance is a leading obstacle in effective management of advanced prostate cancer (PCa). A better understanding of the molecular mechanisms involved in PCa chemoresistance could improve treatment of patients with PCa. In the present study, using immune histochemical, chemistry, and precipitation assays with cells from individuals with benign or malignant prostate cancer or established PCa cell lines, we found that the oncogenic transcription factor pre-B cell leukemia homeobox-1 (PBX1) promotes PCa cell proliferation and confers to resistance against common anti-cancer drugs such as doxorubicin and cisplatin. We observed that genetic PBX1 knockdown abrogates this resistance, and further experiments revealed that PBX1 stability was modulated by the ubiquitin-proteasomal pathway. To directly probe the impact of this pathway on PBX1 activity, we screened for PBX1-specific deubiquitinases (Dubs) and found that ubiquitin-specific peptidase 9 X-linked (USP9x) interacted with and stabilized the PBX1 protein by attenuating its Lys-48-linked polyubiquitination. Moreover, the USP9x inhibitor WP1130 markedly induced PBX1 degradation and promoted PCa cell apoptosis. The results in this study indicate that PBX1 confers to PCa chemoresistance and identify USP9x as a Dub of PBX1. We concluded that targeting the USP9x/PBX1 axis could be a potential therapeutic strategy for managing advanced prostate cancer.


Subject(s)
Apoptosis , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Prostatic Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Humans , Male , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism
19.
Anticancer Drugs ; 29(10): 995-1003, 2018 11.
Article in English | MEDLINE | ID: mdl-30106753

ABSTRACT

The hedgehog-smoothened (HH/SMO) pathway has been proposed as a potential therapeutic target for hematological malignancies. Our previous studies designed a series of HH inhibitors with novel scaffolds distinctive from vismodegib, the first Food and Drug Administration-approved HH inhibitor for the treatment of basal-cell carcinoma and medulloblastoma. In the present study, we evaluated these HH inhibitors against blood cancers and found that HH78 displayed potent activity in suppressing the HH signaling pathway. HH78 competitively bound to SMO and suppressed the transcriptional activity of GLI by the luciferase reporter gene assay and the measurement of HH/SMO-downregulated genes, including cyclin D2, cyclin E, PTCH1, PTCH2, and GLI. HH78 at low micromolar concentrations induced significant cancer cell apoptosis showed by increased caspase-3 activation, annexin V-staining and downregulated prosurvival proteins, including c-Myc, Bcl-2, Mcl-1, and Bcl-xL. In contrast, vismodegib did not show any effects on these apoptotic events. HH78 also suppressed the activation of the AKT/mTOR pathway, which cross-talks with the HH/SMO pathway. Finally, HH78 inhibited the growth of human leukemia K562 in nude mice xenografts with no overt toxicity. Collectively, the present study identified a novel HH inhibitor with great potential for the treatment of hematological malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Hematologic Neoplasms/drug therapy , Smoothened Receptor/antagonists & inhibitors , Anilides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Dose-Response Relationship, Drug , Down-Regulation/genetics , Hematologic Neoplasms/pathology , Humans , K562 Cells , Mice , Mice, Nude , Pyridines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Front Pharmacol ; 9: 673, 2018.
Article in English | MEDLINE | ID: mdl-29997504

ABSTRACT

Saponins are amphipathic glycosides found in traditional Chinese medicines. In the present study, we isolated a panel of saponins from Paris forrestii (Takht.) H. Li, a unique plant found in Tibet and Yunnan provinces, China. By examining their activities in suppressing acute myeloid leukemia (AML) cell proliferation, total saponins from Paris forrestii (TSPf) displayed more potent activity than individual ones. TSPf induced more than 40% AML cell apoptosis and decreased the viability of all leukemia cell lines. TSPf-induced apoptosis was confirmed by both Annexin V staining and caspase-3 activation. In line with these findings, TSPf downregulated pro-survival proteins Mcl-1, Bcl-xL, and Bcl-2 but upregulated the expression of tumor suppressor proteins p53, p27, Bax, and Beclin 1. The AKT/mTOR signaling pathway is frequently overactivated in various AML cells, and TSPf was found to suppress the activation of both AKT and mTOR, but had no effects on their total protein expression. This was further confirmed by the inactivation of 4EBP-1 and p70S6K, two typical downstream signal molecules in the AKT/mTOR pathway. Moreover, TSPf-inactivated AKT/mTOR signaling was found to be associated with downregulated RNF6, a recently identified oncogene in AML. RNF6 activated AKT/mTOR, and consistently, knockdown of RNF6 led to inactivation of the AKT/mTOR pathway. Furthermore, TSPf suppressed the growth of AML xenografts in nude mice models. Oral administration of TSPf almost fully suppressed tumor growth without gross toxicity. Consistent with the findings in cultured cell lines, TSPf also downregulated RNF6 expression along with inactivated AKT/mTOR signaling in tumor tissues. This study thus demonstrated that TSPf displays potent anti-AML activity by suppressing the RNF6/AKT/mTOR pathway. Given its low toxicity, TSPf could be developed for the treatment of AML.

SELECTION OF CITATIONS
SEARCH DETAIL
...