Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
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
2.
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
3.
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
4.
PLoS One ; 15(11): e0241540, 2020.
Article in English | MEDLINE | ID: mdl-33152004

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) has spread to the world. Whether there is an association between lifestyle behaviors and the acquisition of COVID-19 remains unclear. METHODS: In this case-control study, we recruited 105 patients with SARS-CoV-2 infection as a case group from the Wuhan Tongji Hospital (Wuhan, China). For each case two control subjects were recruited. Participants were randomly selected from communities in Wuhan and matched for sex, age (± 2yrs), and pre-existing comorbidities (hypertension and diabetes). RESULTS: A total of 105 patients diagnosed with COVID-19 and 210 controls were included. Compared with control group, the case group had higher proportions of lack of sleep (30.5% vs. 14.8%, P = 0.001) and increased physical activities (56.2% vs. 32.9%, P < 0.001). And patients in the case group were more likely to have alopecia (28.6% vs. 10.0%, P < 0.001) than people from the control group. Overall, we found that lack of sleep [adjusted odds ratio (OR) 1.56, 95% confidence interval (CI) 1.03-2.39)], physical activities (≥ 5 times a week) (adjusted OR 2.05, 95%CI 1.39-3.02) and alopecia (adjusted OR 1.73, 95%CI 1.13-2.66) were independent risk factors for COVID-19 infection. Conversely, low-dose alcohol intake (<100g alcohol per week), hand hygiene, and fruits intake (daily) were significantly associated with a decrease in morbidity. CONCLUSIONS: Individual lifestyle behaviors and health status can affect the occurrence of COVID-19.


Subject(s)
Coronavirus Infections/epidemiology , Health Status , Life Style , Pneumonia, Viral/epidemiology , Adult , Aged , Alcohol Drinking , Betacoronavirus , COVID-19 , Case-Control Studies , China , Comorbidity , Exercise , Female , Humans , Logistic Models , Male , Middle Aged , Pandemics , Risk Factors , SARS-CoV-2 , Sleep
5.
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
6.
Medicine (Baltimore) ; 99(18): e20175, 2020 May.
Article in English | MEDLINE | ID: mdl-32358406

ABSTRACT

RATIONALE: The cystic features of the novel coronavirus disease 2019 (COVID-19) found on computed tomography (CT) have not yet been reported in the published literature. We report the cystic chest CT findings of 2 patients confirmed to have COVID-19-related pneumonia. PATIENT CONCERNS: A 38-year-old man and a 35-year-old man diagnosed with severe COVID-19 pneumonia were admitted to the intensive care unit. DIAGNOSES: Chest CT findings showed multiple cysts in ground-glass opacities (bilaterally) with/without pneumothorax. The cysts had a smooth inner wall. INTERVENTIONS: The patients continued to be given oxygen by mask and received antitussive, phlegm-dispelling treatment. OUTCOMES: At follow up, there was a reduction in the number of multiple cystic lesions on CT. To date, 1 patient was discharged from hospital, while the other had been transferred to the rehabilitation department. LESSONS: COVID-19 may independently result in pulmonary cyst formation and pneumothorax; the application of a ventilator may be another causative factor.


Subject(s)
Chest Pain/etiology , Coronavirus Infections/diagnosis , Coronavirus , Cysts/diagnostic imaging , Dyspnea/etiology , Lung/diagnostic imaging , Pneumonia, Viral/diagnosis , Tomography, X-Ray Computed/methods , Adult , Betacoronavirus , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/complications , Coronavirus Infections/diagnostic imaging , Humans , Lung/pathology , Male , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/diagnostic imaging , Pneumothorax/pathology , SARS-CoV-2 , Thorax
7.
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
8.
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
9.
10.
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
11.
J Biol Chem ; 293(16): 5847-5859, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29467225

ABSTRACT

TMEPAI (transmembrane prostate androgen-induced protein, also called prostate transmembrane protein, androgen-induced 1 (PMEPA1)) is a type I transmembrane (TM) protein, but its cellular function is largely unknown. Here, studying factors influencing the stability of c-Maf, a critical transcription factor in multiple myeloma (MM), we found that TMEPAI induced c-Maf degradation. We observed that TMEPAI recruited NEDD4 (neural precursor cell expressed, developmentally down-regulated 4), a WW domain-containing ubiquitin ligase, to c-Maf, leading to its degradation through the proteasomal pathway. Further investigation revealed that TMEPAI interacts with NEDD4 via its conserved PY motifs. Alanine substitution or deletion of these motifs abrogated the TMEPAI complex formation with NEDD4, resulting in failed c-Maf degradation. Functionally, TMEPAI suppressed the transcriptional activity of c-Maf. Of note, increased TMEPAI expression was positively associated with the overall survival of MM patients. Moreover, TMEPAI was down-regulated in MM cells, and re-expression of TMEPAI induced MM cell apoptosis. In conclusion, this study highlights that TMEPAI decreases c-Maf stability by recruiting the ubiquitin ligase NEDD4 to c-Maf for proteasomal degradation. Our findings suggest that the restoration of functional TMEPA1 expression may represent a promising complementary therapeutic strategy for treating patients with MM.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Multiple Myeloma/metabolism , Proteolysis , Proto-Oncogene Proteins c-maf/metabolism , Humans , Multiple Myeloma/pathology , Nedd4 Ubiquitin Protein Ligases/metabolism , Protein Interaction Maps , Ubiquitination
12.
Cell Death Dis ; 8(9): e3058, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28933784

ABSTRACT

The deubiquitinase USP5 stabilizes c-Maf, a key transcription factor in multiple myeloma (MM), but the mechanisms and significance are unclear. In the present study, USP5 was found to interact with c-Maf and prevented it from degradation by decreasing its polyubiquitination level. Specifically, the 308th and 347th lysine residues in c-Maf were critical for USP5-mediated deubiquitination and stability. There are five key domains in the USP5 protein and subsequent studies revealed that the cryptic ZnF domain and the C-box domain interacted with c-Maf but the UBA1/UBA2 domain partly increased its stability. Notably, MafA and MafB are also members of the c-Maf family, however, USP5 failed to deubiquitinate MafA, suggesting its substrate specificity. In the functional studies, USP5 was found to promoted the transcriptional activity of c-Maf. Consistent with the high level of c-Maf protein in MM cells, USP5 was also highly expressed. When USP5 was knocked down, c-Maf underwent degradation. Interestingly, USP5 silence led to apoptosis of MM cells expressing c-Maf but not MM cells lacking c-Maf, indicating c-Maf is a key factor in USP5-mediated MM cell proliferation and survival. Consistent with this finding, WP1130, an inhibitor of several Dubs including USP5, suppressed the transcriptional activity of c-Maf and induced MM cell apoptosis. When c-Maf was overexpressed, WP1130-induced MM cell apoptosis was abolished. Taken together, these findings suggest that USP5 regulates c-Maf stability and MM cell survival. Targeting the USP5/c-Maf axis could be a potential strategy for MM treatment.


Subject(s)
Apoptosis , Endopeptidases/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Proteolysis , Proto-Oncogene Proteins c-maf/metabolism , Apoptosis/genetics , Endopeptidases/chemistry , Gene Knockdown Techniques , HEK293 Cells , Humans , Lysine/metabolism , Molecular Targeted Therapy , Multiple Myeloma/genetics , Polyubiquitin/metabolism , Protein Binding , Protein Domains , Protein Stability , Structure-Activity Relationship , Transcription, Genetic , Ubiquitination
13.
J Pharmacol Sci ; 134(4): 197-202, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28779993

ABSTRACT

S14161 is a pan-Class I PI3K inhibitor that induces blood cancer cell death, but its mechanism is largely unknown. In the present study, we evaluated the role of S14161 in autophagy, an emerging event in cell destination. Multiple myeloma cell lines RPMI-8226, OPM2, KMS11 and leukemia cell line K562 were treated with S14161. The results showed that S14161 induced autophagy as demonstrated by increased LC3-II and decreased p62, which were prevented by autophagy inhibitors including 3-methyladenine and bafilomycin A1. Mechanistic studies showed that S14161 had no effects on Vps34 expression, but increased Beclin 1 and decreased Bcl-2, two major regulators of autophagy. Furthermore, S14161 dissociated the Beclin 1/Bcl-2 complex and enhanced the formation of Beclin 1/Vps34 complex. Moreover, S14161 inhibited the mTORC1 signaling transduction. S14161 downregulated activation of mTOR and its two critical targets 4E-BP1 and p70S6K, suggesting S14161 inhibited protein synthesis. Taken together, these results demonstrated that Class I PI3K regulates autophagy by modulating protein synthesis and the Beclin 1 signaling pathway. This finding helps understanding the roles of PI3K in autophagy and cancer treatment.


Subject(s)
Autophagy/drug effects , Beclin-1/metabolism , Benzopyrans/pharmacology , Class III Phosphatidylinositol 3-Kinases/metabolism , Hematologic Neoplasms/pathology , Leukemia/pathology , Multiple Myeloma/pathology , Phosphoinositide-3 Kinase Inhibitors , Beclin-1/genetics , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/genetics , Gene Expression/drug effects , Humans , K562 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinases/physiology , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects
14.
J Hematol Oncol ; 10(1): 132, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28673317

ABSTRACT

BACKGROUND: UBE2O is proposed as a ubiquitin-conjugating enzyme, but its function was largely unknown. METHODS: Mass spectrometry was applied to identify c-Maf ubiquitination-associated proteins. Immunoprecipitation was applied for c-Maf and UBE2O interaction. Immunoblotting was used for Maf protein stability. Luciferase assay was used for c-Maf transcriptional activity. Lentiviral infections were applied for UBE2O function in multiple myeloma (MM) cells. Flow cytometry and nude mice xenografts were applied for MM cell apoptosis and tumor growth assay, respectively. RESULTS: UBE2O was found to interact with c-Maf, a critical transcription factor in MM, by the affinity purification/tandem mass spectrometry assay and co-immunoprecipitation assays. Subsequent studies showed that UBE2O mediated c-Maf polyubiquitination and degradation. Moreover, UBE2O downregulated the transcriptional activity of c-Maf and the expression of cyclin D2, a typical gene modulated by c-Maf. DNA microarray revealed that UBE2O was expressed in normal bone marrow cells but downregulated in MGUS, smoldering MM and MM cells, which was confirmed by RT-PCR in primary MM cells, suggesting its potential role in myeloma pathophysiology. When UBE2O was restored, c-Maf protein in MM cells was significantly decreased and MM cells underwent apoptosis. Furthermore, the human MM xenograft in nude mice showed that re-expression of UBE2O delayed the growth of myeloma xenografts in nude mice in association with c-Maf downregulation and activation of the apoptotic pathway. CONCLUSIONS: UBE2O mediates c-Maf polyubiquitination and degradation, induces MM cell apoptosis, and suppresses myeloma tumor growth, which provides a novel insight in understanding myelomagenesis and UBE2O biology.


Subject(s)
Apoptosis , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Animals , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Mice, Nude , Multiple Myeloma/pathology , Protein Interaction Maps , Protein Stability , Proto-Oncogene Proteins c-maf/analysis , Ubiquitin-Conjugating Enzymes/analysis
15.
Oncotarget ; 8(12): 20103-20112, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28223545

ABSTRACT

Ring finger protein 6 (RNF6) is a key oncogene in both prostate cancer and leukemia, but its role is elusive in breast cancer. In the present study, we found that RNF6 was overexpressed in more than 70% of breast cancer tissues and it was associated with overall survival. RNF6 increased breast cancer cell proliferation, migration and reduced cell sensitivity to doxorubicin. Further studies showed that RNF6 was closely associated with increased expression of estrogen receptor, a critical factor in the development of breast cancers. RNF6 was found to induce ERα expression and increased its stability. In doxorubicin-resistant breast cancer cells, RNF6 was found to be elevated in association with increased ERα and anti-apoptotic Bcl-xL, but not pro-apoptotic Bim-1. In consistence with this finding, overexpression of ERα led to increased Bcl-xL but had no effects on Bim-1. Therefore, this study demonstrated that there exists an RNF6/ERα/Bcl-xL axle in breast cancer which promotes cancer cell proliferation and survival. Targeting the RNF6/ERα/Bcl-xL axle could be a promising strategy in the treatment of breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Cell Proliferation , DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/metabolism , bcl-X Protein/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Case-Control Studies , DNA-Binding Proteins/genetics , Estrogen Receptor alpha/genetics , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Tumor Cells, Cultured , bcl-X Protein/genetics
16.
Int J Cardiol ; 227: 589-594, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27836304

ABSTRACT

BACKGROUND: This study examines the clinical utility of plasma neutrophil gelatinase-associated lipocalin (NGAL) as an indicator of myocardial dysfunction and mortality in severe sepsis and septic shock. METHODS: We designed a prospective cohort study in an intensive care unit, and 53 patients with severe sepsis or septic shock were included. Data were used to determine a relationship between NGAL and the development of myocardial dysfunction and mortality. These associations were determined by the Mann-Whitney test, multiple logistic regression, plotting the receiver operating characteristic (ROC) curve, Kaplan-Meier curves and Spearman test. RESULTS: The High NGAL group had higher need for inotropic/vasopressor support (92% vs. 52%, p=0.0186), higher incidence of regional wall motion abnormalities (46% vs. 13%, p=0.0093), higher B-type natriuretic peptide (BNP) level (p=0.0197), higher cardiac troponin I (cTnI) level (p=0.0016), lower ejection fraction (EF) (p<0.0001) and higher mortality (p=0.0262) compared to the Low NGAL group. Patients with High NGAL were more likely to manifest electrocardiogram (ECG) abnormalities (p=0.042) and demonstrate clinical myocardial dysfunction (p=0.0186) as evidenced by clinical or radiological evidence of pulmonary edema as compared to those with Low NGAL group. NGAL, BNP, Acute Physiology and Chronic Health Evaluation (APACHE) II score, cTnI, and PaO2/FIO2 ratio were independent predictor of death by multiple logistic regression analysis. The area under the ROC curve showed that plasma NGAL as a predictor of death in septic shock was significant. CONCLUSIONS: High plasma NGAL correlates with high mortality and myocardial dysfunction in severe sepsis and septic shock.


Subject(s)
Cardiomyopathies/blood , Lipocalin-2/blood , Sepsis/blood , Shock, Septic/blood , Aged , Biomarkers/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/mortality , Cohort Studies , Electrocardiography/mortality , Electrocardiography/trends , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mortality/trends , Predictive Value of Tests , Prospective Studies , Sepsis/diagnostic imaging , Sepsis/mortality , Shock, Septic/diagnostic imaging , Shock, Septic/mortality
17.
Oncotarget ; 7(46): 75539-75550, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27705908

ABSTRACT

The activated JAK2-STAT3 signaling pathway is a high risk factor for multiple myeloma (MM), a fatal malignancy of plasma cells. In the present study, SC09, a potential inhibitor of cholesterol absorption, was identified in a STAT3-targeted drug screen. SC09 suppressed the activation of STAT3 in a time-course and concentration-dependent manner but did not affect its family members STAT1 and STAT5. SC09 inhibited STAT3 transcriptional activity and downregulated the expression of STAT3-regulated genes. Further studies showed that SC09 selectively inhibited JAK2 activation but not other kinases including c-Src, ERK, p38 and mTOR that are all associated with STAT3 activation. Moreover, SC09 obviously induced MM cell death in vitro and delayed MM tumor growth in vivo. SC09-induced MM cell death was dependent on the endogenous STAT3 status, and this effect could be attenuated by enforced expression of STAT3. All the results collectively indicated that SC09 blocks the JAK2-STAT3 signaling thus displaying anti-MM activity. Given its well tolerance and anti-MM potency, SC09 is credited for further investigation as a promising drug for MM treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cholesterol/metabolism , Janus Kinase 2/metabolism , Lipid Metabolism/drug effects , Multiple Myeloma/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/pharmacology , Humans , Janus Kinase 2/chemistry , Mice , Mice, Nude , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Phosphorylation , STAT3 Transcription Factor/genetics , Transcriptional Activation/drug effects
18.
J Biol Chem ; 291(18): 9617-28, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26971355

ABSTRACT

RNF6 is a little-studied ring finger protein. In the present study, we found that RNF6 was overexpressed in various leukemia cells and that it accelerated leukemia cell proliferation, whereas knockdown of RNF6 delayed tumor growth in xenografts. To find out the mechanism of RNF6 overexpression in leukemia, we designed a series of truncated constructs of RNF6 regulatory regions in the luciferase reporter system. The results revealed that the region between -144 and -99 upstream of the RNF6 transcription start site was critical and that this region contained a PBX1 recognition element (PRE). PBX1 modulated RNF6 expression by binding to the specific PRE. When PRE was mutated, RNF6 transcription was completely abolished. Further studies showed that PBX1 collaborated with PREP1 but not MEIS1 to modulate RNF6 expression. Moreover, RNF6 expression could be suppressed by doxorubicin, a major anti-leukemia agent, via down-regulating PBX1. This study thus suggests that RNF6 overexpression in leukemia is under the direction of PBX1 and that the PBX1/RNF6 axis can be developed as a novel therapeutic target of leukemia.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Leukemic , Leukemia/metabolism , Proto-Oncogene Proteins/biosynthesis , Response Elements , Animals , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , HL-60 Cells , Heterografts , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , K562 Cells , Leukemia/drug therapy , Leukemia/genetics , Leukemia/pathology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/genetics
19.
Sci Rep ; 5: 12809, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26329846

ABSTRACT

The mammalian target of rapamycin (mTOR) is extensively involved in multiple myeloma (MM) pathophysiology. In the present study, we reported a novel small molecule SC06 that induced MM cell apoptosis and delayed MM xenograft growth in vivo. Oral administration of SC06 to mice bearing human MM xenografts resulted in significant inhibition of tumor growth at doses that were well tolerated. Mechanistic studies revealed that SC06 selectively inhibited the mTOR signaling pathway but had no effects on other associated kinases, such as AKT, ERK, p38, c-Src and JNK. Further studies showed that SC06-decreased mTOR activation was associated with the downregulation of Raptor, a key component of the mTORC1 complex. SC06 also suppressed the phosphorylation of 4E-BP1 and P70S6K, two typical substrates in the mTORC1 signaling pathway. Notably, expression of Raptor, phosphorylation of mTOR and phosphorylated 4E-BP1 was also decreased in the tumor tissues from SC06-treated mice, which was consistent with the cellular studies. Therefore, given the potency and low toxicity, SC06 could be developed as a potential anti-MM drug candidate by disrupting the mTOR signaling.


Subject(s)
Aminopyridines/therapeutic use , Hydrazones/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aminopyridines/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Female , HEK293 Cells , Humans , Hydrazones/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice, Nude , Multiple Myeloma/pathology , Multiprotein Complexes/metabolism , Proteasome Endopeptidase Complex/metabolism , Regulatory-Associated Protein of mTOR , Small Molecule Libraries/pharmacology
20.
Oncotarget ; 6(1): 185-95, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25474140

ABSTRACT

Recent clinical trials have demonstrated targeting PI3K pathway is a promising strategy for the treatment of blood cancers. To identify novel PI3K inhibitors, we performed a high throughput virtual screen and identified several novel small molecule compounds, including PIK-C98 (C98). The cell-free enzymatic studies showed that C98 inhibited all class I PI3Ks at nano- or low micromolar concentrations but had no effects on AKT or mTOR activity. Molecular docking analysis revealed that C98 interfered with the ATP-binding pockets of PI3Ks by forming H-bonds and arene-H interactions with specific amino acid residues. The cellular assays demonstrated that C98 specifically inhibited PI3K/AKT/mTOR signaling pathway, but had no effects on other kinases and proteins including IGF-1R, ERK, p38, c-Src, PTEN, and STAT3. Inhibition of PI3K by C98 led to myeloma cell apoptosis. Furthermore, oral administration of C98 delayed tumor growth in two independent human myeloma xenograft models in nude mice but did not show overt toxicity. Pharmacokinetic analyses showed that C98 was well penetrated into myeloma tumors. Therefore, through a high throughput virtual screen we identified a novel PI3K inhibitor that is orally active against multiple myeloma with great potential for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Furans/pharmacology , Gene Expression Regulation, Neoplastic , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Thiocarbamates/pharmacology , Administration, Oral , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Female , Humans , Hydrogen Bonding , Mice , Mice, Nude , Mice, SCID , Molecular Dynamics Simulation , Multiple Myeloma/drug therapy , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...