Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell Death Dis ; 15(2): 137, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38351178

ABSTRACT

Yin Yang 1 (YY1) is a key transcription factor that has been implicated in the development of several malignancies. The stability of YY1 is regulated by the ubiquitin-proteasome system. The role of deubiquitinases (DUBs) and their impact on YY1 remain to be fully elucidated. In this study, we screened for ubiquitin-specific proteases that interact with YY1, and identified OTUD3 as a DUB for YY1. Over-expressed OTUD3 inhibited YY1 degradation, thereby increasing YY1 protein levels, whereas OTUD3 knockdown or knockout promoted YY1 degradation, thereby decreasing the proliferation of colorectal cancer (CRC). Furthermore, PLK1 mediates OTUD3 S326 phosphorylation, which further enhances OTUD3 binding and deubiquitination of YY1. In CRC tissues, elevated the expression level of OTUD3 and YY1 were significantly associated with poor prognostic outcomes. These findings suggest that the OTUD3-YY1 pathway has therapeutic potential in CRC, and OTUD3 plays a critical role in regulating YY1.


Subject(s)
Colorectal Neoplasms , Ubiquitin-Specific Proteases , Humans , Phosphorylation , Ubiquitin-Specific Proteases/metabolism , YY1 Transcription Factor/metabolism , Ubiquitin/metabolism , Colorectal Neoplasms/genetics
2.
Biomed Eng Online ; 22(1): 125, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102586

ABSTRACT

BACKGROUND: Multi-omics research has the potential to holistically capture intra-tumor variability, thereby improving therapeutic decisions by incorporating the key principles of precision medicine. The purpose of this study is to identify a robust method of integrating features from different sources, such as imaging, transcriptomics, and clinical data, to predict the survival and therapy response of non-small cell lung cancer patients. METHODS: 2996 radiomics, 5268 transcriptomics, and 8 clinical features were extracted from the NSCLC Radiogenomics dataset. Radiomics and deep features were calculated based on the volume of interest in pre-treatment, routine CT examinations, and then combined with RNA-seq and clinical data. Several machine learning classifiers were used to perform survival analysis and assess the patient's response to adjuvant chemotherapy. The proposed analysis was evaluated on an unseen testing set in a k-fold cross-validation scheme. Score- and concatenation-based multi-omics were used as feature integration techniques. RESULTS: Six radiomics (elongation, cluster shade, entropy, variance, gray-level non-uniformity, and maximal correlation coefficient), six deep features (NasNet-based activations), and three transcriptomics (OTUD3, SUCGL2, and RQCD1) were found to be significant for therapy response. The examined score-based multi-omic improved the AUC up to 0.10 on the unseen testing set (0.74 ± 0.06) and the balance between sensitivity and specificity for predicting therapy response for 106 patients, resulting in less biased models and improving upon the either highly sensitive or highly specific single-source models. Six radiomics (kurtosis, GLRLM- and GLSZM-based non-uniformity from images with no filtering, biorthogonal, and daubechies wavelets), seven deep features (ResNet-based activations), and seven transcriptomics (ELP3, ZZZ3, PGRMC2, TRAK1, ATIC, USP7, and PNPLA2) were found to be significant for the survival analysis. Accordingly, the survival analysis for 115 patients was also enhanced up to 0.20 by the proposed score-based multi-omics in terms of the C-index (0.79 ± 0.03). CONCLUSIONS: Compared to single-source models, multi-omics integration has the potential to improve prediction performance, increase model stability, and reduce bias for both treatment response and survival analysis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Entropy , Gene Expression Profiling , Machine Learning , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases
3.
Yonsei Med J ; 64(12): 745-749, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37992747

ABSTRACT

Progressive familial intrahepatic cholestasis (PFIC) is a group of rare genetic disorders caused by defects in biliary epithelial transporters. It mostly presents as low γ-glutamyltransferase cholestasis. Recently, USP53 has been identified as one of the novel genes associated with PFIC. Herein, we report a 21-year-old Korean male patient with a late-onset PFIC. Initial work-up, including whole genome sequencing, did not find any associated gene. However, reviewing sequencing data identified novel compound heterozygous variants in splicing site of USP53 (NM_001371395.1:c.972+3_972+6del, and c.973-1G>A). The patient's bilirubin level fluctuated during the disease course. At 4.5 years after the initial presentation, the patient's symptom and high bilirubin level were normalized after administration of high-dose ursodeoxycholic acid. Recognition of this disease entity is important for prompt diagnosis and management. USP53 is recommended for the work-up of low γ-glutamyltransferase cholestasis.


Subject(s)
Cholestasis , High-Throughput Nucleotide Sequencing , Male , Adult , Humans , Young Adult , gamma-Glutamyltransferase/genetics , Bilirubin , Republic of Korea , Mutation , Ubiquitin-Specific Proteases
4.
Am J Chin Med ; 51(7): 1905-1925, 2023.
Article in English | MEDLINE | ID: mdl-37646142

ABSTRACT

Hexokinase 2 (HK2), the first glycolytic rate-limiting enzyme, is closely correlated with the occurrence and progression of tumors. Effective therapeutic agents targeting HK2 are urgently needed. Bergenin has exhibited various pharmacological activities, such as antitumor properties. However, the effects of bergenin on the abnormal glucose metabolism of cancer cells are yet unclear. In this study, HK2 was overexpressed in OSCC tissues, and the depletion of HK2 inhibited the growth of OSCC cells in vitro and in vivo. Moreover, these results showed that the natural compound, bergenin, exerted a robust antitumor effect on OSCC cells. Bergenin inhibited cancer cell proliferation, suppressed glycolysis, and induced intrinsic apoptosis in OSCC cells by downregulating HK2. Notably, bergenin restored the antitumor efficacy of irradiation in the radioresistant OSCC cells. A mechanistic study revealed that bergenin upregulated the protein level of phosphatase and the tensin homolog deleted on chromosome 10 (PTEN) by enhancing the interaction between PTEN and ubiquitin-specific protease 13 (USP13) and stabilizing PTEN; this eventually inhibited AKT phosphorylation and HK2 expression. Bergenin was identified as a novel therapeutic agent against glycolysis to inhibit OSCC and overcome radioresistance. Targeting PTEN/AKT/HK2 signaling could be a promising option for clinical OSCC treatment.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Glycolysis/genetics , Cell Line, Tumor , Cell Proliferation , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/pharmacology
5.
ACS Chem Neurosci ; 14(10): 1859-1869, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37116219

ABSTRACT

Cav3.2 channels play an important role in the afferent nociceptive pathway, which is responsible for both physiological and pathological pain transmission. Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the deubiquitinase USP5. In this study, we investigated nine naturally occurring flavonoid derivatives which we tested for their abilities to inhibit transiently expressed Cav3.2 channels and their interactions with USP5. Icariside II (ICA-II), one of the flavonols studied, inhibited the biochemical interactions between USP5 and Cav3.2 and concomitantly and effectively blocked Cav3.2 channels. Molecular docking analysis predicts that ICA-II binds to the cUBP domain and the Cav3.2 interaction region. In addition, ICA-II was predicted to interact with residues in close proximity to the Cav3.2 channel's fenestrations, thus accounting for the observed blocking activity. In mice with inflammatory and neuropathic pain, ICA-II inhibited both phases of the formalin-induced nocifensive responses and abolished thermal hyperalgesia induced by injection of complete Freund's adjuvant (CFA) into the hind paw. Furthermore, ICA-II produced significant and long-lasting thermal anti-hyperalgesia in female mice, whereas Cav3.2 null mice were resistant to the action of ICA-II. Altogether, our data show that ICA-II has analgesic activity via an action on Cav3.2 channels.


Subject(s)
Calcium Channels, T-Type , Neuralgia , Female , Mice , Animals , Calcium Channels, T-Type/metabolism , Molecular Docking Simulation , Neuralgia/drug therapy , Neuralgia/metabolism , Hyperalgesia/metabolism , Flavonoids , Flavonols , Mice, Knockout , Ubiquitin-Specific Proteases/metabolism
6.
Pathol Int ; 73(3): 109-119, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36285444

ABSTRACT

Laryngeal cancer (LC) is a rare and challenging clinical problem. Our aim was to investigate the mechanism of salt-like transcription factor 4 (SALL4) in LC. LC tissue and paracancerous tissue were collected. Relative mRNA or protein levels were measured by quantitative real-time polymerase chain reaction or Western blot. MTT, wound healing, and transwell assay were performed to evaluate cell proliferation, migration and invasion. The binding relationship between SALL4 and USP21 promoter was verified by dual-luciferase assay and ChIP. Co-IP and glutathione-S-transferase (GST)-pull down were performed to measure the protein interaction between USP21 and YY1. Additionally, YY1 ubiquitination level was analyzed. It was found that SALL4 mRNA and SALL4 protein levels were elevated in LC clinical tissues and various LC cells. Knockdown of SALL4 inhibited epithelial-mesenchymal transition (EMT) of LC cells. USP21 was transcriptionally activated by SALL4. Co-IP and GST-pull down confirmed USP21 interacted with YY1. USP21 protected YY1 from degradation through deubiquitination. Furthermore, overexpression of USP21 reversed the effect of knockdown of SALL4 on YY1 and EMT in LC cells. In general, SALL4 facilitated EMT of LC cells through modulating USP21/YY1 axis.


Subject(s)
Laryngeal Neoplasms , Transcription Factors , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Laryngeal Neoplasms/genetics , RNA, Messenger , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Yin-Yang
7.
Proc Natl Acad Sci U S A ; 119(44): e2207975119, 2022 11.
Article in English | MEDLINE | ID: mdl-36279435

ABSTRACT

Stress granules (SGs) are cytoplasmic biomolecular condensates containing proteins and RNAs in response to stress. Ras-GTPase-activating protein binding protein 1 (G3BP1) is a core SG protein. Caprin-1 and ubiquitin specific peptidase 10 (USP10) interact with G3BP1, facilitating and suppressing SG formation, respectively. The crystal structures of the nuclear transport factor 2-like (NTF2L) domain of G3BP1 in complex with the G3BP1-interacting motif (GIM) of Caprin-1 and USP10 show that both GIMs bind to the same hydrophobic pocket of G3BP1. Moreover, both GIMs suppressed the liquid-liquid phase separation (LLPS) of G3BP1, suggesting that Caprin-1 likely facilitates SG formation via other mechanisms. Thus, we dissected various domains of Caprin-1 and investigated their role in LLPS in vitro and SG formation in cells. The C-terminal domain of Caprin-1 underwent spontaneous LLPS, whereas the N-terminal domain and GIM of Caprin-1 suppressed LLPS of G3BP1. The opposing effect of the N- and C-terminal domains of Caprin-1 on SG formation were demonstrated in cells with or without the endogenous Caprin-1. We propose that the N- and C-terminal domains of Caprin-1 regulate SG formation in a "yin and yang" fashion, mediating the dynamic and reversible assembly of SGs.


Subject(s)
DNA Helicases , RNA Helicases , RNA Recognition Motif Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , DNA Helicases/metabolism , Cytoplasmic Granules/metabolism , Stress Granules , GTPase-Activating Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism
8.
Cell Mol Gastroenterol Hepatol ; 14(5): 1103-1122, 2022.
Article in English | MEDLINE | ID: mdl-35934222

ABSTRACT

BACKGROUND & AIMS: Severe acute pancreatitis can easily lead to systemic inflammatory response syndrome and death. Macrophages are known to be involved in the pathophysiology of acute pancreatitis (AP), and macrophage activation correlates with disease severity. In this study, we examined the role of ubiquitin-specific protease 25, a deubiquitinating enzyme and known regulator of macrophages, in the pathogenesis of AP. METHODS: We used L-arginine, cerulein, and choline-deficient ethionine-supplemented diet-induced models of AP in Usp25-/- mice and wild-type mice. We also generated bone marrow Usp25-/- chimeric mice and initiated L-arginine-mediated AP. Primary acinar cells and bone marrow-derived macrophages were isolated from wild-type and Usp25-/- mice to dissect molecular mechanisms. RESULTS: Our results show that Usp25 deficiency exacerbates pancreatic and lung injury, neutrophil and macrophage infiltration, and systemic inflammatory responses in L-arginine, cerulein, and choline-deficient ethionine-supplemented diet-induced models of AP. Bone marrow Usp25-/- chimeric mice challenged with L-arginine show that Usp25 deficiency in macrophages exaggerates AP by up-regulating the TANK-binding kinase 1 (TBK1)-nuclear factor-κB (NF-κB) signaling pathway. Similarly, in vitro data confirm that Usp25 deficiency enhances the TBK1-NF-κB pathway, leading to increased expression of inflammatory cytokines in bone marrow-derived macrophages. CONCLUSIONS: Usp25 deficiency in macrophages enhances TBK1-NF-κB signaling, and the induction of inflammatory chemokines and type I interferon-related genes exacerbates pancreatic and lung injury in AP.


Subject(s)
Pancreatitis , Ubiquitin Thiolesterase , Animals , Mice , Acute Disease , Arginine , Ceruletide , Choline , Cytokines/metabolism , Deubiquitinating Enzymes/metabolism , Disease Models, Animal , Ethionine , Interferon Type I , Lung Injury , Macrophages/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Signal Transduction , Ubiquitin-Specific Proteases/metabolism , Ubiquitin Thiolesterase/metabolism
9.
Proc Natl Acad Sci U S A ; 119(36): e2119854119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037364

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by the loss of tumor suppressor Von Hippel Lindau (VHL) function. VHL is the component of an E3 ligase complex that promotes the ubiquitination and degradation of hypoxia inducible factor α (HIF-α) (including HIF1α and HIF2α) and Zinc Fingers And Homeoboxes 2 (ZHX2). Our recent research showed that ZHX2 contributed to ccRCC tumorigenesis in a HIF-independent manner. However, it is still unknown whether ZHX2 could be modified through deubiquitination even in the absence of pVHL. Here, we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and identified USP13 as a DUB that bound ZHX2 and promoted ZHX2 deubiquitination. As a result, USP13 promoted ZHX2 protein stability in an enzymatically dependent manner, and depletion of USP13 led to ZHX2 down-regulation in ccRCC. Functionally, USP13 depletion led to decreased cell proliferation measured by two-dimensional (2D) colony formation and three-dimensional (3D) anchorage-independent growth. Furthermore, USP13 was essential for ccRCC tumor growth in vivo, and the effect was partially mediated by its regulation on ZHX2. Our findings support that USP13 may be a key effector in ccRCC tumorigenesis.


Subject(s)
Carcinoma, Renal Cell , Homeodomain Proteins , Kidney Neoplasms , Transcription Factors , Ubiquitin-Specific Proteases , Carcinogenesis/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
10.
Ren Fail ; 44(1): 752-766, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35509185

ABSTRACT

Chronic kidney disease (CKD) has recently become a serious health and social concern. Vascular calcification, a common complication of CKD, is a risk factor that increases the incidence and mortality of cardiovascular events in patients with CKD. However, there are currently no effective therapeutic targets that can facilitate treatment with fewer side effects for vascular calcification in CKD. To identify potential therapeutic targets, we performed label-free quantification (LFQ) analyses of protein samples from rat aortic vascular smooth muscle cells (RASMCs) after high-phosphorus treatment by nano-UPLC-MS/MS. We determined that ubiquitin-specific protease 47 (USP47) may be associated with CKD vascular calcification by regulating the osteogenic transdifferentiation of the vascular smooth muscle cell (VSMC) phenotype, thus suggesting a novel and potentially effective therapeutic target for CKD vascular calcification. USP47 knockdown significantly reduced the expression of ß-transducin repeat-containing protein (BTRC), serine/threonine-protein kinase akt-1 (AKT1), Klotho, fibroblast growth factor (FGF23), and matrix Gla protein (MGP) in RASMCs after high-phosphorus treatment. Consistent with the results of protein-protein interaction (PPI) analyses, USP47 may be involved in regulating osteogenic transdifferentiation markers, such as runt-related transcription factor 2 (RUNX2), Klotho, FGF23, and MGP through the BTRC/AKT1 pathway upon CKD vascular calcification. These data indicate that USP47 may be associated with vascular calcification in CKD by regulating osteogenic differentiation of VSMCs. USP47 may regulate osteogenic transdifferentiation in VSMCs upon CKD vascular calcification through a process involving the BTRC/AKT1 pathway. This study identified a novel potential therapeutic target for the treatment of vascular calcification in CKD.


Subject(s)
Renal Insufficiency, Chronic , Ubiquitin-Specific Proteases , Vascular Calcification , Animals , Cell Transdifferentiation/genetics , Cells, Cultured , Female , Humans , Male , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/metabolism , Osteogenesis/genetics , Phosphorus/metabolism , Rats , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Tandem Mass Spectrometry , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/pharmacology , Vascular Calcification/metabolism
11.
ACS Chem Neurosci ; 13(4): 524-536, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35113527

ABSTRACT

Cav3.2 calcium channels are important mediators of nociceptive signaling in the primary afferent pain pathway, and their expression is increased in various rodent models of chronic pain. Previous work from our laboratory has shown that this is in part mediated by an aberrant expression of deubiquitinase USP5, which associates with these channels and increases their stability. Here, we report on a novel bioactive rhodanine compound (II-1), which was identified in compound library screens. II-1 inhibits biochemical interactions between USP5 and the Cav3.2 domain III-IV linker in a dose-dependent manner, without affecting the enzymatic activity of USP5. Molecular docking analysis reveals two potential binding pockets at the USP5-Cav3.2 interface that are distinct from the binding site of the deubiquitinase inhibitor WP1130 (a.k.a. degrasyn). With an understanding of the ability of some rhodanines to produce false positives in high-throughput screening, we have conducted several orthogonal assays to confirm the validity of this hit, including in vivo experiments. Intrathecal delivery of II-1 inhibited both phases of formalin-induced nocifensive behaviors in mice, as well as abolished thermal hyperalgesia induced by the delivery of complete Freund's adjuvant (CFA) to the hind paw. The latter effects were abolished in Cav3.2 null mice, thus confirming that Cav3.2 is required for the action of II-1. II-1 also mediated a robust inhibition of mechanical allodynia induced by injury to the sciatic nerve. Altogether, our data uncover a novel class of analgesics─well suited to rapid structure-activity relationship studies─that target the Cav3.2/USP5 interface.


Subject(s)
Analgesics , Calcium Channels, T-Type , Neuralgia , Ubiquitin-Specific Proteases , Analgesics/pharmacology , Animals , Calcium Channel Blockers , Calcium Channels, T-Type/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Mice , Molecular Docking Simulation , Neuralgia/metabolism , Structure-Activity Relationship , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism
12.
Pediatr Dev Pathol ; 25(2): 207-212, 2022.
Article in English | MEDLINE | ID: mdl-34809518

ABSTRACT

Whole-exome sequencing studies have recently identified novel genes implicated in normal- or low-GGT pediatric cholestasis including ubiquitin-specific peptidase 53 (USP53). We identified novel biallelic mutations in the USP53 gene in a 7-month-old infant with pruritus and progressive intrahepatic cholestasis. His liver biopsy showed portal and perivenular fibrosis with bland bilirubinostasis. His parents were asymptomatic heterozygous for the same mutation. He is currently on vitamin supplements and cholestyramine and his family has also been counseled for liver transplantation. Our report confirms that patients with biallelic mutation in USP53 develop cholestatic liver disease.


Subject(s)
Cholestasis, Intrahepatic , Cholestasis , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/genetics , Humans , Infant , Male , Mutation , Ubiquitin-Specific Proteases/genetics , Exome Sequencing
13.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925279

ABSTRACT

Ubiquitylation and deubiquitylation are reversible protein post-translational modification (PTM) processes involving the regulation of protein degradation under physiological conditions. Loss of balance in this regulatory system can lead to a wide range of diseases, such as cancer and inflammation. As the main members of the deubiquitinases (DUBs) family, ubiquitin-specific peptidases (USPs) are closely related to biological processes through a variety of molecular signaling pathways, including DNA damage repair, p53 and transforming growth factor-ß (TGF-ß) pathways. Over the past decade, increasing attention has been drawn to USPs as potential targets for the development of therapeutics across diverse therapeutic areas. In this review, we summarize the crucial roles of USPs in different signaling pathways and focus on advances in the development of USP inhibitors, as well as the methods of screening and identifying USP inhibitors.


Subject(s)
Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism , Deubiquitinating Enzymes/metabolism , Drug Evaluation, Preclinical , Humans , Neoplasms/metabolism , Protein Processing, Post-Translational/physiology , Proteolysis , Signal Transduction , Ubiquitin/metabolism , Ubiquitination/physiology
14.
Oncogene ; 40(13): 2367-2381, 2021 04.
Article in English | MEDLINE | ID: mdl-33658627

ABSTRACT

Histone deacetylase (HDAC) inhibitors are effective in MYCN-driven cancers, because of a unique need for HDAC recruitment by the MYCN oncogenic signal. However, HDAC inhibitors are much more effective in combination with other anti-cancer agents. To identify novel compounds which act synergistically with HDAC inhibitor, such as suberanoyl hydroxamic acid (SAHA), we performed a cell-based, high-throughput drug screen of 10,560 small molecule compounds from a drug-like diversity library and identified a small molecule compound (SE486-11) which synergistically enhanced the cytotoxic effects of SAHA. Effects of drug combinations on cell viability, proliferation, apoptosis and colony forming were assessed in a panel of neuroblastoma cell lines. Treatment with SAHA and SE486-11 increased MYCN ubiquitination and degradation, and markedly inhibited tumorigenesis in neuroblastoma xenografts, and, MYCN transgenic zebrafish and mice. The combination reduced ubiquitin-specific protease 5 (USP5) levels and increased unanchored polyubiquitin chains. Overexpression of USP5 rescued neuroblastoma cells from the cytopathic effects of the combination and reduced unanchored polyubiquitin, suggesting USP5 is a therapeutic target of the combination. SAHA and SE486-11 directly bound to USP5 and the drug combination exhibited a 100-fold higher binding to USP5 than individual drugs alone in microscale thermophoresis assays. MYCN bound to the USP5 promoter and induced USP5 gene expression suggesting that USP5 and MYCN expression created a forward positive feedback loop in neuroblastoma cells. Thus, USP5 acts as an oncogenic cofactor with MYCN in neuroblastoma and the novel combination of HDAC inhibitor with SE486-11 represents a novel therapeutic approach for the treatment of MYCN-driven neuroblastoma.


Subject(s)
Carcinogenesis/drug effects , Histone Deacetylase Inhibitors/pharmacology , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Ubiquitin-Specific Proteases/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Small Molecule Libraries/pharmacology , Vorinostat/pharmacology , Zebrafish/genetics
15.
Biochem Cell Biol ; 99(2): 223-230, 2021 04.
Article in English | MEDLINE | ID: mdl-32845162

ABSTRACT

Abraxas brother protein 1 (ABRO1) is a subunit of the deubiquitinating enzyme BRCC36-containing isopeptidase complex and plays important roles in cellular responses to stress by interacting with its binding partners, such as ubiquitin-specific peptidase 7, p53, activating transcription factor 4, THAP-domain containing 5, and serine hydroxymethyltransferase. However, the transcriptional regulation of ABRO1 remains unexplored. In this study, we identified and characterized the core regulatory elements of the human ABRO1 gene and mapped them to the ABRO1 promoter region. Additionally, 5' rapid amplification of cDNA ends revealed that the transcriptional start site (TSS) was located -13 bp upstream from the start codon. Reporter gene, chromatin immunoprecipitation, and electrophoretic mobility shift assays demonstrated that ABRO1 transcription was regulated through cis-acting elements located in the region -89 to -59 bp upstream of the ABRO1 TSS and that these elements were targeted by yin yang 1 transcription factor (YY1). Moreover, YY1 overexpression increased human ABRO1 mRNA and protein expression, and small-interfering RNA-mediated downregulation of YY1 attenuated ABRO1 expression. These results suggested that YY1 positively regulated human ABRO1 expression by binding to cis-acting elements located in the ABRO1 TSS.


Subject(s)
Nuclear Matrix-Associated Proteins/genetics , Ubiquitin-Specific Proteases/genetics , YY1 Transcription Factor/metabolism , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Nuclear Matrix-Associated Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Specific Proteases/metabolism
16.
Mol Brain ; 11(1): 60, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30340616

ABSTRACT

Cav3.2 T-type calcium channels are important for the signaling of nociceptive information in the primary afferent pain pathway. During neuropathy and peripheral inflammation, Cav3.2 channels are upregulated due to an increased association with the deubiquitinase USP5. Disrupting these interactions in male mice by the use of cell permeant peptides reverses mechanical and thermal hypersensitivity. Here we explore the effects of interfering with USP5 binding to the channel in female mice with synchronized estrous cycle. We show that intrathecal delivery of a cell-penetrating TAT peptide corresponding to the UBPc domain of USP5 fully reverses mechanical hypersensitivity in mice intraplantarly injected with Complete Freund's Adjuvant. Hence, the USP5 mediated dysregulation of Cav3.2 channel activity does not exhibit sex differences, and potential therapeutics targeting this interaction should be effective in both male and female subjects.


Subject(s)
Calcium Channels, T-Type/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Chronic Pain/metabolism , Chronic Pain/pathology , Female , Hyperalgesia/pathology , Inflammation/pathology , Male , Mice, Inbred C57BL , Peptides/metabolism
17.
J Nutr Biochem ; 49: 63-70, 2017 11.
Article in English | MEDLINE | ID: mdl-28886438

ABSTRACT

Ubiquitin-specific protease 19 (USP19) is a key player in the negative regulation of muscle mass during muscle atrophy. Loss-of-function approaches demonstrate that 17ß-estradiol (E2) increases USP19 expression through estrogen receptor (ER) α and consequently decreases soleus muscle mass in young female mice under physiological conditions. Daidzein is one of the main isoflavones in soy, and activates ERß-dependent transcription. Here, we investigated the effects of daidzein on E2-increased USP19 expression and E2-decreased soleus muscle mass in young female mice. Daidzein stimulated the transcriptional activity of ERß in murine C2C12 cells and down-regulated USP19 expression. Consistently, daidzein inhibited E2-induced USP19 expression in a reporter activity using a functional half-estrogen response element (hERE) from Usp19. Daidzein inhibited E2-induced recruitment of ERα and promoted recruitment of ERß to the Usp19 hERE. Dietary daidzein down-regulated the expression of USP19 at the mRNA and protein levels and increased soleus muscle mass in female mice, but not in males. In soleus muscle from ovariectomized (OVX) female mice, dietary daidzein inhibited E2-increased USP19 mRNA expression and E2-decreased muscle mass. Furthermore, E2 induced the recruitment of ERα and ERß to the hERE, whereas daidzein inhibited E2-induced recruitment of ERα, and enhanced E2-increased recruitment of ERß, to the Usp19 hERE. These results demonstrate that dietary daidzein decreases USP19 mRNA expression through ERß and increases soleus muscle mass in young female mice, but not in male mice, under physiological conditions.


Subject(s)
Dietary Supplements , Estrogen Receptor beta/agonists , Isoflavones/therapeutic use , Muscle, Skeletal/metabolism , Phytoestrogens/therapeutic use , Sarcopenia/prevention & control , Ubiquitin-Specific Proteases/antagonists & inhibitors , Active Transport, Cell Nucleus , Animals , Animals, Outbred Strains , Cell Line , Endopeptidases , Enzyme Repression , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Genes, Reporter , Male , Mice , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Ovariectomy/adverse effects , Random Allocation , Response Elements , Sarcopenia/etiology , Sarcopenia/metabolism , Sarcopenia/pathology , Sex Characteristics , Signal Transduction , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
18.
Sci Rep ; 7(1): 6427, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743993

ABSTRACT

The ICR (Imprinting Control Region) of the Peg3 (Paternally Expressed Gene 3) domain contains an unusual cluster of YY1 binding sites. In the current study, these YY1 binding sites were mutated to characterize the unknown roles in the mouse Peg3 domain. According to the results, paternal and maternal transmission of the mutant allele did not cause any major effect on the survival of the pups. In the mutants, the maternal-specific DNA methylation on the ICR was properly established and maintained, causing no major effect on the imprinting of the domain. In contrast, the paternal transmission resulted in changes in the expression levels of several genes: down-regulation of Peg3 and Usp29 and up-regulation of Zim1. These changes were more pronounced during the neonatal stage than during the adult stage. In the case of Peg3 and Zim1, the levels of the observed changes were also different between males and females, suggesting the different degrees of YY1 involvement between two sexes. Overall, the results indicated that YY1 is mainly involved in controlling the transcriptional levels, but not the DNA methylation, of the Peg3 domain.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , YY1 Transcription Factor/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Chromatin/chemistry , Chromatin/genetics , DNA Methylation , DNA-Binding Proteins , Female , Gene Expression Regulation , Genomic Imprinting , Male , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Phenotype , Protein Domains , Ubiquitin-Specific Proteases/genetics , YY1 Transcription Factor/genetics
19.
Phytomedicine ; 23(12): 1344-1355, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27765354

ABSTRACT

BACKGROUND AND PURPOSE: Aberrant expression of ß-catenin is highly associated with progression of various cancers including head and neck cancer (HNC). Green tea is most commonly used beverage in the world and one of the more bioactive compounds is the antioxidant epigallocatechin gallate (EGCG). This study was performed to investigate the mechanism by which EGCG inhibits the growth of HNC, focusing on the modulation of the expression and activity of ß-catenin. METHODS: In vitro effects of EGCG on the transcription, translation, or degradation of ß-catenin were investigated. Antitumor effects of EGCG in vivo were evaluated in a syngeneic mouse model and ß-catenin expression was checked in HNC patients' samples. RESULTS: ß-catenin expression was elevated in tumor samples of HNC patients. EGCG induced apoptosis in KB and FaDu cells through the suppression of ß-catenin signaling. Knockdown of ß-catenin using siRNA enhanced the proapoptotic activities of EGCG. EGCG decreased mRNA and transcriptional activity of ß-catenin in p53 wild-type KB cells. EGCG also enhanced the ubiquitination and proteasomal degradation of ß-catenin. The suppression of ß-catenin and consequent apoptosis were observed in response to EGCG treatment in a syngeneic mouse model. In conclusion, we report that EGCG inhibits ß-catenin expression through multiple mechanisms including decreased transcription and increased ubiquitin-mediated 26S proteasomal degradation. CONCLUSION: This study proposes a novel molecular rationale for antitumor activities of green tea in HNCs.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Catechin/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Transcriptional Activation/drug effects , beta Catenin/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Knockdown Techniques , Genes, p53/drug effects , Humans , Male , Mice , Mice, Inbred C3H , Middle Aged , Neoplasm Transplantation , Signal Transduction/drug effects , Ubiquitin-Specific Proteases/genetics , beta Catenin/drug effects , beta Catenin/genetics
20.
EBioMedicine ; 12: 219-226, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27658736

ABSTRACT

Cutaneous abscess infections are difficult to treat with current therapies and alternatives to conventional antibiotics are needed. Understanding the regulatory mechanisms that govern abscess pathology should reveal therapeutic interventions for these recalcitrant infections. Here we demonstrated that the stringent stress response employed by bacteria to cope and adapt to environmental stressors was essential for the formation of lesions, but not bacterial growth, in a methicillin resistant Staphylococcus aureus (MRSA) cutaneous abscess mouse model. To pharmacologically confirm the role of the stringent response in abscess formation, a cationic peptide that causes rapid degradation of the stringent response mediator, guanosine tetraphosphate (ppGpp), was employed. The therapeutic application of this peptide strongly inhibited lesion formation in mice infected with Gram-positive MRSA and Gram-negative Pseudomonas aeruginosa. Overall, we provide insights into the mechanisms governing abscess formation and a paradigm for treating multidrug resistant cutaneous abscesses.


Subject(s)
Abscess/metabolism , Abscess/microbiology , Stress, Physiological , Abscess/drug therapy , Abscess/pathology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Bacterial Toxins/metabolism , Biofilms/drug effects , Disease Models, Animal , Female , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL