Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 25(14): 7089-7094, 2021 07.
Article in English | MEDLINE | ID: mdl-34114734

ABSTRACT

MYSM1 is a chromatin-binding protein, widely investigated for its functions in haematopoiesis in human and mouse; however, its role in haematologic malignancies remains unexplored. Here, we investigate the cross-talk between MYSM1 and oncogenic cMYC in the transcriptional regulation of genes encoding ribosomal proteins, and the implications of these mechanisms for cMYC-driven carcinogenesis. We demonstrate that in cMYC-driven B cell lymphoma in mouse models, MYSM1-loss represses ribosomal protein gene expression and protein synthesis. Importantly, the loss of MYSM1 also strongly inhibits cMYC oncogenic activity and protects against B cell lymphoma onset and progression in the mouse models. This advances the understanding of the molecular and transcriptional mechanisms of lymphomagenesis, and suggests MYSM1 as a possible drug target for cMYC-driven malignancies.


Subject(s)
Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Trans-Activators/deficiency , Ubiquitin-Specific Proteases/deficiency , Animals , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Mice , Proto-Oncogene Proteins c-myc/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
2.
J Pediatr Gastroenterol Nutr ; 72(5): 667-673, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33075013

ABSTRACT

OBJECTIVES: Although a number of genetic forms of cholestasis have been identified, the genetic etiology of disease remains unidentified in a subset of cholestasis patients. METHODS: Whole exome sequencing (WES) was performed in DNA from patients diagnosed with cholestasis, at different points on the continuum from progressive familial intrahepatic cholestasis to benign recurrent intrahepatic cholestasis, in whom no disease mutations in known cholestasis genes had been identified. Candidate genes were then assessed in a larger patient sample, by targeted next-generation sequencing (NGS). Disease features at presentation and follow-up were collected from available medical records. RESULTS: By WES, we identified 3 patients with homozygous mutations in USP53. Screening of USP53 in a larger set of patients identified 4 additional patients with homozygous mutations in USP53. Six of the 7 patients had deletion mutations, and 1 had a missense mutation; 3 of the patients were siblings, all bearing a deletion that also disrupted neighboring MYOZ2. Age of onset ranged from early infancy to adolescence. Cholestasis tended to be biochemically mild and intermittent, and responsive to medication. Liver fibrosis was, however, present in all 4 patients who were biopsied, and splenomegaly was apparent in 5 of 7 at last ultrasound. CONCLUSIONS: Two groups recently identified patients with liver disease and mutation in USP53. We have now identified biallelic mutation in USP53 in 7 further patients with cholestasis, from 5 families. Most individuals had evidence of chronic liver disease, and long-term follow-up is recommended.


Subject(s)
Cholestasis, Intrahepatic , Cholestasis , Ubiquitin-Specific Proteases/deficiency , Adolescent , Carrier Proteins , Child , Child, Preschool , Cholestasis/genetics , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/genetics , Homozygote , Humans , Infant , Muscle Proteins , Mutation , Ubiquitin-Specific Proteases/genetics , Exome Sequencing
3.
Curr Cancer Drug Targets ; 20(9): 689-699, 2020.
Article in English | MEDLINE | ID: mdl-32359336

ABSTRACT

BACKGROUND: Ubiquitin specific protease 48 (USP48) is a member of the deubiquitinating enzymes (DUBs) family. However, the function of USP48 in ovarian cancer remains unclear. OBJECTIVE: The present study reveals that USP48 knockdown could significantly inhibit cell migration and invasion in ES2, 3AO and A2780 cells, without affecting cell proliferation. METHODS: After carboplatin (CBP) treatment, the USP48 ablation increases the apoptosis rate, and the cleaved PARP and cleaved caspase 3 expression levels in ES2, 3AO and A2780 cells. The subcutaneous tumor and intraperitoneally injected experiments demonstrated that the USP48 knockdown significantly increases responsiveness to CBP, and alleviates the metastasis in vivo. Meanwhile, USP48 deficiency results in the improved survival of mice. RESULTS: Finally, the analysis of clinical samples and the TCGA and Kaplan-Meier Plot database revealed that the high expression of USP48 in ovarian cancer patients is associated with poor survival and resistance to CBP therapy. CONCLUSION: In summary, USP48 may be a potential therapeutic target for ovarian cancer patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ubiquitin-Specific Proteases/deficiency , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Metastasis/genetics , Ovarian Neoplasms/pathology , Progression-Free Survival , Transfection , Tumor Burden/drug effects , Tumor Burden/genetics , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
4.
J Immunol ; 203(8): 2049-2054, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31534008

ABSTRACT

Cyclic GMP-AMP synthase (cGAS), a cytosolic DNA sensor, catalyzes the formation of the second messenger 2'3'-cGAMP that binds to STING and triggers the type I IFN signaling. Activation of cGAS can be modulated by several protein posttranslational modifications, including ubiquitination. However, the cGAS activation regulated by protein deubiquitination remains poorly understood. In this study, we identified that deubiquitinase USP27X could interact with cGAS and cleave K48-linked polyubiquitination chains from cGAS, leading to cGAS stabilization. Consistently, knockout of Usp27x in mice macrophages resulted in an accelerated turnover of cGAS, decreased cGAMP production, phosphorylation of TBK1 and IRF3, and IFN-ß production. Furthermore, Usp27x knockout mice macrophages showed impaired innate antiviral responses against HSV type 1 infection. Our data suggest that USP27X is a novel regulator of the cGAS-STING cytosolic DNA sensing pathway.


Subject(s)
Cytosol/metabolism , DNA/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Ubiquitin-Specific Proteases/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Knockout , RAW 264.7 Cells , Ubiquitin-Specific Proteases/deficiency , Ubiquitination
5.
Sci China Life Sci ; 62(4): 544-552, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30919279

ABSTRACT

The balanced actions between ubiquitination and deubiquitination precisely control the levels of various proteins vital for spermatogenesis. Ubiquitin-specific processing proteases (USPs) are the largest family of deubiquitinatingenzymes(DUBs), containing more than 50 members. So far, the functions of only a few USPs in male fertility have been studied, the roles of the majority are yet unknown. The present study aimed to explore the function of Usp29 (ubiquitin-specific protease 29) in male fertility. We found that Usp29 showed predominant expression in mouse testis, and its mRNA expression started to increase at 14 days postpartum (dpp), with a peak at 28 and 35 dpp. Using CRISPR/Cas9 technology, we generated Usp29 knockout mice (Usp29-/-). Usp29-/- mice exhibited no overt developmental anomalies. Further examination revealed that Usp29-/- mice had normal fertility and showed no detectable difference in the testis/body weight ratio, testicular and epididymal histology as well as epididymal sperm count from the wild-type littermates. Moreover, Usp29 is not a pseudogene in mice. Taken together, our study first reported that though Usp29 is predominantly expressed in the testis, it is not essential for male fertility in mice.


Subject(s)
Fertility/genetics , Ubiquitin-Specific Proteases/metabolism , Animals , Epididymis/anatomy & histology , Female , Genome/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phylogeny , RNA, Messenger/metabolism , Sperm Count , Spermatogenesis , Testis/anatomy & histology , Testis/metabolism , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics
6.
Clin Sci (Lond) ; 133(2): 335-349, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30622220

ABSTRACT

Ubiquitin-specific peptidase 4 (USP4) protein is a type of deubiquitination enzyme that is correlated with many important biological processes. However, the function of USP4 in hepatic ischaemia/reperfusion (I/R) injury remains unknown. The aim of the present study was to explore the role of USP4 in hepatic I/R injury. USP4 gene knockout mice and primary hepatocytes were used to construct hepatic I/R models. The effect of USP4 on hepatic I/R injury was examined via pathological and molecular analyses. Our results indicated that USP4 was significantly up-regulated in liver of mice subjected to hepatic I/R injury. USP4 knockout mice exhibited exacerbated hepatic I/R injury, as evidenced by enhanced liver inflammation via the nuclear factor κB (NF-κB) signalling pathway and increased hepatocyte apoptosis. Additionally, USP4 overexpression inhibited hepatocyte inflammation and apoptosis on hepatic I/R stimulation. Mechanistically, our study demonstrates that USP4 deficiency exerts its detrimental effects on hepatic I/R injury by inducing activation of the transforming growth factor ß-activated kinase 1 (TAK1)/JNK signalling pathways. TAK1 was required for USP4 function in hepatic I/R injury as TAK1 inhibition abolished USP4 function in vitro In conclusion, our study demonstrates that USP4 deficiency plays a detrimental role in hepatic I/R injury by promoting activation of the TAK1/JNK signalling pathways. Modulation of this axis may be a novel strategy to alleviate the pathological process of hepatic I/R injury.


Subject(s)
Liver/blood supply , Liver/enzymology , MAP Kinase Kinase Kinases/metabolism , Reperfusion Injury/enzymology , Ubiquitin-Specific Proteases/deficiency , Animals , Apoptosis , Disease Models, Animal , HEK293 Cells , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , MAP Kinase Kinase Kinases/genetics , Male , Mice, Knockout , NF-kappa B/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Ubiquitin-Specific Proteases/genetics
7.
Nat Commun ; 9(1): 2280, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891926

ABSTRACT

Defects in DNA repair can cause various genetic diseases with severe pathological phenotypes. Fanconi anemia (FA) is a rare disease characterized by bone marrow failure, developmental abnormalities, and increased cancer risk that is caused by defective repair of DNA interstrand crosslinks (ICLs). Here, we identify the deubiquitylating enzyme USP48 as synthetic viable for FA-gene deficiencies by performing genome-wide loss-of-function screens across a panel of human haploid isogenic FA-defective cells (FANCA, FANCC, FANCG, FANCI, FANCD2). Thus, as compared to FA-defective cells alone, FA-deficient cells additionally lacking USP48 are less sensitive to genotoxic stress induced by ICL agents and display enhanced, BRCA1-dependent, clearance of DNA damage. Consequently, USP48 inactivation reduces chromosomal instability of FA-defective cells. Our results highlight a role for USP48 in controlling DNA repair and suggest it as a potential target that could be therapeutically exploited for FA.


Subject(s)
DNA Repair/genetics , DNA Repair/physiology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , BRCA1 Protein/metabolism , CRISPR-Cas Systems , Cell Line , Chromosomal Instability , DNA Damage , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Knockout Techniques , Genetic Therapy , Histones/metabolism , Humans , Mutation , Rad51 Recombinase/metabolism , Ubiquitin-Specific Proteases/deficiency , Ubiquitination
8.
Cell Rep ; 13(11): 2470-2479, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686633

ABSTRACT

USP15 is a deubiquitinase that negatively regulates activation of naive CD4(+) T cells and generation of IFN-γ-producing T helper 1 (Th1) cells. USP15 deficiency in mice promotes antitumor T cell responses in a transplantable cancer model; however, it has remained unclear how deregulated T cell activation impacts primary tumor development during the prolonged interplay between tumors and the immune system. Here, we find that the USP15-deficient mice are hypersensitive to methylcholantrene (MCA)-induced fibrosarcomas. Excessive IFN-γ production in USP15-deficient mice promotes expression of the immunosuppressive molecule PD-L1 and the chemokine CXCL12, causing accumulation of T-bet(+) regulatory T cells and CD11b(+)Gr-1(+) myeloid-derived suppressor cells at tumor site. Mixed bone marrow adoptive transfer studies further reveals a T cell-intrinsic role for USP15 in regulating IFN-γ production and tumor development. These findings suggest that T cell intrinsic USP15 deficiency causes excessive production of IFN-γ, which promotes an immunosuppressive tumor microenvironment during MCA-induced primary tumorigenesis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interferon-gamma/metabolism , Th1 Cells/metabolism , Ubiquitin-Specific Proteases/genetics , Animals , Antibodies, Neutralizing/immunology , B7-H1 Antigen/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemokine CXCL12/metabolism , Fibrosarcoma/chemically induced , Fibrosarcoma/mortality , Fibrosarcoma/pathology , Forkhead Transcription Factors/metabolism , Interferon-gamma/immunology , Methylcholanthrene/toxicity , Mice , Mice, Knockout , Signal Transduction , Survival Rate , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Tumor Microenvironment , Ubiquitin-Specific Proteases/deficiency
9.
Nature ; 518(7539): 417-21, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25470037

ABSTRACT

T-helper type 17 (TH17) cells that produce the cytokines interleukin-17A (IL-17A) and IL-17F are implicated in the pathogenesis of several autoimmune diseases. The differentiation of TH17 cells is regulated by transcription factors such as RORγt, but post-translational mechanisms preventing the rampant production of pro-inflammatory IL-17A have received less attention. Here we show that the deubiquitylating enzyme DUBA is a negative regulator of IL-17A production in T cells. Mice with DUBA-deficient T cells developed exacerbated inflammation in the small intestine after challenge with anti-CD3 antibodies. DUBA interacted with the ubiquitin ligase UBR5, which suppressed DUBA abundance in naive T cells. DUBA accumulated in activated T cells and stabilized UBR5, which then ubiquitylated RORγt in response to TGF-ß signalling. Our data identify DUBA as a cell-intrinsic suppressor of IL-17 production.


Subject(s)
Interleukin-17/biosynthesis , Protein Biosynthesis , Th17 Cells/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Enzyme Stability , Female , Inflammation/genetics , Inflammation/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Signal Transduction , Substrate Specificity , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/biosynthesis , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics , Ubiquitination
10.
J Exp Med ; 211(9): 1759-77, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25113974

ABSTRACT

Histone ubiquitination at DNA breaks is required for activation of the DNA damage response (DDR) and DNA repair. How the dynamic removal of this modification by deubiquitinating enzymes (DUBs) impacts genome maintenance in vivo is largely unknown. To address this question, we generated mice deficient for Ub-specific protease 3 (USP3; Usp3Δ/Δ), a histone H2A DUB which negatively regulates ubiquitin-dependent DDR signaling. Notably, USP3 deletion increased the levels of histone ubiquitination in adult tissues, reduced the hematopoietic stem cell (HSC) reserves over time, and shortened animal life span. Mechanistically, our data show that USP3 is important in HSC homeostasis, preserving HSC self-renewal, and repopulation potential in vivo and proliferation in vitro. A defective DDR and unresolved spontaneous DNA damage contribute to cell cycle restriction of Usp3Δ/Δ HSCs. Beyond the hematopoietic system, Usp3Δ/Δ animals spontaneously developed tumors, and primary Usp3Δ/Δ cells failed to preserve chromosomal integrity. These findings broadly support the regulation of chromatin ubiquitination as a key pathway in preserving tissue function through modulation of the response to genotoxic stress.


Subject(s)
DNA Damage/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Carcinogenesis , Cell Proliferation , Cellular Senescence , DNA Breaks, Double-Stranded , DNA Repair/physiology , Female , Histones/metabolism , Homeostasis , Lymphopenia/etiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...