Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 596
Filter
1.
Nat Commun ; 15(1): 8183, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294185

ABSTRACT

Synthesis and maturation of Okazaki Fragments is an incessant and highly efficient metabolic process completing the synthesis of the lagging strands at replication forks during S phase. Accurate Okazaki fragment maturation (OFM) is crucial to maintain genome integrity and, therefore, cell survival in all living organisms. In eukaryotes, OFM involves the consecutive action of DNA polymerase Pol ∂, 5' Flap endonuclease Fen1 and DNA ligase I, and constitutes the best example of a sequential process coordinated by the sliding clamp PCNA. For OFM to occur efficiently, cooperation of these enzymes with PCNA must be highly regulated. Here, we present evidence of a role for the K164-PCNA-deubiquitylase Ubp10 in the maturation of Okazaki fragments in the budding yeast Saccharomyces cerevisiae. We show that Ubp10 associates with lagging-strand DNA synthesis machineries on replicating chromatin to ensure timely ligation of Okazaki fragments by promoting PCNA dissociation from chromatin requiring lysine 164 deubiquitylation.


Subject(s)
Chromatin , DNA Replication , Proliferating Cell Nuclear Antigen , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Chromatin/metabolism , DNA/metabolism , Ubiquitination , Endopeptidases/metabolism , DNA, Fungal/metabolism , DNA, Fungal/genetics , Deubiquitinating Enzymes/metabolism , Flap Endonucleases/metabolism , Flap Endonucleases/genetics , DNA Ligase ATP/metabolism , DNA Ligase ATP/genetics , Ubiquitin Thiolesterase
2.
Cell Death Dis ; 15(9): 693, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333496

ABSTRACT

Circular RNAs (circRNAs) are a type of regulatory RNA that feature covalently closed single-stranded loops. Evidence suggested that circRNAs play important roles in the progression and development of various cancers. However, the impact of circRNA on autophagy-mediated progression of colorectal cancer (CRC) remains unclear. The objective of this project was to investigate the influence of circSEC24B on autophagy and its underlying mechanisms in CRC. To validate the presence and circular structure of circSEC24B in CRC cells and tissues, PCR and Sanger sequencing techniques were employed. Drug resistance and invasive phenotype of CRC cells were evaluated using CCK8, transwell, and Edu assays. Gain- and loss-of-function experiments were conducted to assess the effects of circSEC24B and its protein partner on the growth, invasion, and metastasis of CRC cells in vitro and in vivo. Interactions between circSEC24B, OTUB1, and SRPX2 were analyzed through immunofluorescence, RNA-pulldown, and RIP assays. Mass spectrometry analysis was used to identify potential binding proteins of circRNA in CRC cells. Vectors were constructed to investigate the specific structural domain of the deubiquitinating enzyme OTUB1 that binds to circSEC24B. Results showed that circSEC24B expression was increased in CRC tissues and cell lines, and it enhanced CRC cell proliferation and autophagy levels. Mechanistically, circSEC24B promoted CRC cell proliferation by regulating the protein stability of SRPX2. Specifically, circSEC24B acted as a scaffold, facilitating the binding of OTUB1 to SRPX2 and thereby enhancing its protein stability. Additionally, evidence suggested that OTUB1 regulated SRPX2 expression through an acetylation-dependent mechanism. In conclusion, this study demonstrated that circSEC24B activated autophagy and induced chemoresistance in CRC by promoting the deubiquitination of SRPX2, mediated by the deubiquitinating enzyme OTUB1.


Subject(s)
Autophagy , Colorectal Neoplasms , Cysteine Endopeptidases , Deubiquitinating Enzymes , Drug Resistance, Neoplasm , Membrane Proteins , RNA, Circular , Ubiquitination , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Drug Resistance, Neoplasm/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Cell Line, Tumor , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Nude , Animals , Mice , Cell Proliferation , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Male , Female
3.
Nat Commun ; 15(1): 7856, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251623

ABSTRACT

Despite recent advances in systemic therapy for hepatocellular carcinoma (HCC), the prognosis of hepatitis B virus (HBV)-induced HCC patients remains poor. By screening a sgRNA library targeting human deubiquitinases, we find that ubiquitin-specific peptidase 26 (USP26) deficiency impairs HBV-positive HCC cell proliferation. Genetically engineered murine models with Usp26 knockout confirm that Usp26 drives HCC tumorigenesis. Mechanistically, we find that the HBV-encoded protein HBx binds to the promoter and induces the production of USP26, which is an X-linked gene exclusively expressed in the testis. HBx consequently promotes the association of USP26 with SIRT1 to synergistically stabilize SIRT1 by deubiquitination, which promotes cell proliferation and impedes cell apoptosis to accelerate HCC tumorigenesis. In patients with HBV-positive HCC, USP26 is robustly induced, and its levels correlate with SIRT1 levels and poor prognosis. Collectively, our study highlights a causative link between HBV infection, deubiquitinase induction and development of HCC, identifying a druggable target, USP26.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Epigenesis, Genetic , Hepatitis B virus , Liver Neoplasms , Sirtuin 1 , Trans-Activators , Viral Regulatory and Accessory Proteins , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Animals , Liver Neoplasms/virology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Hepatitis B virus/genetics , Mice , Sirtuin 1/metabolism , Sirtuin 1/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Male , Cell Proliferation/genetics , Viral Regulatory and Accessory Proteins/metabolism , Carcinogenesis/genetics , Hepatitis B/virology , Hepatitis B/complications , Hepatitis B/genetics , Hepatitis B/metabolism , Cell Line, Tumor , Mice, Knockout , Gene Expression Regulation, Neoplastic , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Apoptosis/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Promoter Regions, Genetic/genetics
4.
Cell Mol Life Sci ; 81(1): 349, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136771

ABSTRACT

Multiple myeloma (MM) is the second most common hematological tumor in adults. Immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide (Len), are effective drugs for the treatment of multiple myeloma. Len can recruit IKZF1 and IKZF3 to cereblon (CRBN), a substrate receptor of the cullin 4-RING E3 ligase (CRL4), promote their ubiquitination and degradation, and finally inhibit the proliferation of myeloma cells. However, MM patients develop resistance to IMiDs over time, leading to disease recurrence and deterioration. To explore the possible approaches that may enhance the sensitivity of IMiDs to MM, in this study, we used the proximity labeling technique TurboID and quantitative proteomics to identify Lys-63-specific deubiquitinase BRCC36 as a CRBN-interacting protein. Biochemical experiments demonstrated that BRCC36 in the BRISC complex protects CRBN from lysosomal degradation by specifically cleaving the K63-linked polyubiquitin chain on CRBN. Further studies found that a small-molecule compound SHIN1, which binds to BRISC complex subunit SHMT2, can upregulate CRBN by elevating BRCC36. The combination of SHIN1 and Len can further increase the sensitivity of MM cells to IMiDs. Therefore, this study provides the basis for the exploration of a possible strategy for the SHIN1 and Len combination treatment for MM.


Subject(s)
Adaptor Proteins, Signal Transducing , Lenalidomide , Lysosomes , Multiple Myeloma , Ubiquitin-Protein Ligases , Humans , Multiple Myeloma/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Lenalidomide/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Lysosomes/metabolism , Lysosomes/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Ubiquitination/drug effects , Proteolysis/drug effects , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors
5.
Biomed Pharmacother ; 179: 117348, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208669

ABSTRACT

In cancer research, oncogenesis can be affected by modulating the deubiquitination pathway. Ubiquitination regulates proteins post-translationally in variety of physiological processes. The Otubain Subfamily includes OTUB1 (ovarian tumor-associated proteinase B1) and OTUB2(ovarian tumor-associated proteinase B2). They are deubiquitinating enzymes, which are research hotspots in tumor immunotherapy, with their implications extending across the spectrum of tumor development. Understanding their important role in tumorigenesis, includ-ing hepatocellular carcinoma (HCC) is crucial. HCC has alarming global incidence rates and mortality statistics, ranking among the top five prevalent cancers in Malaysia1. Numerous studies have consistently indicated significant expression of OTUB1 and OTUB2 in HCC cells. In addition, OTUB1 has important biological functions in cancer, suggesting its important role in tumorigenesis. However, the mechanism underlying the action of OTUB1 and OTUB2 in liver cancer remains inadequately explored. Therefore, Otubain Subfamily, as potential molecular target, holds promise for advancing HCC treatments. However, further clinical studies are required to verify its efficacy and application prospects.


Subject(s)
Carcinoma, Hepatocellular , Deubiquitinating Enzymes , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Deubiquitinating Enzymes/metabolism , Animals , Cysteine Endopeptidases/metabolism
6.
Aging (Albany NY) ; 16(15): 11553-11567, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39115875

ABSTRACT

BACKGROUND: Breast cancer is a prevalent disease that has a dismal prognosis for patients and a bad outlook for treatments. Ubiquitination is a reversible biological process that regulates protein production and degradation, as well as plays a vital role in protein transport, localization, and biological activity. METHODS: We obtained the breast cancer patient sample data and used a machine learning technique to create a novel index called Deubiquitinating enzyme related index (DUBRI) by gathering genes associated to deubiquitinating enzymes. Based on DUBRI, we systematically analyze patients' prognosis, clinical characteristics, tumor immune microenvironment, chemotherapy response and immunotherapy response. Finally, the function of OTUB2 was explored in breast cancer cells. RESULTS: DUBRI, which consists of five deubiquitinating enzyme genes (OTUB2, USP41, MINDY2, YOD1, and PSMD7), is a reliable predictor of survival in breast cancer patients. We found that the high DUBRI group presented higher levels of immune cell infiltration. We performed molecular docking prediction of core target proteins in deubiquitinating enzymes. In vitro experiments verified that knockdown of OTUB2 could inhibit the proliferation and migration of breast cancer. CONCLUSIONS: The DUBRI discovered in this research may effectively evaluate the outlook of breast cancer patients and identify groups of patients who would gain advantages from immunotherapy, offering vital knowledge for the future targeted treatment of breast cancer patients.


Subject(s)
Breast Neoplasms , Deubiquitinating Enzymes , Immunotherapy , Humans , Breast Neoplasms/immunology , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Female , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Prognosis , Immunotherapy/methods , Tumor Microenvironment/immunology , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Molecular Docking Simulation , Ubiquitination , Machine Learning , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
7.
Int J Biol Macromol ; 277(Pt 4): 134476, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111477

ABSTRACT

The single-stranded RNA genome of SARS-CoV-2 encodes several structural and non-structural proteins, among which the papain-like protease (PLpro) is crucial for viral replication and immune evasion and has emerged as a promising therapeutic target. The current study aims to discover new inhibitors of PLpro that can simultaneously disrupt its protease and deubiquitinase activities. Using multiple computational approaches, six compounds (CP1-CP6) were selected from our in-house compounds database, with higher docking scores (-7.97 kcal/mol to -8.14 kcal/mol) and fitted well in the active pocket of PLpro. Furthermore, utilizing microscale molecular dynamics simulations (MD), the dynamic behavior of selected compounds was studied. Those molecules strongly binds at the PLpro active site and forms stable complexes. The dynamic motions suggest that the binding of CP1-CP6 brought the protein to a closed conformational state, thereby altering its normal function. In an in vitro evaluation, CP2 showed the most significant inhibitory potential for PLpro (protease activity = 2.71 ± 0.33 µM and deubiquitinase activity = 3.11 ± 0.75 µM), followed by CP1, CP5, CP4 and CP6. Additionally, CP1-CP6 showed no cytotoxicity at a concentration of 30 µM in the human BJ cell line.


Subject(s)
Coronavirus Papain-Like Proteases , Deubiquitinating Enzymes , Molecular Docking Simulation , Molecular Dynamics Simulation , SARS-CoV-2 , SARS-CoV-2/enzymology , SARS-CoV-2/drug effects , Humans , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/chemistry , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Catalytic Domain , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Biological Products/pharmacology , Biological Products/chemistry , COVID-19 Drug Treatment , COVID-19/virology , Protein Binding
8.
Cell Commun Signal ; 22(1): 420, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210373

ABSTRACT

BACKGROUND: Colon cancer is one of the most prevalent tumors in the digestive tract, and its stemness feature significantly contribute to chemoresistance, promote the epithelial-mesenchymal transition (EMT) process, and ultimately lead to tumor metastasis. Therefore, it is imperative for researchers to elucidate the molecular mechanisms underlying the enhancement of stemness feature, chemoresistance, and EMT in colon cancer. METHODS: Sphere-formation and western blotting assays were conducted to assess the stemness feature. Edu, flow cytometry, and cell viability assays were employed to evaluate the chemoresistance. Immunofluorescence and western blotting assays were utilized to detect EMT. Immunoprecipitation, ubiquitination, agarose gel electrophoresis, chromatin immunoprecipitation followed by quantitative PCR (chip-qPCR), and dual luciferase reporter gene assays were employed for mechanistic investigations. RESULTS: We demonstrated a markedly higher expression level of OTUB2 in colon cancer tissues compared to adjacent tissues. Furthermore, elevated OTUB2 expression was closely associated with poor prognosis and distant tumor metastasis. Functional experiments revealed that knockdown of OTUB2 attenuated stemness feature of colon cancer, enhanced its sensitivity to oxaliplatin, inhibited its EMT process, ultimately reduced the ability of tumor metastasis. Conversely, overexpression of OTUB2 exerted opposite effects. Mechanistically, we identified OTUB2 as a deubiquitinase for SP1 protein which bound specifically to SP1 protein, thereby inhibiting K48 ubiquitination of SP1 protein. The SP1 protein functioned as a transcription factor for the GINS1, exerting its regulatory effect by binding to the 1822-1830 region of the GINS1 promoter and enhancing its transcriptional activity. Ultimately, alterations in GINS1 expression directly regulated stemness feature, chemosensitivity, and EMT progression in colon cancer. CONCLUSION: Collectively, the OTUB2/SP1/GINS1 axis played a pivotal role in driving stemness feature, chemoresistance, and EMT in colon cancer. These results shed new light on understanding chemoresistance and metastasis mechanisms involved in colon cancer.


Subject(s)
Colonic Neoplasms , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells , Humans , Epithelial-Mesenchymal Transition/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Male , Animals , Female , Oxaliplatin/pharmacology , Ubiquitination , Middle Aged , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Mice, Nude , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Mice
9.
Int J Biol Sci ; 20(10): 3784-3801, 2024.
Article in English | MEDLINE | ID: mdl-39113709

ABSTRACT

Ubiquitination, a prevalent and highly dynamic reversible post-translational modification, is tightly regulated by the deubiquitinating enzymes (DUBs) superfamily. Among them, OTU Domain-Containing Ubiquitin Aldehyde-Binding Protein 1 (OTUB1) stands out as a critical member of the OTU deubiquitinating family, playing a pivotal role as a tumor regulator across various cancers. However, its specific involvement in BLCA (BLCA) and its clinical significance have remained ambiguous. This study aimed to elucidate the biofunctions of OTUB1 in BLCA and its implications for clinical prognosis. Our investigation revealed heightened OTUB1 expression in BLCA, correlating with unfavorable clinical outcomes. Through in vivo and in vitro experiments, we demonstrated that increased OTUB1 levels promote BLCA tumorigenesis and progression, along with conferring resistance to cisplatin treatment. Notably, we established a comprehensive network involving OTUB1, ß-catenin, necroptosis, and BLCA, delineating their regulatory interplay. Mechanistically, we uncovered that OTUB1 exerts its influence by deubiquitinating and stabilizing ß-catenin, leading to its nuclear translocation. Subsequently, nuclear ß-catenin enhances the transcriptional activity of c-myc and cyclin D1 while suppressing the expression of RIPK3 and MLKL, thereby fostering BLCA progression and cisplatin resistance. Importantly, our clinical data suggest that the OTUB1/ß-catenin/RIPK3/MLKL axis holds promise as a potential biomarker for BLCA.


Subject(s)
Cysteine Endopeptidases , Signal Transduction , beta Catenin , Humans , beta Catenin/metabolism , Animals , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Mice , Deubiquitinating Enzymes/metabolism , Cell Line, Tumor , Mice, Nude , Ubiquitination , Cisplatin/pharmacology , Cisplatin/therapeutic use
10.
EMBO J ; 43(17): 3677-3709, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009674

ABSTRACT

The ubiquitination and proteasome-mediated degradation of Hypoxia Inducible Factors (HIFs) is central to metazoan oxygen-sensing, but the involvement of deubiquitinating enzymes (DUBs) in HIF signalling is less clear. Here, using a bespoke DUBs sgRNA library we conduct CRISPR/Cas9 mutagenesis screens to determine how DUBs are involved in HIF signalling. Alongside defining DUBs involved in HIF activation or suppression, we identify USP43 as a DUB required for efficient activation of a HIF response. USP43 is hypoxia regulated and selectively associates with the HIF-1α isoform, and while USP43 does not alter HIF-1α stability, it facilitates HIF-1 nuclear accumulation and binding to its target genes. Mechanistically, USP43 associates with 14-3-3 proteins in a hypoxia and phosphorylation dependent manner to increase the nuclear pool of HIF-1. Together, our results highlight the multifunctionality of DUBs, illustrating that they can provide important signalling functions alongside their catalytic roles.


Subject(s)
Deubiquitinating Enzymes , Hypoxia-Inducible Factor 1, alpha Subunit , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Mutagenesis , CRISPR-Cas Systems , HEK293 Cells , Signal Transduction , Ubiquitination
11.
Redox Biol ; 75: 103287, 2024 09.
Article in English | MEDLINE | ID: mdl-39079388

ABSTRACT

Hepatic ischemia/reperfusion (I/R) injury is an important cause of liver function impairment following liver surgery. The ubiquitin-proteasome system (UPS) plays a crucial role in protein quality control and has substantial impact on the hepatic I/R process. Although OTU deubiquitinase 1 (OTUD1) is involved in diverse biological processes, its specific functional implications in hepatic I/R are not yet fully understood. This study demonstrates that OTUD1 alleviates oxidative stress, apoptosis, and inflammation induced by hepatic I/R injury. Mechanistically, OTUD1 deubiquitinates and activates nuclear factor erythroid 2-related factor 2 (NRF2) through its catalytic site cysteine 320 residue and ETGE motif, thereby attenuating hepatic I/R injury. Additionally, administration of a short peptide containing the ETGE motif significantly mitigates hepatic I/R injury in mice. Overall, our study elucidates the mechanism and role of OTUD1 in ameliorating hepatic I/R injury, providing a theoretical basis for potential treatment using ETGE-peptide.


Subject(s)
Liver , NF-E2-Related Factor 2 , Oxidative Stress , Reperfusion Injury , Animals , Humans , Male , Mice , Apoptosis , Deubiquitinating Enzymes/metabolism , Disease Models, Animal , Liver/metabolism , Liver/pathology , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
12.
Sci Rep ; 14(1): 15696, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977909

ABSTRACT

As the largest organ in the human body, skeletal muscle is essential for breathing support, movement initiation, and maintenance homeostasis. It has been shown that programmed cell death (PCD), which includes autophagy, apoptosis, and necrosis, is essential for the development of skeletal muscle. A novel form of PCD called ferroptosis is still poorly understood in relation to skeletal muscle. In this study, we observed that the activation of ferroptosis significantly impeded the differentiation of C2C12 myoblasts into myotubes and concurrently suppressed the expression of OTUB1, a crucial deubiquitinating enzyme. OTUB1-silenced C2C12 mouse myoblasts were used to investigate the function of OTUB1 in ferroptosis. The results show that OTUB1 knockdown in vitro significantly increased C2C12 ferroptosis and inhibited myogenesis. Interestingly, the induction of ferroptosis resulting from OTUB1 knockdown was concomitant with the activation of autophagy. Furthermore, OTUB1 interacted with the P62 protein and stabilized its expression by deubiquitinating it, thereby inhibiting autophagy-dependent ferroptosis and promoting myogenesis. All of these findings demonstrate the critical role that OTUB1 plays in controlling ferroptosis, and we suggest that focusing on the OTUB1-P62 axis may be a useful tactic in the treatment and prevention of disorders involving the skeletal muscle.


Subject(s)
Autophagy , Cell Differentiation , Cysteine Endopeptidases , Ferroptosis , Muscle Development , Muscle Fibers, Skeletal , Myoblasts , Animals , Mice , Muscle Fibers, Skeletal/metabolism , Ferroptosis/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Myoblasts/metabolism , Myoblasts/cytology , Cell Line , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Ubiquitination , Humans , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics
13.
Nature ; 631(8022): 843-849, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39020180

ABSTRACT

Ubiquitination pathways have crucial roles in protein homeostasis, signalling and innate immunity1-3. In these pathways, an enzymatic cascade of E1, E2 and E3 proteins conjugates ubiquitin or a ubiquitin-like protein (Ubl) to target-protein lysine residues4. Bacteria encode ancient relatives of E1 and Ubl proteins involved in sulfur metabolism5,6, but these proteins do not mediate Ubl-target conjugation, leaving open the question of whether bacteria can perform ubiquitination-like protein conjugation. Here we demonstrate that a bacterial operon associated with phage defence islands encodes a complete ubiquitination pathway. Two structures of a bacterial E1-E2-Ubl complex reveal striking architectural parallels with canonical eukaryotic ubiquitination machinery. The bacterial E1 possesses an amino-terminal inactive adenylation domain and a carboxy-terminal active adenylation domain with a mobile α-helical insertion containing the catalytic cysteine (CYS domain). One structure reveals a pre-reaction state with the bacterial Ubl C terminus positioned for adenylation, and a second structure mimics an E1-to-E2 transthioesterification state with the E1 CYS domain adjacent to the bound E2. We show that a deubiquitinase in the same pathway preprocesses the bacterial Ubl, exposing its C-terminal glycine for adenylation. Finally, we show that the bacterial E1 and E2 collaborate to conjugate Ubl to target-protein lysine residues. Together, these data reveal that bacteria possess bona fide ubiquitination systems with strong mechanistic and architectural parallels to canonical eukaryotic ubiquitination pathways, suggesting that these pathways arose first in bacteria.


Subject(s)
Bacterial Proteins , Bacteriophages , Escherichia , Ubiquitin-Activating Enzymes , Ubiquitin-Conjugating Enzymes , Ubiquitination , Ubiquitins , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacteriophages/chemistry , Bacteriophages/immunology , Bacteriophages/metabolism , Catalytic Domain , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/metabolism , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/metabolism , Escherichia/chemistry , Escherichia/enzymology , Escherichia/immunology , Escherichia/virology , Evolution, Molecular , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Operon/genetics , Protein Domains , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitins/metabolism , Ubiquitins/chemistry , Eukaryota/enzymology , Eukaryota/metabolism
14.
Nature ; 631(8022): 850-856, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39020165

ABSTRACT

Several immune pathways in humans conjugate ubiquitin-like proteins to virus and host molecules as a means of antiviral defence1-5. Here we studied an antiphage defence system in bacteria, comprising a ubiquitin-like protein, ubiquitin-conjugating enzymes E1 and E2, and a deubiquitinase. We show that during phage infection, this system specifically conjugates the ubiquitin-like protein to the phage central tail fibre, a protein at the tip of the tail that is essential for tail assembly as well as for recognition of the target host receptor. Following infection, cells encoding this defence system release a mixture of partially assembled, tailless phage particles and fully assembled phages in which the central tail fibre is obstructed by the covalently attached ubiquitin-like protein. These phages show severely impaired infectivity, explaining how the defence system protects the bacterial population from the spread of phage infection. Our findings demonstrate that conjugation of ubiquitin-like proteins is an antiviral strategy conserved across the tree of life.


Subject(s)
Bacterial Proteins , Bacteriophages , Deubiquitinating Enzymes , Escherichia coli , Ubiquitin-Conjugating Enzymes , Ubiquitins , Virus Assembly , Bacteriophages/chemistry , Bacteriophages/metabolism , Bacteriophages/pathogenicity , Bacteriophages/physiology , Deubiquitinating Enzymes/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Escherichia coli/virology , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/metabolism , Viral Tail Proteins/metabolism , Viral Tail Proteins/chemistry , Bacterial Proteins/metabolism , Evolution, Molecular , Conserved Sequence
15.
Mol Cancer ; 23(1): 148, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39048965

ABSTRACT

Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.


Subject(s)
Deubiquitinating Enzymes , Neoplasms , Proteasome Endopeptidase Complex , Ubiquitination , Humans , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Proteasome Endopeptidase Complex/metabolism , Deubiquitinating Enzymes/metabolism , Proteolysis , Ubiquitin/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Protein Processing, Post-Translational , Molecular Targeted Therapy , Ubiquitin-Protein Ligases/metabolism
16.
Insect Biochem Mol Biol ; 172: 104162, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067716

ABSTRACT

Deubiquitinases (DUBs) are essential for the maintenance of protein homeostasis and assembly of proteins into functional complexes. Despite growing interest in DUBs biological functions, the roles of DUBs in regulating intestinal stem cells (ISCs) and gut homeostasis remain largely unknown. Here, we perform an in vivo RNAi screen through induced knock-down of DUBs expression in adult midgut ISCs and enteroblasts (EBs) to identify DUB regulators of intestinal homeostasis in Drosophila. We screen 43 DUBs and identify 8 DUBs that are required for ISCs homeostasis. Knocking-down of usp1, CG7857, usp5, rpn8, usp10 and csn5 decreases the number of ISCs/EBs, while knocking-down of CG4968 and usp8 increases the number of ISCs/EBs. Moreover, knock-down of usp1, CG4968, CG7857, or rpn8 in ISCs/EBs disrupts the intestinal barrier integrity and shortens the lifespan, indicating the requirement of these DUBs for the maintenance of gut homeostasis. Furthermore, we provide evidences that USP1 mediates ISC lineage differentiation via modulating the Notch signaling activity. Our study identifies, for the first time, the deubiquitinases required for the maintenance of intestinal homeostasis in Drosophila, and provide new insights into the functional links between the DUBs and intestinal homeostasis.


Subject(s)
Deubiquitinating Enzymes , Drosophila Proteins , Homeostasis , Intestines , RNA Interference , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Stem Cells/metabolism , Drosophila/genetics , Drosophila/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics
17.
Antiviral Res ; 228: 105944, 2024 08.
Article in English | MEDLINE | ID: mdl-38914283

ABSTRACT

SARS-CoV-2 papain-like protease (PLpro) could facilitate viral replication and host immune evasion by respectively hydrolyzing viral polyprotein and host ubiquitin conjugates, thereby rendering itself as an important antiviral target. Yet few noncovalent PLpro inhibitors of SARS-CoV-2 have been reported with improved directed towards pathogenic deubiquitinating activities inhibition. Herein, we report that coronavirus PLpro proteases have distinctive substrate bias and are conserved to deubiquitylate K63-linked polyubiquitination, thereby attenuating host type I interferon response. We identify a noncovalent compound specifically optimized towards halting the K63-deubiquitinase activity of SARS-CoV-2 PLpro, but not other coronavirus (CoV) counterparts or host deubiquitinase. Contrasting with GRL-0617, a SARS-CoV-1 PLpro inhibitor, SIMM-036 is 50-fold and 7-fold (half maximal inhibitory concentration (IC50)) more potent to inhibit viral replication during SARS-CoV-2 infection and restore the host interferon-ß (IFN-ß) response in human angiotensin-converting enzyme 2 (hACE2)-HeLa cells, respectively. Structure-activity relationship (SAR) analysis further reveals the importance of BL2 groove of PLpro, which could determine the selectivity of K63-deubiquitinase activity of the enzyme.


Subject(s)
Antiviral Agents , SARS-CoV-2 , Virus Replication , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism , Coronavirus Papain-Like Proteases/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , COVID-19/virology , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/metabolism , Ubiquitination/drug effects , COVID-19 Drug Treatment , Vero Cells , Chlorocebus aethiops , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Animals , HEK293 Cells
18.
Nat Commun ; 15(1): 5441, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937512

ABSTRACT

Recent studies have shown the crucial role of podocyte injury in the development of diabetic kidney disease (DKD). Deubiquitinating modification of proteins is widely involved in the occurrence and development of diseases. Here, we explore the role and regulating mechanism of a deubiquitinating enzyme, OTUD5, in podocyte injury and DKD. RNA-seq analysis indicates a significantly decreased expression of OTUD5 in HG/PA-stimulated podocytes. Podocyte-specific Otud5 knockout exacerbates podocyte injury and DKD in both type 1 and type 2 diabetic mice. Furthermore, AVV9-mediated OTUD5 overexpression in podocytes shows a therapeutic effect against DKD. Mass spectrometry and co-immunoprecipitation experiments reveal an inflammation-regulating protein, TAK1, as the substrate of OTUD5 in podocytes. Mechanistically, OTUD5 deubiquitinates K63-linked TAK1 at the K158 site through its active site C224, which subsequently prevents the phosphorylation of TAK1 and reduces downstream inflammatory responses in podocytes. Our findings show an OTUD5-TAK1 axis in podocyte inflammation and injury and highlight the potential of OTUD5 as a promising therapeutic target for DKD.


Subject(s)
Diabetic Nephropathies , Inflammation , MAP Kinase Kinase Kinases , Mice, Knockout , Podocytes , Ubiquitination , Animals , Humans , Male , Mice , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , HEK293 Cells , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Mice, Inbred C57BL , Phosphorylation , Podocytes/metabolism , Podocytes/pathology , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics
19.
J Med Chem ; 67(13): 10710-10742, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38897928

ABSTRACT

Upon infection by an intracellular pathogen, host cells activate apoptotic pathways to limit pathogen replication. Consequently, efficient proliferation of the obligate intracellular pathogen Chlamydia trachomatis, a major cause of trachoma and sexually transmitted diseases, depends on the suppression of host cell apoptosis. C. trachomatis secretes deubiquitinase ChlaDUB1 into the host cell, leading among other interactions to the stabilization of antiapoptotic proteins and, thus, suppression of host cell apoptosis. Targeting the bacterial effector protein may, therefore, lead to new therapeutic possibilities. To explore the active site of ChlaDUB1, an iterative cycle of computational docking, synthesis, and enzymatic screening was applied with the aim of lead structure development. Hereby, covalent inhibitors were developed, which show enhanced inhibition with a 22-fold increase in IC50 values compared to previous work. Comprehensive insights into the binding prerequisites to ChlaDUB1 are provided, establishing the foundation for an additional specific antichlamydial therapy by small molecules.


Subject(s)
Chlamydia trachomatis , Drug Design , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/enzymology , Structure-Activity Relationship , Molecular Docking Simulation , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/metabolism , Molecular Structure , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism
20.
Biol Direct ; 19(1): 46, 2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38880876

ABSTRACT

BACKGROUND: There is growing evidence indicating that deubiquitinating enzymes may contribute to tumor progression and can serve as promising therapeutic targets. METHODS: The overexpression of deubiquitinase OTUD6B in lung adenocarcinoma (LUAD) and its adjacent tissues was analyzed by immunohistochemistry and TCGA/GO database. Survival analysis further supported OTUD6B as a potential target for LUAD treatment. We assessed the effect of OTUD6B on LUAD cell growth using cell viability assays and conducted TUNEL staining, migration, and invasion experiments to investigate the impact of OTUD6B on the apoptosis and metastasis of LUAD cells. Additionally, we established a transplanted tumor model in nude mice to validate our findings in vivo. Finally, using IP mass spectrometry and co-IP experiments, we screened and confirmed the influence of RIPK1 as a substrate of OTUD6B in LUAD. RESULTS: OTUD6B is highly overexpressed in human LUAD and predicts poor prognosis in LUAD patients. OTUD6B knockdown inhibited the proliferation of LUAD cells and enhanced apoptosis and inhibited metastasis in LUAD cells suppressed. A549 xenografts revealed that OTUD6B deletion can slow down tumour growth. Additionally, OTUD6B can bind to RIPK1, reduce its ubiquitination level and increase its protein stability. CONCLUSIONS: Our results suggest that OTUD6B is a promising clinical target for LUAD treatment and that targeting OTUD6B may constitute an effective anti-LUAD strategy.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mice, Nude , Receptor-Interacting Protein Serine-Threonine Kinases , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Disease Progression , Cell Proliferation , Apoptosis , Cell Line, Tumor , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , A549 Cells , Ubiquitination , Protein Stability , Endopeptidases/metabolism , Endopeptidases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL