Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 681
Filter
1.
J Cell Mol Med ; 28(16): e70037, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39183260

ABSTRACT

Ubiquitin like modifier activating enzyme 1 (UBA1) plays an important role in immune regulation and cellular function. However, the functional mechanism and role of UBA1 in pan-cancer have not been fully elucidated and its value in haematological tumours (diffuse large B cell lymphoma (DLBC/DLBCL) and acute myeloid leukaemia (AML/LAML)) has not been explored. We conducted a comprehensive analysis of the functional mechanism and role of UBA1 in pan-cancer using multiple databases, including differential expression analysis, clinical pathological staging analysis, prognosis analysis and immune analysis. Then, we confirmed the function of UBA1 in haematological tumours through cell experiments. The results showed that the expression of UBA1 was significantly increased in most cancers and the differential expression of UBA1 was mainly concentrated in digestive tumours, haematological tumours and brain tumours. Moreover, the high expression of UBA1 had poor prognosis in most tumours, which may be related to its involvement in various cancer-related pathways such as cell cycle, as well as its methylation level, protein phosphorylation level, immune cell infiltration and immune therapy response. Cell experiments have confirmed that UBA1 can significantly regulate the cycle progression and apoptosis of DLBCL cells and AML cells. Therefore, UBA1 may be a potential therapeutic target for haematological tumours. In summary, our study not only comprehensively analysed the functional mechanisms and clinical value of UBA1 in pan-cancer, but also validated for the first time the regulatory role of UBA1 in haematological tumours.


Subject(s)
Computational Biology , Gene Expression Regulation, Neoplastic , Ubiquitin-Activating Enzymes , Humans , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , Prognosis , Computational Biology/methods , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/genetics , Cell Line, Tumor , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Apoptosis , Cell Proliferation
2.
Cell Death Dis ; 15(7): 544, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085203

ABSTRACT

UFMylation is a highly conserved ubiquitin-like post-translational modification that catalyzes the covalent linkage of UFM1 to its target proteins. This modification plays a critical role in the maintenance of endoplasmic reticulum proteostasis, DNA damage response, autophagy, and transcriptional regulation. Mutations in UFM1, as well as in its specific E1 enzyme UBA5 and E2 enzyme UFC1, have been genetically linked to microcephaly. Our previous research unveiled the important role of UFMylation in regulating mitosis. However, the underlying mechanisms have remained unclear due to the limited identification of substrates. In this study, we identified Eg5, a motor protein crucial for mitotic spindle assembly and maintenance, as a novel substrate for UFMylation and identified Lys564 as the crucial UFMylation site. UFMylation did not alter its transcriptional level, phosphorylation level, or protein stability, but affected the mono-ubiquitination of Eg5. During mitosis, Eg5 and UFM1 co-localize at the centrosome and spindle apparatus, and defective UFMylation leads to diminished spindle localization of Eg5. Notably, the UFMylation-defective Eg5 mutant (K564R) exhibited shorter spindles, metaphase arrest, spindle checkpoint activation, and a failure of cell division in HeLa cells. Overall, Eg5 UFMylation is essential for proper spindle organization, mitotic progression, and cell proliferation.


Subject(s)
Kinesins , Mitosis , Spindle Apparatus , Ubiquitination , Humans , Spindle Apparatus/metabolism , HeLa Cells , Kinesins/metabolism , Kinesins/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , HEK293 Cells , Centrosome/metabolism , Protein Processing, Post-Translational , Proteins
3.
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
4.
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
5.
Cell Rep Med ; 5(7): 101653, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39019009

ABSTRACT

Drug-induced liver injury (DILI) is a significant cause of acute liver failure (ALF) and liver transplantation in the Western world. Acetaminophen (APAP) overdose is a main contributor of DILI, leading to hepatocyte cell death through necrosis. Here, we identified that neddylation, an essential post-translational modification involved in the mitochondria function, was upregulated in liver biopsies from patients with APAP-induced liver injury (AILI) and in mice treated with an APAP overdose. MLN4924, an inhibitor of the neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8)-activating enzyme (NAE-1), ameliorated necrosis and boosted liver regeneration in AILI. To understand how neddylation interferes in AILI, whole-body biotinylated NEDD8 (bioNEDD8) and ubiquitin (bioUB) transgenic mice were investigated under APAP overdose with and without MLN4924. The cytidine diphosphate diacylglycerol (CDP-DAG) synthase TAM41, responsible for producing cardiolipin essential for mitochondrial activity, was found modulated under AILI and restored its levels by inhibiting neddylation. Understanding this ubiquitin-like crosstalk in AILI is essential for developing promising targeted inhibitors for DILI treatment.


Subject(s)
Acetaminophen , Cardiolipins , Chemical and Drug Induced Liver Injury , Cyclopentanes , NEDD8 Protein , Pyrimidines , Acetaminophen/adverse effects , Animals , NEDD8 Protein/metabolism , NEDD8 Protein/genetics , Humans , Pyrimidines/pharmacology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Cardiolipins/metabolism , Mice , Cyclopentanes/pharmacology , Male , Liver/metabolism , Liver/pathology , Liver/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Signal Transduction/drug effects , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/antagonists & inhibitors
6.
Biomolecules ; 14(7)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-39062453

ABSTRACT

(1) Background: The neddylation pathway assumes a pivotal role in the initiation and progression of cancer. MLN4924, a potent small-molecule inhibitor of the NEDD8-activating enzyme (NAE), effectively intervenes in the early stages of the neddylation pathway. By instigating diverse cellular responses, such as senescence and apoptosis in cancer cells, MLN4924 also exerts regulatory effects on non-malignant cells within the tumor microenvironment (TME) and tumor virus-infected cells, thereby impeding the onset of tumors. Consequently, MLN4924 has been widely acknowledged as a potent anti-cancer drug. (2) Recent findings: Nevertheless, recent findings have illuminated additional facets of the neddylation pathway, revealing its active involvement in various biological processes detrimental to the survival of cancer cells. This newfound understanding underscores the dual role of MLN4924 in tumor therapy, characterized by both anti-cancer and pro-cancer effects. This dichotomy is herein referred to as the "double-edged effects" of MLN4924. This paper delves into the intricate relationship between the neddylation pathway and cancer, offering a mechanistic exploration and analysis of the causes underlying the double-edged effects of MLN4924-specifically, the accumulation of pro-cancer neddylation substrates. (3) Perspectives: Here, the objective is to furnish theoretical support and novel insights that can guide the development of next-generation anti-cancer drugs targeting the neddylation pathway.


Subject(s)
Antineoplastic Agents , Cyclopentanes , NEDD8 Protein , Neoplasms , Pyrimidines , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Humans , Pyrimidines/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , NEDD8 Protein/metabolism , Animals , Signal Transduction/drug effects , Apoptosis/drug effects , Tumor Microenvironment/drug effects , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/antagonists & inhibitors
7.
Exp Cell Res ; 440(2): 114148, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38936760

ABSTRACT

UBA5, a ubiquitin-like activated enzyme involved in ufmylation and sumoylation, presents a viable target for pancreatic and breast cancer treatments, yet its role in lung adenocarcinoma (LUAD) remains underexplored. This study reveals UBA5's tumor-promoting effect in LUAD, as evidenced by its upregulation in patients and positive correlation with TNM stages. Elevated UBA5 levels predict poor outcomes for these patients. Pharmacological inhibition of UBA5 using DKM 2-93 significantly curtails the growth of A549, H1299, and cisplatin-resistant A549 (A549/DDP) LUAD cells in vitro. Additionally, UBA5 knockdown via shRNA lentivirus suppresses tumor growth both in vitro and in vivo. High UBA5 expression adversely alters the tumor immune microenvironment, affecting immunostimulators, MHC molecules, chemokines, receptors, and immune cell infiltration. Notably, UBA5 expression correlates positively with M2 macrophage infiltration, the predominant immune cells in LUAD. Co-culture experiments further demonstrate that UBA5 knockdown directly inhibits M2 macrophage polarization and lactate production in LUAD. Moreover, in vivo studies show reduced M2 macrophage infiltration following UBA5 knockdown. UBA5 expression is also associated with increased tumor heterogeneity, including tumor mutational burden, microsatellite instability, neoantigen presence, and homologous recombination deficiency. Experiments indicate that UBA5 overexpression promotes cisplatin resistance in vitro, whereas UBA5 inhibition enhances cisplatin sensitivity in both in vitro and in vivo settings. Overall, these findings suggest that targeting UBA5 inhibits LUAD by impeding cancer cell proliferation, M2 macrophage polarization, and cisplatin resistance.


Subject(s)
Adenocarcinoma of Lung , Cisplatin , Drug Resistance, Neoplasm , Lung Neoplasms , Humans , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Animals , Mice , Cell Proliferation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Female , Tumor Microenvironment/drug effects , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Male , Gene Expression Regulation, Neoplastic/drug effects
8.
Bioorg Med Chem Lett ; 107: 129779, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38729317

ABSTRACT

Targeted protein degradation is mediated by small molecules that induce or stabilize protein-protein interactions between targets and the ubiquitin-proteasome machinery. Currently, there remains a need to expand the repertoire of viable E3 ligases available for hijacking. Notably, covalent chemistry has been employed to engage a handful of E3 ligases, including DCAF11. Here, we disclose a covalent PROTAC that enables DCAF11-dependent degradation, featuring a cyanoacrylamide warhead. Our findings underscore DCAF11 as an interesting candidate with a capacity to accommodate diverse electrophilic chemistries compatible with targeted protein degradation.


Subject(s)
Acrylamides , Humans , Acrylamides/chemistry , Acrylamides/pharmacology , Acrylamides/chemical synthesis , Molecular Structure , Proteolysis/drug effects , Drug Discovery , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Structure-Activity Relationship
9.
Free Radic Biol Med ; 219: 127-140, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38614228

ABSTRACT

Doxorubicin (DOX) is a widely utilized chemotherapeutic agent in clinical oncology for treating various cancers. However, its clinical use is constrained by its significant side effects. Among these, the development of cardiomyopathy, characterized by cardiac remodeling and eventual heart failure, stands as a major concern following DOX chemotherapy. In our current investigation, we have showcased the efficacy of MLN4924 in mitigating doxorubicin-induced cardiotoxicity through direct inhibition of the NEDD8-activating enzyme, NAE. MLN4924 demonstrated the ability to stabilize mitochondrial function post-doxorubicin treatment, diminish cardiomyocyte apoptosis, alleviate oxidative stress-induced damage in the myocardium, enhance cardiac contractile function, mitigate cardiac fibrosis, and impede cardiac remodeling associated with heart failure. At the mechanistic level, MLN4924 intervened in the neddylation process by inhibiting the NEDD8 activating enzyme, NAE, within the murine cardiac tissue subsequent to doxorubicin treatment. This intervention resulted in the suppression of NEDD8 protein expression, reduction in neddylation activity, and consequential manifestation of cardioprotective effects. Collectively, our findings posit MLN4924 as a potential therapeutic avenue for mitigating doxorubicin-induced cardiotoxicity by attenuating heightened neddylation activity through NAE inhibition, thereby offering a viable and promising treatment modality for afflicted patients.


Subject(s)
Cardiotoxicity , Cyclopentanes , Doxorubicin , Myocytes, Cardiac , NEDD8 Protein , Pyrimidines , Animals , Mice , Apoptosis/drug effects , Cardiotoxicity/drug therapy , Cardiotoxicity/pathology , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Doxorubicin/adverse effects , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NEDD8 Protein/metabolism , NEDD8 Protein/antagonists & inhibitors , Oxidative Stress/drug effects , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics
10.
Front Biosci (Landmark Ed) ; 29(4): 144, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38682183

ABSTRACT

BACKGROUND: Gliomas are characterized by aggressive behavior, leading to severe disability and high mortality. Ubiquitin-like modifier activating enzyme 2 (UBA2) is a subunit of the E1-activating enzyme involved in the SUMOylation (SUMO, small ubiquitin-related modifier) of numerous proteins. Although the abnormality of UBA2 is linked to the progression of various tumor types, the role of UBA2 in glioma is still unknown. METHODS: A bioinformatic analysis using several public databases was conducted to examine the expression level, clinicopathological correlations, and prognostic significance of UBA2 in glioma. The correlation between UBA2 expression and drug sensitivity in cancers was also explored. Multiple cellular experiments were conducted to validate the role of UBA2 in glioma. RESULTS: Analysis of multiple databases and cellular experiments revealed that UBA2 was overexpressed in glioma tissues and cell lines, respectively. UBA2 expression in gliomas correlated with World Health Organization (WHO) grade, IDH gene status, 1p19q deletion, histological type, and immune cell infiltration in glioma. UBA2 expression in carcinomas also correlated with drug sensitivity. Kaplan-Meier analysis revealed that high expression of UBA2 predicted poorer survival in glioma patients. A nomogram model containing UBA2 expression was constructed for clinical practice. Knockdown of UBA2 was observed to suppress glioma cell progression and sensitize glioma cells to irradiation in vitro. CONCLUSION: Overall, this research showed that UBA2 might be involved not only in the development of glioma but also in the regulation of immunity, drug sensitivity, and radiosensitivity. Therefore, UBA2 may be a potential target for therapy and a candidate biomarker for glioma diagnosis and prognosis.


Subject(s)
Biomarkers, Tumor , Glioma , Ubiquitin-Activating Enzymes , Glioma/diagnosis , Glioma/immunology , Glioma/mortality , Glioma/therapy , Cell Line, Tumor , Prognosis , Ubiquitin-Activating Enzymes/analysis , Ubiquitin-Activating Enzymes/metabolism , Immunotherapy , Radiation Tolerance , Disease Progression
11.
Leuk Lymphoma ; 65(7): 978-988, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38489672

ABSTRACT

Adult T-cell leukemia (ATL), caused by HTLV-1, is the most lethal hematological malignancy. NEDD8-activating enzyme (NAE) is a component of the NEDD8 conjunction pathway that regulates cullin-RING ubiquitin ligase (CRL) activity. HTLV-1-infected T cells expressed higher levels of NAE catalytic subunit UBA3 than normal peripheral blood mononuclear cells. NAE1 knockdown inhibited proliferation of HTLV-1-infected T cells. The NAE1 inhibitor MLN4924 suppressed neddylation of cullin and inhibited the CRL-mediated turnover of tumor suppressor proteins. MLN4924 inhibited proliferation of HTLV-1-infected T cells by inducing DNA damage, leading to S phase arrest and caspase-dependent apoptosis. S phase arrest was associated with CDK2 and cyclin A downregulation. MLN4924-induced apoptosis was mediated by the upregulation of pro-apoptotic and downregulation of anti-apoptotic proteins. Furthermore, MLN4924 inhibited NF-κB, AP-1, and Akt signaling pathways and activated JNK. Therefore, neddylation inhibition is an attractive strategy for ATL therapy. Our findings support the use of MLN4924 in ATL clinical trials.


Subject(s)
Apoptosis , Cell Proliferation , Cyclopentanes , Human T-lymphotropic virus 1 , NEDD8 Protein , NF-kappa B , Proto-Oncogene Proteins c-akt , Pyrimidines , Signal Transduction , Transcription Factor AP-1 , Ubiquitin-Activating Enzymes , Humans , Pyrimidines/pharmacology , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Cyclopentanes/pharmacology , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects , NEDD8 Protein/metabolism , Cell Proliferation/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Ubiquitins/metabolism , Cullin Proteins/metabolism
12.
Commun Biol ; 7(1): 382, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553562

ABSTRACT

Autophagy is a dynamic self-renovation biological process that maintains cell homeostasis and is responsible for the quality control of proteins, organelles, and energy metabolism. The E1-like ubiquitin-activating enzyme autophagy-related gene 7 (ATG7) is a critical factor that initiates classic autophagy reactions by promoting the formation and extension of autophagosome membranes. Recent studies have identified the key functions of ATG7 in regulating the cell cycle, apoptosis, and metabolism associated with the occurrence and development of multiple diseases. This review summarizes how ATG7 is precisely programmed by genetic, transcriptional, and epigenetic modifications in cells and the relationship between ATG7 and aging-related diseases.


Subject(s)
Autophagosomes , Autophagy , Autophagy-Related Protein 7/genetics , Autophagosomes/metabolism , Autophagy/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
13.
Int J Mol Sci ; 25(5)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38474091

ABSTRACT

Ubiquitin-like modifier-activating enzyme 6 (UBA6) is a member of the E1 enzyme family, which initiates the ubiquitin-proteasome system (UPS). The UPS plays critical roles not only in protein degradation but also in various cellular functions, including neuronal signaling, myocardial remodeling, immune cell differentiation, and cancer development. However, the specific role of UBA6 in cellular functions is not fully elucidated in comparison with the roles of the UPS. It has been known that the E1 enzyme is associated with the motility of cancer cells. In this study, we verified the physiological roles of UBA6 in lung cancer cells through gene-silencing siRNA targeting UBA6 (siUBA6). The siUBA6 treatment attenuated the migration of H1975 cells, along with a decrease in lysosomal Ca2+ release. While autophagosomal proteins remained unchanged, lysosomal proteins, including TRPML1 and TPC2, were decreased in siUBA6-transfected cells. Moreover, siUBA6 induced the production of multivesicular bodies (MVBs), accompanied by an increase in MVB markers in siUBA6-transfected H1975 cells. Additionally, the expression of the exosomal marker CD63 and extracellular vesicles was increased by siUBA6 treatment. Our findings suggest that knock-down of UBA6 induces lysosomal TRPML1 depletion and inhibits endosomal trafficking to lysosome, and subsequently, leads to the accumulation of MVBs and enhanced exosomal secretion in lung cancer cells.


Subject(s)
Lung Neoplasms , Humans , Lung Neoplasms/metabolism , Lysosomes/metabolism , Multivesicular Bodies/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/metabolism
14.
J Transl Med ; 22(1): 148, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38351014

ABSTRACT

Helicobacter pylori (H. pylori) is a major risk factor of gastric cancer (GC). The SUMO-activating enzyme SAE1(SUMO-activating enzyme subunit 1), which is indispensable for protein SUMOylation, involves in human tumorigenesis. In this study, we used the TIMER and TCGA database to explore the SAE1 expression in GC and normal tissues and Kaplan-Meier Plotter platform for survival analysis of GC patients. GC tissue microarray and gastric samples from patients who underwent endoscopic treatment were employed to detect the SAE1expression. Our results showed that SAE1 was overexpressed in GC tissues and higher SAE1 expression was associated with worse clinical characteristics of GC patients. Cell and animal models showed that H. pylori infection upregulated SAE1, SUMO1, and SUMO2/3 protein expression. Functional assays suggested that suppression of SAE1 attenuated epithelial-mesenchymal transition (EMT) biomarkers and cell proliferation abilities induced by H. pylori. Cell and animal models of ROS inhibition in H. pylori showed that ROS could mediate the H. pylori-induced upregulation of SAE1, SUMO1, and SUMO2/3 protein. RNA sequencing was performed and suggested that knockdown of SAE1 could exert an impact on IGF-1 expression. General, increased SUMOylation modification is involved in H. pylori-induced GC.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Animals , Humans , Up-Regulation/genetics , Stomach Neoplasms/pathology , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Reactive Oxygen Species/metabolism , Cell Transformation, Neoplastic , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
15.
EMBO J ; 43(10): 1919-1946, 2024 May.
Article in English | MEDLINE | ID: mdl-38360993

ABSTRACT

Most cellular ubiquitin signaling is initiated by UBA1, which activates and transfers ubiquitin to tens of E2 enzymes. Clonally acquired UBA1 missense mutations cause an inflammatory-hematologic overlap disease called VEXAS (vacuoles, E1, X-linked, autoinflammatory, somatic) syndrome. Despite extensive clinical investigation into this lethal disease, little is known about the underlying molecular mechanisms. Here, by dissecting VEXAS-causing UBA1 mutations, we discovered that p.Met41 mutations alter cytoplasmic isoform expression, whereas other mutations reduce catalytic activity of nuclear and cytoplasmic isoforms by diverse mechanisms, including aberrant oxyester formation. Strikingly, non-p.Met41 mutations most prominently affect transthioesterification, revealing ubiquitin transfer to cytoplasmic E2 enzymes as a shared property of pathogenesis amongst different VEXAS syndrome genotypes. A similar E2 charging bottleneck exists in some lung cancer-associated UBA1 mutations, but not in spinal muscular atrophy-causing UBA1 mutations, which instead, render UBA1 thermolabile. Collectively, our results highlight the precision of conformational changes required for faithful ubiquitin transfer, define distinct and shared mechanisms of UBA1 inactivation in diverse diseases, and suggest that specific E1-E2 modules control different aspects of tissue differentiation and maintenance.


Subject(s)
Ubiquitin-Activating Enzymes , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/genetics , Humans , Mutation, Missense , Ubiquitin/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism
16.
Phytomedicine ; 126: 155148, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387271

ABSTRACT

BACKGROUND: Finding a drug for early intervention in the hepatic fibrosis process has important clinical significance. Previous studies have suggested SUMOylation as a potential target for intervention in hepatic fibrosis. However, the role of SAE1, a marker of SUMOylation, in hepatic fibrosis is unknown. Additionally, whether ginkgolic acid (GA), a SUMOylation inhibitor, inhibits hepatic fibrosis by inhibiting SUMO1-activating enzyme subunit 1 (SAE1) should be further investigated. METHODS: Liver tissues of patients with hepatic cirrhosis and a rat model of hepatic fibrosis constructed with CCl4 (400 mg/kg, twice weekly) or TAA (200 mg/kg, twice weekly) were selected, and the degree of hepatic fibrosis was then evaluated using H&E, Sirius red, and Masson's trichrome staining. After knockdown or overexpression of SAE1 in hepatic stellate cells, the expression levels of ferroptosis and hepatic fibrosis markers were measured in vitro. After intervention with a ferroptosis inhibitor, the expression levels were again measured in vivo and in vitro. RESULTS: We first demonstrated that SAE1 increased in patients with hepatic cirrhosis. Subsequently, testing of the rat hepatic fibrosis model confirmed that GA reduced the expression of SAE1 and improved hepatic fibrosis in rats. Then, we used hepatic stellate cell lines to confirm in vitro that GA inhibited SAE1 expression and induced ferroptosis, and that overexpression of SAE1 or inhibition of ferroptosis reversed this process. Finally, we confirmed in vivo that GA induced ferroptosis and alleviated the progression of hepatic fibrosis, while inhibiting ferroptosis also reversed the progression of hepatic fibrosis in rats. CONCLUSION: SAE1 is a potential anti-fibrotic target protein, and GA induces ferroptosis of hepatic stellate cells by targeting SAE1 to exert an anti-hepatic fibrosis effect, which lays an experimental foundation for the future clinical application of its anti-hepatic fibrosis effect.


Subject(s)
Ferroptosis , Salicylates , Humans , Rats , Animals , Signal Transduction , Liver Cirrhosis/metabolism , Liver , Hepatic Stellate Cells , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/pharmacology
17.
Acta Biochim Biophys Sin (Shanghai) ; 56(2): 199-209, 2024 02 25.
Article in English | MEDLINE | ID: mdl-38298057

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) accounts for approximately 15% of primary liver cancers, and the incidence rate has been increasing in recent years. Surgical resection is the best treatment for ICC, but the 5-year survival rate is less than 30%. ICC signature genes are crucial for the early diagnosis of ICC, so it is especially important to identify signature genes. The aim of this study is to screen the signature genes of ICC and find the potential target for the treatment of ICC. We find that UBA3 is highly expressed in ICC, and knockdown of UBA3 inhibits ICC proliferation, invasion and migration. Mechanistic experiments show that UBA3 promotes ICC proliferation, invasion and migration by affecting ANXA2 through the MAPK signaling pathway. UBA3 is a target of bufalin, and bufalin targeting UBA3 inhibits ICC development and progression through the MAPK signaling pathway. In conclusion, our study shows that bufalin inhibits ICC by targeting UBA3, which has emerged as a new biomarker and potential therapeutic target for ICC.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Ubiquitin-Activating Enzymes , Humans , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Signal Transduction , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
18.
Int J Mol Sci ; 25(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38397039

ABSTRACT

Human brain development involves a tightly regulated sequence of events that starts shortly after conception and continues up to adolescence. Before birth, neurogenesis occurs, implying an extensive differentiation process, sustained by changes in the gene expression profile alongside proteome remodeling, regulated by the ubiquitin proteasome system (UPS) and autophagy. The latter processes rely on the selective tagging with ubiquitin of the proteins that must be disposed of. E3 ubiquitin ligases accomplish the selective recognition of the target proteins. At the late stage of neurogenesis, the brain starts to take shape, and neurons migrate to their designated locations. After birth, neuronal myelination occurs, and, in parallel, neurons form connections among each other throughout the synaptogenesis process. Due to the malfunctioning of UPS components, aberrant brain development at the very early stages leads to neurodevelopmental disorders. Through deep data mining and analysis and by taking advantage of machine learning-based models, we mapped the transcriptomic profile of the genes encoding HECT- and ring-between-ring (RBR)-E3 ubiquitin ligases as well as E2 ubiquitin-conjugating and E1 ubiquitin-activating enzymes during human brain development, from early post-conception to adulthood. The inquiry outcomes unveiled some implications for neurodevelopment-related disorders.


Subject(s)
Ubiquitin-Activating Enzymes , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Brain/metabolism
19.
EMBO J ; 43(2): 250-276, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177505

ABSTRACT

Expansion mutations in polyalanine stretches are associated with a growing number of diseases sharing a high degree of genotypic and phenotypic commonality. These similarities prompted us to query the normal function of physiological polyalanine stretches and to investigate whether a common molecular mechanism is involved in these diseases. Here, we show that UBA6, an E1 ubiquitin-activating enzyme, recognizes a polyalanine stretch within its cognate E2 ubiquitin-conjugating enzyme USE1. Aberrations in this polyalanine stretch reduce ubiquitin transfer to USE1 and, subsequently, polyubiquitination and degradation of its target, the ubiquitin ligase E6AP. Furthermore, we identify competition for the UBA6-USE1 interaction by various proteins with polyalanine expansion mutations in the disease state. The deleterious interactions of expanded polyalanine tract proteins with UBA6 in mouse primary neurons alter the levels and ubiquitination-dependent degradation of E6AP, which in turn affects the levels of the synaptic protein Arc. These effects are also observed in induced pluripotent stem cell-derived autonomic neurons from patients with polyalanine expansion mutations, where UBA6 overexpression increases neuronal resilience to cell death. Our results suggest a shared mechanism for such mutations that may contribute to the congenital malformations seen in polyalanine tract diseases.


Subject(s)
Peptides , Ubiquitin-Activating Enzymes , Ubiquitin , Humans , Animals , Mice , Ubiquitination , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Mutation
20.
Nat Struct Mol Biol ; 31(2): 351-363, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182926

ABSTRACT

UBR4 is a 574 kDa E3 ligase (E3) of the N-degron pathway with roles in neurodevelopment, age-associated muscular atrophy and cancer. The catalytic module that carries out ubiquitin (Ub) transfer remains unknown. Here we identify and characterize a distinct E3 module within human UBR4 consisting of a 'hemiRING' zinc finger, a helical-rich UBR zinc-finger interacting (UZI) subdomain, and an N-terminal region that can serve as an affinity factor for the E2 conjugating enzyme (E2). The structure of an E2-E3 complex provides atomic-level insight into the specificity determinants of the hemiRING toward the cognate E2s UBE2A/UBE2B. Via an allosteric mechanism, the UZI subdomain modestly activates the Ub-loaded E2 (E2∼Ub). We propose attenuated activation is complemented by the intrinsically high lysine reactivity of UBE2A, and their cooperation imparts a reactivity profile important for substrate specificity and optimal degradation kinetics. These findings reveal the mechanistic underpinnings of a neuronal N-degron E3, its specific recruitment of UBE2A, and highlight the underappreciated architectural diversity of cross-brace domains with Ub E3 activity.


Subject(s)
Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/metabolism , Catalysis , Ubiquitination , Calmodulin-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL