Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.699
Filter
1.
Front Immunol ; 15: 1445338, 2024.
Article in English | MEDLINE | ID: mdl-39247192

ABSTRACT

Background: Defective ribosomal products (DRiPs) are non-functional proteins rapidly degraded during or after translation being an essential source for MHC class I ligands. DRiPs are characterized to derive from a substantial subset of nascent gene products that degrade more rapidly than their corresponding native retiree pool. So far, mass spectrometry analysis revealed that a large number of HLA class I peptides derive from DRiPs. However, a specific viral DRiP on protein level was not described. In this study, we aimed to characterize and identify DRiPs derived from a viral protein. Methods: Using the nucleoprotein (NP) of the lymphocytic choriomeningitis virus (LCMV) which is conjugated N-terminally to ubiquitin, or the ubiquitin-like modifiers FAT10 or ISG15 the occurrence of DRiPs was studied. The formation and degradation of DRiPs was monitored by western blot with the help of a FLAG tag. Flow cytometry and cytotoxic T cells were used to study antigen presentation. Results: We identified several short lived DRiPs derived from LCMV-NP. Of note, these DRiPs could only be observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, but not in the wild type form. Using proteasome inhibitors, we could show that degradation of LCMV-NP derived DRiPs were proteasome dependent. Interestingly, the synthesis of DRiPs could be enhanced when cells were stressed with the help of FCS starvation. An enhanced NP118-126 presentation was observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, or under FCS starvation. Conclusion: Taken together, we visualize for the first time DRiPs derived from a viral protein. Furthermore, DRiPs formation, and therefore MHC-I presentation, is enhanced under cellular stress conditions. Our investigations on DRiPs in MHC class I antigen presentation open up new approaches for the development of vaccination strategies.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I , Lymphocytic choriomeningitis virus , Antigen Presentation/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Lymphocytic choriomeningitis virus/immunology , Animals , Humans , Stress, Physiological/immunology , T-Lymphocytes, Cytotoxic/immunology , Mice , Ubiquitins/metabolism , Ubiquitins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/immunology , Proteolysis , Nucleoproteins/immunology , Nucleoproteins/metabolism
2.
Viruses ; 16(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205213

ABSTRACT

Emergence of newer variants of SARS-CoV-2 underscores the need for effective antivirals to complement the vaccination program in managing COVID-19. The multi-functional papain-like protease (PLpro) of SARS-CoV-2 is an essential viral protein that not only regulates the viral replication but also modulates the host immune system, making it a promising therapeutic target. To this end, we developed an in vitro interferon stimulating gene 15 (ISG15)-based Förster resonance energy transfer (FRET) assay and screened the National Cancer Institute (NCI) Diversity Set VI compound library, which comprises 1584 small molecules. Subsequently, we assessed the PLpro enzymatic activity in the presence of screened molecules. We identified three potential PLpro inhibitors, namely, NSC338106, 651084, and 679525, with IC50 values in the range from 3.3 to 6.0 µM. These molecules demonstrated in vitro inhibition of the enzyme activity and exhibited antiviral activity against SARS-CoV-2, with EC50 values ranging from 0.4 to 4.6 µM. The molecular docking of all three small molecules to PLpro suggested their specificity towards the enzyme's active site. Overall, our study contributes promising prospects for further developing potential antivirals to combat SARS-CoV-2 infection.


Subject(s)
Antiviral Agents , Coronavirus Papain-Like Proteases , Cytokines , High-Throughput Screening Assays , SARS-CoV-2 , Ubiquitins , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , High-Throughput Screening Assays/methods , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , Cytokines/metabolism , Ubiquitins/metabolism , Ubiquitins/chemistry , Ubiquitins/antagonists & inhibitors , Molecular Docking Simulation , COVID-19 Drug Treatment , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Fluorescence Resonance Energy Transfer , COVID-19/virology
3.
J Virol ; 98(9): e0086924, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194248

ABSTRACT

Interferon (IFN)-stimulated gene 15 (ISG15), a ubiquitin-like protein, is covalently conjugated to host immune proteins such as MDA5 and IRF3 in a process called ISGylation, thereby promoting type I IFN induction to limit the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, whether SARS-CoV-2 proteins can be directly targeted for ISGylation remains elusive. In this study, we identified the nucleocapsid (N) protein of SARS-CoV-2 as a major substrate of ISGylation catalyzed by the host E3 ligase HERC5; however, N ISGylation is readily removed through deISGylation by the papain-like protease (PLpro) activity of NSP3. Mass spectrometry analysis identified that the N protein undergoes ISGylation at four lysine residues (K266, K355, K387, and K388), and mutational analysis of these sites in the context of a SARS-CoV-2 replicon (N-4KR) abolished N ISGylation and alleviated ISGylation-mediated inhibition of viral RNA synthesis. Furthermore, our results indicated that HERC5 targets preferentially phosphorylated N protein for ISGylation to regulate its oligomeric assembly. These findings reveal a novel mechanism by which the host ISGylation machinery directly targets SARS-CoV-2 proteins to restrict viral replication and illuminate how an intricate interplay of host (HERC5) and viral (PLpro) enzymes coordinates viral protein ISGylation and thereby regulates virus replication.IMPORTANCEThe role of protein ISGylation in regulating host cellular processes has been studied extensively; however, how ISG15 conjugation influences the activity of viral proteins, particularly coronaviral proteins, is largely unknown. Our study uncovered that the nucleocapsid (N) protein of SARS-CoV-2 is ISGylated by the HERC5 ISGylation machinery and that this modification impedes the functional assembly of N into oligomers ultimately inhibiting viral RNA synthesis. This antiviral restriction mechanism is antagonized by the PLpro deISGylation activity of SARS-CoV-2 NSP3. This study deepens our understanding of SARS-CoV-2 protein regulation by posttranslational modifications and may open new avenues for designing antiviral strategies for COVID-19.


Subject(s)
Coronavirus Nucleocapsid Proteins , Coronavirus Papain-Like Proteases , Cytokines , RNA, Viral , SARS-CoV-2 , Ubiquitin-Protein Ligases , Ubiquitins , Virus Replication , Humans , Ubiquitins/metabolism , Ubiquitins/genetics , SARS-CoV-2/metabolism , Ubiquitin-Protein Ligases/metabolism , Coronavirus Papain-Like Proteases/metabolism , RNA, Viral/metabolism , RNA, Viral/genetics , Cytokines/metabolism , HEK293 Cells , Coronavirus Nucleocapsid Proteins/metabolism , Phosphoproteins/metabolism , COVID-19/virology , COVID-19/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Intracellular Signaling Peptides and Proteins
4.
J Virol ; 98(9): e0085524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39120134

ABSTRACT

A severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes mild-to-severe respiratory symptoms, including acute respiratory distress. Despite remarkable efforts to investigate the virological and pathological impacts of SARS-CoV-2, many of the characteristics of SARS-CoV-2 infection still remain unknown. The interferon-inducible ubiquitin-like protein ISG15 is covalently conjugated to several viral proteins to suppress their functions. It was reported that SARS-CoV-2 utilizes its papain-like protease (PLpro) to impede ISG15 conjugation, ISGylation. However, the role of ISGylation in SARS-CoV-2 infection remains unclear. We aimed to elucidate the role of ISGylation in SARS-CoV-2 replication. We observed that the SARS-CoV-2 nucleocapsid protein is a target protein for the HERC5 E3 ligase-mediated ISGylation in cultured cells. Site-directed mutagenesis reveals that the residue K374 within the C-terminal spacer B-N3 (SB/N3) domain is required for nucleocapsid-ISGylation, alongside conserved lysine residue in MERS-CoV (K372) and SARS-CoV (K375). We also observed that the nucleocapsid-ISGylation results in the disruption of nucleocapsid oligomerization, thereby inhibiting viral replication. Knockdown of ISG15 mRNA enhanced SARS-CoV-2 replication in the SARS-CoV-2 reporter replicon cells, while exogenous expression of ISGylation components partially hampered SARS-CoV-2 replication. Taken together, these results suggest that SARS-CoV-2 PLpro inhibits ISGylation of the nucleocapsid protein to promote viral replication by evading ISGylation-mediated disruption of the nucleocapsid oligomerization.IMPORTANCEISG15 is an interferon-inducible ubiquitin-like protein that is covalently conjugated to the viral protein via specific Lys residues and suppresses viral functions and viral propagation in many viruses. However, the role of ISGylation in SARS-CoV-2 infection remains largely unclear. Here, we demonstrated that the SARS-CoV-2 nucleocapsid protein is a target protein for the HERC5 E3 ligase-mediated ISGylation. We also found that the residue K374 within the C-terminal spacer B-N3 (SB/N3) domain is required for nucleocapsid-ISGylation. We obtained evidence suggesting that nucleocapsid-ISGylation results in the disruption of nucleocapsid-oligomerization, thereby suppressing SARS-CoV-2 replication. We discovered that SARS-CoV-2 papain-like protease inhibits ISG15 conjugation of nucleocapsid protein via its de-conjugating enzyme activity. The present study may contribute to gaining new insight into the roles of ISGylation-mediated anti-viral function in SARS-CoV-2 infection and may lead to the development of more potent and selective inhibitors targeted to SARS-CoV-2 nucleocapsid protein.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Coronavirus Papain-Like Proteases , Cytokines , SARS-CoV-2 , Ubiquitin-Protein Ligases , Ubiquitins , Virus Replication , Humans , Ubiquitins/metabolism , Ubiquitins/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Cytokines/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/genetics , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Coronavirus Papain-Like Proteases/metabolism , HEK293 Cells , Phosphoproteins/metabolism , Phosphoproteins/genetics , Immune Evasion , Nucleocapsid Proteins/metabolism , Coronavirus 3C Proteases/metabolism , Intracellular Signaling Peptides and Proteins
5.
Mol Brain ; 17(1): 57, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148092

ABSTRACT

Discovery of novel post-translational modifications provides new insights into changes in protein function, localization, and stability. They are also key elements in understanding disease mechanisms and developing therapeutic strategies. We have previously reported that ubiquitin-like 3 (UBL3) serves as a novel post-translational modifier that is highly expressed in the cerebral cortex and hippocampus, in addition to various other organs, and that 60% of proteins contained in small extracellular vesicles (sEVs), including exosomes, are influenced by UBL3. In this study, we generated transgenic mice expressing biotinylated UBL3 in the forebrain under control of the alpha-CaMKII promoter (Ubl3Tg/+). Western blot analysis revealed that the expression of UBL3 in the cerebral cortex and hippocampus was 6- to 7-fold higher than that in the cerebellum. Therefore, we performed immunoprecipitation of protein extracts from the cerebral cortex of Ubl3+/+ and Ubl3Tg/+ mice using avidin beads to comprehensively discover UBL3 interacting proteins, identifying 35 new UBL3 interacting proteins. Nine proteins were annotated as extracellular exosomes. Gene Ontology (GO) analysis suggested a new relationship between sEVs and RNA metabolism in neurodegenerative diseases. We confirmed the association of endogenous UBL3 with the RNA-binding proteins FUS and HPRT1-both listed in the Neurodegenerative Diseases Variation Database (NDDVD)-and with LYPLA1, which is involved in Huntington's disease, using immunoprecipitation (IP)-western blotting analysis. These UBL3 interacting proteins will accelerate the continued elucidation of sEV research about proteins regulated by novel post-translational modifications by UBL3 in the brain.


Subject(s)
Brain , Ubiquitins , Animals , Mice , Brain/metabolism , Cerebral Cortex/metabolism , Exosomes/metabolism , Gene Ontology , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Ubiquitins/metabolism
6.
Theranostics ; 14(11): 4536-4553, 2024.
Article in English | MEDLINE | ID: mdl-39113797

ABSTRACT

Rationale: Acute kidney injury (AKI) has substantial rates of mortality and morbidity, coupled with an absence of efficacious treatment options. AKI commonly transits into chronic kidney disease (CKD) and ultimately culminates in end-stage renal failure. The interferon-stimulated gene 15 (ISG15) level was upregulated in the kidneys of mice injured by ischemia-reperfusion injury (IRI), cisplatin, or unilateral ureteral obstruction (UUO), however, its role in AKI development and subsequent AKI-to-CKD transition remains unknown. Methods: Isg15 knockout (Isg15 KO) mice challenged with bilateral or unilateral IRI, cisplatin, or UUO were used to investigate its role in AKI. We established cellular models with overexpression or knockout of ISG15 and subjected them to hypoxia-reoxygenation, cisplatin, or transforming growth factor- ß1 (TGF-ß1) stimulation. Renal RNA-seq data obtained from AKI models sourced from public databases and our studies, were utilized to examine the expression profiles of ISG15 and its associated genes. Additionally, published single cell RNA-seq data from human kidney allograft biopsies and mouse IRI model were analyzed to investigate the expression patterns of ISG15 and the type I TGF-ß receptor (TGFßR1). Western blotting, qPCR, co-immunoprecipitation, and immunohistochemical staining assays were performed to validate our findings. Results: Alleviated pathological injury and renal function were observed in Isg15 KO mice with IRI-, cisplatin-, or UUO-induced AKI and the following AKI-to-CKD transition. In hypoxia-reoxygenation, cisplatin or TGF-ß1 treated HK-2 cells, knockout ISG15 reduced stimulus-induced cell fibrosis, while overexpression of ISG15 with modification capacity exacerbated cell fibrosis. Immunoprecipitation assays demonstrated that ISG15 promoted ISGylation of TGFßR1, and inhibited its ubiquitination. Moreover, knockout of TGFßR1 blocked ISG15's fibrosis-exacerbating effect in HK-2 cells, while overexpression of TGFßR1 abolished the renal protective effect of ISG15 knockout during IRI-induced kidney injury. Conclusions: ISG15 plays an important role in the development of AKI and subsequent AKI-to-CKD transition by promoting TGFßR1 ISGylation.


Subject(s)
Acute Kidney Injury , Cisplatin , Cytokines , Mice, Knockout , Reperfusion Injury , Ubiquitins , Animals , Humans , Male , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Cisplatin/pharmacology , Cytokines/metabolism , Disease Models, Animal , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Ubiquitins/metabolism , Ubiquitins/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/genetics
7.
Cell Mol Life Sci ; 81(1): 292, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38976080

ABSTRACT

Cisplatin resistance is a major challenge for systemic therapy against advanced bladder cancer (BC). Little information is available on the regulation of cisplatin resistance and the underlying mechanisms require elucidation. Here, we detected that downregulation of the tumor suppressor, PPP2R2B (a serine/threonine protein phosphatase 2 A regulatory subunit), in BC promoted cell proliferation and migration. What's more, low PPP2R2B expression was correlated with cisplatin resistance. In vitro and in vivo experiments verified that PPP2R2B could promote BC sensitivity to cisplatin. In terms of mechanism, we identified a novel function of PPP2R2B as a nucleocytoplasmic transport molecule. PPP2R2B promoted ISG15 entry into the nucleus by mediating binding of IPO5 with ISG15. Nuclear translocation of ISG15 inhibited DNA repair, further increasing ISG15 expression through activation of the STING pathway. Besides, PPP2R2B was down-regulated by SUV39H1-mediated histone 3 lysine 9 trimethylation, which could be restored by the SUV39H1-specific inhibitor, chaetocin. Our data suggest that PPP2R2B expression level is a potential biomarker for chemotherapy response and that chemotherapy in combination with chaetocin may be a feasible treatment strategy for patients with BC.


Subject(s)
Cisplatin , Cytokines , Drug Resistance, Neoplasm , Protein Phosphatase 2 , Ubiquitins , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Humans , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Ubiquitins/metabolism , Ubiquitins/genetics , Cytokines/metabolism , Animals , Cell Line, Tumor , Mice , Cell Proliferation/drug effects , Mice, Nude , Cell Nucleus/metabolism , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C , Gene Expression Regulation, Neoplastic/drug effects , Cell Movement/drug effects , Female , Nerve Tissue Proteins
8.
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
9.
Acta Trop ; 257: 107283, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955322

ABSTRACT

Toxoplasmosis, a zoonotic parasitic disease caused by Toxoplasma gondii (T. gondii), is prevalent worldwide. The fact should be emphasized that a considerable proportion of individuals infected with T. gondii may remain asymptomatic; nevertheless, the condition can have severe implications for pregnant women or immunocompromised individuals. The current treatment of toxoplasmosis primarily relies on medication; however, traditional anti-toxoplasmosis drugs exhibit significant limitations in terms of efficacy, side effects, and drug resistance. The life cycles of T. gondii are characterized by distinct stages and its body morphology goes through dynamic alterations during the growth cycle that are intricately governed by a wide array of post-translational modifications (PTMs). Ubiquitin (Ub) signaling and ubiquitin-like (Ubl) signaling are two crucial post-translational modification pathways within cells, regulating protein function, localization, stability, or interactions by attaching Ub or ubiquitin-like proteins (Ubls) to target proteins. While these signaling mechanisms share some functional similarities, they have distinct regulatory mechanisms and effects. T. gondii possesses both Ub and Ubls and plays a significant role in regulating the parasite's life cycle and maintaining its morphology through PTMs of substrate proteins. Investigating the role and mechanism of protein ubiquitination in T. gondii will provide valuable insights for preventing and treating toxoplasmosis. This review explores the distinctive characteristics of Ub and Ubl signaling in T. gondii, with the aim of inspiring research ideas for the identification of safer and more effective drug targets against toxoplasmosis.


Subject(s)
Signal Transduction , Toxoplasma , Toxoplasmosis , Ubiquitin , Toxoplasma/metabolism , Toxoplasma/physiology , Toxoplasma/drug effects , Ubiquitin/metabolism , Humans , Toxoplasmosis/parasitology , Toxoplasmosis/drug therapy , Toxoplasmosis/metabolism , Animals , Protozoan Proteins/metabolism , Ubiquitination , Protein Processing, Post-Translational , Ubiquitins/metabolism , Life Cycle Stages
10.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38977311

ABSTRACT

The ubiquitin-like modifier FAT10 is upregulated under pro-inflammatory conditions, targets its substrates for proteasomal degradation and functions as a negative regulator of the type-I IFN response. Influenza A virus infection upregulates the production of type-I IFN and the expression of the E3 ligase TRIM21, which regulates type-I IFN production in a positive feedback manner. In this study, we show that FAT10 becomes covalently conjugated to TRIM21 and that this targets TRIM21 for proteasomal degradation. We further show that the coiled-coil and PRYSPRY domains of TRIM21 and the C-terminal diglycine motif of FAT10 are important for the TRIM21-FAT10 interaction. Moreover, upon influenza A virus infection and in the presence of FAT10 the total ubiquitination of TRIM21 is reduced and our data reveal that the FAT10-mediated degradation of TRIM21 diminishes IFNß production. Overall, this study provides strong evidence that FAT10 down-regulates the antiviral type-I IFN production by modulating additional molecules of the RIG-I signaling pathway besides the already published OTUB1. In addition, we elucidate a novel mechanism of FAT10-mediated proteasomal degradation of TRIM21 that regulates its stability.


Subject(s)
Interferon Type I , Proteasome Endopeptidase Complex , Ribonucleoproteins , Ubiquitination , Ubiquitins , Humans , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Interferon Type I/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , Proteasome Endopeptidase Complex/metabolism , Down-Regulation , HEK293 Cells , Signal Transduction , Influenza A virus/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Proteolysis , Animals
11.
mBio ; 15(8): e0053424, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38975783

ABSTRACT

Ubiquitin-like proteins (Ubls) in eukaryotes and bacteria mediate sulfur transfer for the biosynthesis of sulfur-containing biomolecules and form conjugates with specific protein targets to regulate their functions. Here, we investigated the functions and physiological importance of Ubls in a hyperthermophilic archaeon by constructing a series of deletion mutants. We found that the Ubls (TK1065, TK1093, and TK2118) in Thermococcus kodakarensis are conjugated to their specific target proteins, and all three are involved in varying degrees in the biosynthesis of sulfur-containing biomolecules such as tungsten cofactor (Wco) and tRNA thiouridines. TK2118 (named UblB) is involved in the biosynthesis of Wco in a glyceraldehyde 3-phosphate:ferredoxin oxidoreductase, which is required for glycolytic growth, whereas TK1093 (named UblA) plays a key role in the efficient thiolation of tRNAs, which contributes to cellular thermotolerance. Intriguingly, in the presence of elemental sulfur (S0) in the culture medium, defective synthesis of these sulfur-containing molecules in Ubl mutants was restored, indicating that T. kodakarensis can use S0 as an alternative sulfur source without Ubls. Our analysis indicates that the Ubl-mediated sulfur-transfer system in T. kodakarensis is important for efficient sulfur assimilation, especially under low S0 conditions, which may allow this organism to survive in a low sulfur environment.IMPORTANCESulfur is a crucial element in living organisms, occurring in various sulfur-containing biomolecules including iron-sulfur clusters, vitamins, and RNA thionucleosides, as well as the amino acids cysteine and methionine. In archaea, the biosynthesis routes and sulfur donors of sulfur-containing biomolecules are largely unknown. Here, we explored the functions of Ubls in the deep-blanched hyperthermophilic archaeon, Thermococcus kodakarensis. We demonstrated functional redundancy of these proteins in the biosynthesis of tungsten cofactor and tRNA thiouridines and the significance of these sulfur-carrier functions, especially in low sulfur environments. We propose that acquisition of a Ubl sulfur-transfer system, in addition to an ancient inorganic sulfur assimilation pathway, enabled the primordial archaeon to advance into lower-sulfur environments and expand their habitable zone.


Subject(s)
Archaeal Proteins , Sulfur , Thermococcus , Thermococcus/genetics , Thermococcus/metabolism , Sulfur/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , RNA, Transfer/metabolism , RNA, Transfer/genetics
12.
Cell Death Dis ; 15(7): 502, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003255

ABSTRACT

Dysfunction of the ubiquitin-proteasome system (UPS) is involved in the pathogenesis of various malignancies including colorectal cancer (CRC). Ubiquitin domain containing 1 (UBTD1), a ubiquitin-like protein, regulates UPS-mediated protein degradation and tumor progression in some cancer types. However, the biological function and mechanism of UBTD1 are far from being well elucidated, and its role in CRC has not been explored yet. In our study, we analyzed CRC patients' clinical information and UBTD1 expression data, and found that the expression of UBTD1 in cancer tissue was significantly higher than that in adjacent normal tissue. Higher UBTD1 expression was significantly associated with poorer survival and more lymph node metastasis. Overexpression of UBTD1 could facilitate, while knockdown could inhibit CRC cell proliferation and migration, respectively. RNA-seq and proteomics indicated that c-Myc is an important downstream target of UBTD1. Metabolomics showed the products of the glycolysis pathway were significantly increased in UBTD1 overexpression cells. In vitro, we verified UBTD1 upregulating c-Myc protein and promoting CRC cell proliferation and migration via regulating c-Myc. UBTD1 promoted CRC cells' glycolysis, evidenced by the increased lactate production and glucose uptake following UBTD1 overexpression. Mechanistically, UBTD1 prolonged the half-life of the c-Myc protein by binding to E3 ligase ß-transducin repeat-containing protein (ß-TrCP), thereby upregulated the expression of glycolysis rate-limiting enzyme hexokinase II (HK2), and enhanced glycolysis and promoted CRC progression. In conclusion, our study revealed that UBTD1 promotes CRC progression by upregulating glycolysis via the ß-TrCP/c-Myc/HK2 pathway, suggesting its potential as a prognostic biomarker and therapeutic target in CRC.


Subject(s)
Cell Proliferation , Colorectal Neoplasms , Disease Progression , Glycolysis , Proto-Oncogene Proteins c-myc , Up-Regulation , Animals , Female , Humans , Male , Mice , Middle Aged , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hexokinase/metabolism , Hexokinase/genetics , Mice, Nude , Protein Stability , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Ubiquitins/metabolism , Ubiquitins/genetics
13.
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
14.
Clin Exp Med ; 24(1): 140, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951255

ABSTRACT

Although renal cell carcinoma (RCC) is a prevalent type of cancer, the most common pathological subtype, clear cell renal cell carcinoma (ccRCC), still has poorly understood molecular mechanisms of progression. Moreover, interferon-stimulated gene 15 (ISG15) is associated with various types of cancer; however, its biological role in ccRCC remains unclear.This study aimed to explore the role of ISG15 in ccRCC progression.ISG15 expression was upregulated in ccRCC and associated with poor prognosis. RNA sequence analysis and subsequent experiments indicated that ISG15 modulated IL6/JAK2/STAT3 signaling to promote ccRCC proliferation, migration, and invasion. Additionally, our animal experiments confirmed that sustained ISG15 knockdown reduced tumor growth rate in nude mice and promoted cell apoptosis. ISG15 modulates the IL6/JAK2/STAT3 pathway, making it a potential therapeutic target and prognostic biomarker for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Cell Proliferation , Cytokines , Interleukin-6 , Janus Kinase 2 , Kidney Neoplasms , Mice, Nude , STAT3 Transcription Factor , Signal Transduction , Ubiquitins , Humans , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/genetics , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Janus Kinase 2/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Cytokines/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/genetics , Mice , Cell Line, Tumor , Male , Cell Movement , Female , Apoptosis , Gene Expression Regulation, Neoplastic , Prognosis , Disease Progression
15.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000460

ABSTRACT

Aberrant aggregation of misfolded alpha-synuclein (α-syn), a major pathological hallmark of related neurodegenerative diseases such as Parkinson's disease (PD), can translocate between cells. Ubiquitin-like 3 (UBL3) is a membrane-anchored ubiquitin-fold protein and post-translational modifier. UBL3 promotes protein sorting into small extracellular vesicles (sEVs) and thereby mediates intercellular communication. Our recent studies have shown that α-syn interacts with UBL3 and that this interaction is downregulated after silencing microsomal glutathione S-transferase 3 (MGST3). However, how MGST3 regulates the interaction of α-syn and UBL3 remains unclear. In the present study, we further explored this by overexpressing MGST3. In the split Gaussia luciferase complementation assay, we found that the interaction between α-syn and UBL3 was upregulated by MGST3. While Western blot and RT-qPCR analyses showed that silencing or overexpression of MGST3 did not significantly alter the expression of α-syn and UBL3, the immunocytochemical staining analysis indicated that MGST3 increased the co-localization of α-syn and UBL3. We suggested roles for the anti-oxidative stress function of MGST3 and found that the effect of MGST3 overexpression on the interaction between α-syn with UBL3 was significantly rescued under excess oxidative stress and promoted intracellular α-syn to extracellular transport. In conclusion, our results demonstrate that MGST3 upregulates the interaction between α-syn with UBL3 and promotes the interaction to translocate intracellular α-syn to the extracellular. Overall, our findings provide new insights and ideas for promoting the modulation of UBL3 as a therapeutic agent for the treatment of synucleinopathy-associated neurodegenerative diseases.


Subject(s)
Glutathione Transferase , Oxidative Stress , Ubiquitins , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Humans , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Ubiquitins/metabolism , Ubiquitins/genetics , Up-Regulation , Protein Transport , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Binding
16.
Medicine (Baltimore) ; 103(26): e38737, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941371

ABSTRACT

Alterations in signaling pathways and modulation of cell metabolism are associated with the pathogenesis of cancers, including hepatocellular carcinoma (HCC). Small ubiquitin-like modifier (SUMO) proteins and NF-κB family play major roles in various cellular processes. The current study aims to determine the expression profile of SUMO and NF-κB genes in HCC tumors and investigate their association with the clinical outcome of HCC. The expression of 5 genes - SUMO1, SUMO2, SUMO3, NF-κB p65, and NF-κB p50 - was quantified in tumor and adjacent non-tumor tissues of 58 HBV-related HCC patients by real-time quantitative PCR and was analyzed for the possible association with clinical parameters of HCC. The expression of SUMO2 was significantly higher in HCC tumor tissues compared to the adjacent non-tumor tissues (P = .01), while no significant difference in SUMO1, SUMO3, NF-κB p65, and NF-κB p50 expression was observed between HCC tumor and non-tumor tissues (P > .05). In HCC tissues, a strong correlation was observed between the expression of SUMO2 and NF-κB p50, between SUMO3 and NF-κB p50, between SUMO3 and NF-κB p65 (Spearman rho = 0.83; 0.82; 0.772 respectively; P < .001). The expression of SUMO1, SUMO2, SUMO3, NF-κB p65, and NF-κB p50 was decreased in grade 3 compared to grades 1 and 2 in HCC tumors according to the World Health Organization grades system. Our results highlighted that the SUMO2 gene is upregulated in tumor tissues of patients with HCC, and is related to the development of HCC, thus it may be associated with the pathogenesis of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Small Ubiquitin-Related Modifier Proteins , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/virology , Liver Neoplasms/metabolism , Male , Female , Middle Aged , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , NF-kappa B/metabolism , Adult , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Hepatitis B virus/genetics , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Aged , Gene Expression Regulation, Neoplastic , Ubiquitins/genetics , Ubiquitins/metabolism , Hepatitis B/complications , Hepatitis B/genetics
17.
Cell ; 187(17): 4656-4673.e28, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-38942013

ABSTRACT

The ability of proteins and RNA to coalesce into phase-separated assemblies, such as the nucleolus and stress granules, is a basic principle in organizing membraneless cellular compartments. While the constituents of biomolecular condensates are generally well documented, the mechanisms underlying their formation under stress are only partially understood. Here, we show in yeast that covalent modification with the ubiquitin-like modifier Urm1 promotes the phase separation of a wide range of proteins. We find that the drop in cellular pH induced by stress triggers Urm1 self-association and its interaction with both target proteins and the Urm1-conjugating enzyme Uba4. Urmylation of stress-sensitive proteins promotes their deposition into stress granules and nuclear condensates. Yeast cells lacking Urm1 exhibit condensate defects that manifest in reduced stress resilience. We propose that Urm1 acts as a reversible molecular "adhesive" to drive protective phase separation of functionally critical proteins under cellular stress.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Stress, Physiological , Ubiquitins , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitins/metabolism , Biomolecular Condensates/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Hydrogen-Ion Concentration , Stress Granules/metabolism
18.
Cells ; 13(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891017

ABSTRACT

Telomeres, potential biomarkers of aging, are known to shorten with continued cigarette smoke exposure. In order to further investigate this process and its impact on cellular stress and inflammation, we used an in vitro model with cigarette smoke extract (CSE) and observed the downregulation of telomere stabilizing TRF2 and POT1 genes after CSE treatment. hTERT is a subunit of telomerase and a well-known oncogenic marker, which is overexpressed in over 85% of cancers and may contribute to lung cancer development in smokers. We also observed an increase in hTERT and ISG15 expression levels after CSE treatment, as well as increased protein levels revealed by immunohistochemical staining in smokers' lung tissue samples compared to non-smokers. The effects of ISG15 overexpression were further studied by quantifying IFN-γ, an inflammatory protein induced by ISG15, which showed greater upregulation in smokers compared to non-smokers. Similar changes in gene expression patterns for TRF2, POT1, hTERT, and ISG15 were observed in blood and buccal swab samples from smokers compared to non-smokers. The results from this study provide insight into the mechanisms behind smoking causing telomere shortening and how this may contribute to the induction of inflammation and/or tumorigenesis, which may lead to comorbidities in smokers.


Subject(s)
Aging , Cytokines , Inflammation , Shelterin Complex , Smoking , Telomerase , Telomere , Telomeric Repeat Binding Protein 2 , Humans , Inflammation/genetics , Inflammation/pathology , Aging/genetics , Telomeric Repeat Binding Protein 2/metabolism , Telomeric Repeat Binding Protein 2/genetics , Cytokines/metabolism , Telomere/metabolism , Telomerase/metabolism , Telomerase/genetics , Smoking/adverse effects , Ubiquitins/metabolism , Ubiquitins/genetics , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Interferon-gamma/metabolism , Telomere Homeostasis , Male , Telomere Shortening , Female , Middle Aged
19.
JCI Insight ; 9(13)2024 May 23.
Article in English | MEDLINE | ID: mdl-38781019

ABSTRACT

Immunosuppression is a common feature of esophageal adenocarcinoma (EAC) and has been linked to poor overall survival (OS). We hypothesized that upstream factors might negatively influence CD3 levels and T cell activity, thus promoting immunosuppression and worse survival. We used clinical data and patient samples of those who progressed from Barrett's to dysplasia to EAC, investigated gene (RNA-Seq) and protein (tissue microarray) expression, and performed cell biology studies to delineate a pathway impacting CD3 protein stability that might influence EAC outcome. We showed that the loss of both CD3-ε expression and CD3+ T cell number correlated with worse OS in EAC. The gene related to anergy in lymphocytes isoform 1 (GRAIL1), which is the prominent isoform in EACs, degraded (ε, γ, δ) CD3s and inactivated T cells. In contrast, isoform 2 (GRAIL2), which is reduced in EACs, stabilized CD3s. Further, GRAIL1-mediated CD3 degradation was facilitated by interferon-stimulated gene 15 (ISG15), a ubiquitin-like protein. Consequently, the overexpression of a ligase-dead GRAIL1, ISG15 knockdown, or the overexpression of a conjugation-defective ISG15-leucine-arginine-glycine-glycine mutant could increase CD3 levels. Together, we identified an ISG15/GRAIL1/mutant p53 amplification loop negatively influencing CD3 levels and T cell activity, thus promoting immunosuppression in EAC.


Subject(s)
Adenocarcinoma , CD3 Complex , Cytokines , Esophageal Neoplasms , Ubiquitins , Humans , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/immunology , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/immunology , CD3 Complex/metabolism , CD3 Complex/genetics , Cytokines/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , Male , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Female , Gene Expression Regulation, Neoplastic , Barrett Esophagus/pathology , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Middle Aged
20.
PLoS Pathog ; 20(5): e1012230, 2024 May.
Article in English | MEDLINE | ID: mdl-38776321

ABSTRACT

While macrophage is one of the major type I interferon (IFN-I) producers in multiple tissues during viral infections, it also serves as an important target cell for many RNA viruses. However, the regulatory mechanism for the IFN-I response of macrophages to respond to a viral challenge is not fully understood. Here we report ADAP, an immune adaptor protein, is indispensable for the induction of the IFN-I response of macrophages to RNA virus infections via an inhibition of the conjugation of ubiquitin-like ISG15 (ISGylation) to RIG-I. Loss of ADAP increases RNA virus replication in macrophages, accompanied with a decrease in LPS-induced IFN-ß and ISG15 mRNA expression and an impairment in the RNA virus-induced phosphorylation of IRF3 and TBK1. Moreover, using Adap-/- mice, we show ADAP deficiency strongly increases the susceptibility of macrophages to RNA-virus infection in vivo. Mechanically, ADAP selectively interacts and functionally cooperates with RIG-I but not MDA5 in the activation of IFN-ß transcription. Loss of ADAP results in an enhancement of ISGylation of RIG-I, whereas overexpression of ADAP exhibits the opposite effect in vitro, indicating ADAP is detrimental to the RNA virus-induced ISGylation of RIG-I. Together, our data demonstrate a novel antagonistic activity of ADAP in the cell-intrinsic control of RIG-I ISGylation, which is indispensable for initiating and sustaining the IFN-I response of macrophages to RNA virus infections and replication.


Subject(s)
Adaptor Proteins, Signal Transducing , DEAD Box Protein 58 , Interferon Type I , Macrophages , Mice, Knockout , RNA Virus Infections , Ubiquitins , Animals , Macrophages/virology , Macrophages/metabolism , Macrophages/immunology , Mice , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , DEAD Box Protein 58/metabolism , Interferon Type I/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cytokines/metabolism , Mice, Inbred C57BL , Humans , Receptors, Immunologic/metabolism , Interferon-beta/metabolism , RNA Viruses/immunology , Interferon Regulatory Factor-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL