Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.839
Filter
1.
Front Immunol ; 15: 1436174, 2024.
Article in English | MEDLINE | ID: mdl-39315102

ABSTRACT

The ubiquitin-proteasome system (UPS) plays a crucial role in modulating the proliferation, activation, and normal functioning of immune cells through the regulation of protein degradation and function. By influencing the expression of immune checkpoint-associated proteins, the UPS modulates T cell-mediated anti-tumor immune responses and can potentially facilitate the immune escape of tumor cells. Additionally, the UPS contributes to the remodeling of the tumor immunosuppressive microenvironment (TIME) by regulating B cells, dendritic cells (DCs), macrophages, and Treg cells. Targeting the UPS in conjunction with immune checkpoint-associated proteins, and combining these with other therapeutic approaches, may significantly enhance the efficacy of combination therapies and pave the way for novel cancer treatment strategies. In this review, we first summarize the composition and alterations of the TIME, with a particular emphasis on the role of the UPS in TIME and its interactions with various immune cell types. Finally, we explore the potential of combining UPS-targeted therapies with immunotherapy to substantially improve the effectiveness of immunotherapy and enhance patient survival outcomes.


Subject(s)
Neoplasms , Proteasome Endopeptidase Complex , Tumor Microenvironment , Ubiquitin , Humans , Tumor Microenvironment/immunology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Ubiquitin/metabolism , Animals , Immunotherapy/methods , Combined Modality Therapy
2.
Redox Biol ; 76: 103349, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260061

ABSTRACT

Chemoresistance remains a principal culprit for the treatment failure in colorectal cancer (CRC), especially for patients with recurrent or metastatic disease. Deciphering the molecular basis of chemoresistance may lead to novel therapeutic strategies for this fatal disease. Here, UBR5, an E3 ubiquitin ligase frequently overexpressed in human CRC, is demonstrated to mediate chemoresistance principally by inhibiting ferroptosis. Paradoxically, UBR5 shields oxaliplatin-activated Smad3 from proteasome-dependent degradation via Lys 11-linked polyubiquitination. This novel chemical modification of Smad3 facilitates the transcriptional repression of ATF3, induction of SLC7A11 and inhibition of ferroptosis, contributing to chemoresistance. Consequently, targeting UBR5 in combination with a ferroptosis inducer synergistically sensitizes CRC to oxaliplatin-induced cell death and control of tumor growth. This study reveals, for the first time, a major clinically relevant chemoresistance mechanism in CRC mediated by UBR5 in sustaining TGFß-Smad3 signaling and tuning ferroptosis, unveiling its potential as a viable therapeutic target for chemosensitization.


Subject(s)
Amino Acid Transport System y+ , Colorectal Neoplasms , Drug Resistance, Neoplasm , Ferroptosis , Signal Transduction , Smad3 Protein , Ubiquitin-Protein Ligases , Ferroptosis/drug effects , Ferroptosis/genetics , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Smad3 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Drug Resistance, Neoplasm/genetics , Signal Transduction/drug effects , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Mice , Animals , Cell Line, Tumor , Ubiquitination , Oxaliplatin/pharmacology , Ubiquitin/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Lysine/metabolism
3.
Int J Mol Sci ; 25(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39337505

ABSTRACT

Glaucoma is chronic optic neuropathy whose pathogenesis has been associated with the altered metabolism of Trabecular Meshwork Cells, which is a cell type involved in the synthesis and remodeling of the trabecular meshwork, the main drainage pathway of the aqueous humor. Starting from previous findings supporting altered ubiquitin signaling, in this study, we investigated the ubiquitin-mediated turnover of myocilin (MYOC/TIGR gene), which is a glycoprotein with a recognized role in glaucoma pathogenesis, in a human Trabecular Meshwork strain cultivated in vitro in the presence of dexamethasone. This is a validated experimental model of steroid-induced glaucoma, and myocilin upregulation by glucocorticoids is a phenotypic marker of Trabecular Meshwork strains. Western blotting and native-gel electrophoresis first uncovered that, in the presence of dexamethasone, myocilin turnover by proteasome particles was slower than in the absence of the drug. Thereafter, co-immunoprecipitation, RT-PCR and gene-silencing studies identified STUB1/CHIP as a candidate E3-ligase of myocilin. In this regard, dexamethasone treatment was found to downregulate STUB1/CHIP levels by likely promoting its proteasome-mediated turnover. Hence, to strengthen the working hypothesis about global alterations of ubiquitin-signaling, the first profiling of TMCs ubiquitylome, in the presence and absence of dexamethasone, was here undertaken by diGLY proteomics. Application of this workflow effectively highlighted a robust dysregulation of key pathways (e.g., phospholipid signaling, ß-catenin, cell cycle regulation) in dexamethasone-treated Trabecular Meshwork Cells, providing an ubiquitin-centered perspective around the effect of glucocorticoids on metabolism and glaucoma pathogenesis.


Subject(s)
Cytoskeletal Proteins , Dexamethasone , Eye Proteins , Glycoproteins , Proteasome Endopeptidase Complex , Trabecular Meshwork , Ubiquitination , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/cytology , Humans , Dexamethasone/pharmacology , Glycoproteins/metabolism , Glycoproteins/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Cells, Cultured , Ubiquitin/metabolism , Glaucoma/metabolism , Glaucoma/pathology
4.
Cell Chem Biol ; 31(9): 1627-1635, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303699

ABSTRACT

The dynamic process of membrane shaping and remodeling plays a vital role in cellular functions, with proteins and cellular membranes interacting intricately to adapt to various cellular needs and environmental cues. Ubiquitination-a posttranslational modification-was shown to be essential in regulating membrane structure and shape. It influences virtually all pathways relying on cellular membranes, such as endocytosis and autophagy by directing protein degradation, sorting, and oligomerization. Ubiquitin is mostly known as a protein modifier; however, it was reported that ubiquitin and ubiquitin-like proteins can associate directly with lipids, affecting membrane curvature and dynamics. In this review, we summarize some of the current knowledge on ubiquitin-mediated membrane remodeling in the context of endocytosis, autophagy, and ER-phagy.


Subject(s)
Cell Membrane , Ubiquitin , Ubiquitination , Ubiquitin/metabolism , Humans , Cell Membrane/metabolism , Autophagy , Endocytosis , Animals , Endoplasmic Reticulum/metabolism
5.
Arthritis Res Ther ; 26(1): 171, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342401

ABSTRACT

OBJECTIVE: Based on the recent evidence of IL-1 inhibition in patients with rheumatoid arthritis (RA) and concomitant type 2 diabetes (T2D), we evaluated the synovial tissue expression of IL-1 related genes in relationship to the ubiquitin-proteasome system and the effects of insulin on ubiquitinated proteins in fibroblast-like synoviocytes (FLSs). METHODS: The synovial expression of IL-1 pathway genes was compared in early (< 1 year) treatment-naïve RA patients with T2D (RA/T2D n = 16) and age- and sex-matched RA patients without T2D (n = 16), enrolled in the Pathobiology of Early Arthritis Cohort (PEAC). The synovial expression of ubiquitin in macrophages and synovial lining fibroblasts was also assessed by Immunohistochemistry/immunofluorescence and correlated with synovial pathotypes. Finally, FLSs from RA patients (n = 5) were isolated and treated with human insulin (200 and 500 nM) and ubiquitinated proteins were assessed by western blot. RESULTS: Synovial tissues of RA/T2D patients were characterised by a consistent reduced expression of ubiquitin-proteasome genes. More specifically, ubiquitin genes (UBB, UBC, and UBA52) and genes codifying proteasome subunits (PSMA2, PSMA6, PSMA7, PSMB1, PSMB3, PSMB4, PSMB6, PSMB8, PSMB9, PSMB10, PSMC1, PSMD9, PSME1, and PSME2) were significantly lower in RA/T2D patients. On the contrary, genes regulating fibroblast functions (FGF7, FGF10, FRS2, FGFR3, and SOS1), and genes linked to IL-1 pathway hyper-activity (APP, IRAK2, and OSMR) were upregulated in RA/T2D. Immunohistochemistry showed a significant reduction of the percentage of ubiquitin-positive cells in synovial tissues of RA/T2D patients. Ubiquitin-positive cells were also increased in patients with a lympho-myeloid pathotype compared to diffuse myeloid or pauci-immune-fibroid. Finally, in vitro experiments showed a reduction of ubiquitinated proteins in RA-FLSs treated with a high concentration of insulin (500 nM). CONCLUSIONS: A different IL-1 pathway gene expression was observed in the synovial tissues of early treatment-naïve RA/T2D patients, linked to decreased expression of the ubiquitin-proteasome system. These findings may provide a mechanistic explanation of the observed clinical benefits of IL-1 inhibition in patients with RA and concomitant T2D.


Subject(s)
Arthritis, Rheumatoid , Diabetes Mellitus, Type 2 , Interleukin-1 , Proteasome Endopeptidase Complex , Synovial Membrane , Ubiquitin , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Diabetes Mellitus, Type 2/metabolism , Male , Female , Middle Aged , Ubiquitin/metabolism , Interleukin-1/metabolism , Interleukin-1/genetics , Synovial Membrane/metabolism , Signal Transduction/physiology , Aged , Cohort Studies , Synoviocytes/metabolism , Synoviocytes/drug effects , Blotting, Western , Adult , Immunohistochemistry , Cells, Cultured , Insulin/metabolism
6.
Sci Adv ; 10(38): eadj4122, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39303039

ABSTRACT

Sarcopenia is characterized by accelerated muscle mass and function loss, which burdens and challenges public health worldwide. Several studies indicated that selenium deficiency is associated with sarcopenia; however, the specific mechanism remains unclear. Here, we demonstrated that selenoprotein W (SELENOW) containing selenium in the form of selenocysteine functioned in sarcopenia. SELENOW expression is up-regulated in dexamethasone (DEX)-induced muscle atrophy and age-related sarcopenia mouse models. Knockout (KO) of SELENOW profoundly aggravated the process of muscle mass loss in the two mouse models. Mechanistically, SELENOW KO suppressed the RAC1-mTOR cascade by the interaction between SELENOW and RAC1 and induced the imbalance of protein synthesis and degradation. Consistently, overexpression of SELENOW in vivo and in vitro alleviated the muscle and myotube atrophy induced by DEX. SELENOW played a role in age-related sarcopenia and regulated the genes associated with aging. Together, our study uncovered the function of SELENOW in age-related sarcopenia and provides promising evidence for the prevention and treatment of sarcopenia.


Subject(s)
Mice, Knockout , Proteasome Endopeptidase Complex , Protein Biosynthesis , Sarcopenia , Selenoprotein W , Ubiquitin , Animals , Proteasome Endopeptidase Complex/metabolism , Mice , Sarcopenia/metabolism , Sarcopenia/genetics , Sarcopenia/pathology , Ubiquitin/metabolism , Selenoprotein W/genetics , Selenoprotein W/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Dexamethasone/pharmacology , TOR Serine-Threonine Kinases/metabolism , Disease Models, Animal , Muscular Atrophy/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/chemically induced , Aging/metabolism , Male , Signal Transduction , Neuropeptides
7.
Mol Cell ; 84(18): 3378-3380, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303679

ABSTRACT

In two recent studies in Nature, Hör et al.1 and Chambers et al.2 report that ubiquitin-like conjugation in bacteria antagonizes phage replication.


Subject(s)
Ubiquitination , Ubiquitin/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacteriophages/metabolism , Bacteriophages/physiology , Virus Replication , Bacteria/metabolism , Bacteria/genetics , Bacteria/virology
8.
Nat Commun ; 15(1): 7707, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300082

ABSTRACT

Mutations in parkin and PINK1 cause early-onset Parkinson's disease (EOPD). The ubiquitin ligase parkin is recruited to damaged mitochondria and activated by PINK1, a kinase that phosphorylates ubiquitin and the ubiquitin-like domain of parkin. Activated phospho-parkin then ubiquitinates mitochondrial proteins to target the damaged organelle for degradation. Here, we present the mechanism of activation of a new class of small molecule allosteric modulators that enhance parkin activity. The compounds act as molecular glues to enhance the ability of phospho-ubiquitin (pUb) to activate parkin. Ubiquitination assays and isothermal titration calorimetry with the most active compound (BIO-2007817) identify the mechanism of action. We present the crystal structure of a closely related compound (BIO-1975900) bound to a complex of parkin and two pUb molecules. The compound binds next to pUb on RING0 and contacts both proteins. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) experiments confirm that activation occurs through release of the catalytic Rcat domain. In organello and mitophagy assays demonstrate that BIO-2007817 partially rescues the activity of parkin EOPD mutants, R42P and V56E, offering a basis for the design of activators as therapeutics for Parkinson's disease.


Subject(s)
Parkinson Disease , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/chemistry , Humans , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinases/metabolism , Protein Kinases/genetics , Protein Kinases/chemistry , Crystallography, X-Ray , Mutation , Phosphorylation , Allosteric Regulation , Mitophagy/drug effects , Ubiquitin/metabolism , Models, Molecular , Protein Binding , HEK293 Cells
9.
Elife ; 132024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221915

ABSTRACT

Loss-of-function Parkin mutations lead to early-onset of Parkinson's disease. Parkin is an auto-inhibited ubiquitin E3 ligase activated by dual phosphorylation of its ubiquitin-like (Ubl) domain and ubiquitin by the PINK1 kinase. Herein, we demonstrate a competitive binding of the phospho-Ubl and RING2 domains towards the RING0 domain, which regulates Parkin activity. We show that phosphorylated Parkin can complex with native Parkin, leading to the activation of autoinhibited native Parkin in trans. Furthermore, we show that the activator element (ACT) of Parkin is required to maintain the enzyme kinetics, and the removal of ACT slows the enzyme catalysis. We also demonstrate that ACT can activate Parkin in trans but less efficiently than when present in the cis molecule. Furthermore, the crystal structure reveals a donor ubiquitin binding pocket in the linker connecting REP and RING2, which plays a crucial role in Parkin activity.


Subject(s)
Protein Binding , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/chemistry , Humans , Phosphorylation , Crystallography, X-Ray , Models, Molecular , Ubiquitin/metabolism , Kinetics
10.
Cell Death Dis ; 15(9): 660, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251572

ABSTRACT

The WD repeat-containing protein 4 (WDR4) has repeatedly been associated with primary microcephaly, a condition of impaired brain and skull growth. Often, faulty centrosomes cause microcephaly, yet aberrant cilia may also be involved. Here, we show using a combination of approaches in human fibroblasts, zebrafish embryos and patient-derived cells that WDR4 facilitates cilium formation. Molecularly, we associated WDR4 loss-of-function with increased protein synthesis and concomitant upregulation of proteasomal activity, while ubiquitin precursor pools are reduced. Inhibition of proteasomal activity as well as supplementation with free ubiquitin restored normal ciliogenesis. Proteasome inhibition ameliorated microcephaly phenotypes. Thus, we propose that WDR4 loss-of-function impairs head growth and neurogenesis via aberrant cilia formation, initially caused by disturbed protein and ubiquitin homeostasis.


Subject(s)
Cilia , Proteasome Endopeptidase Complex , Ubiquitin , Zebrafish , Proteasome Endopeptidase Complex/metabolism , Humans , Cilia/metabolism , Cilia/pathology , Animals , Ubiquitin/metabolism , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Fibroblasts/metabolism , Neurogenesis
11.
Sci Rep ; 14(1): 21195, 2024 09 11.
Article in English | MEDLINE | ID: mdl-39261509

ABSTRACT

It is estimated that there are 544.9 million people suffering from chronic respiratory diseases in the world, which is the third largest chronic disease. Although there are various clinical treatment methods, there is no specific drug for chronic pulmonary diseases, including chronic obstructive pulmonary disease (COPD), interstitial lung disease (ILD) and idiopathic pulmonary fibrosis (IPF). Therefore, it is urgent to clarify the pathological mechanism and medication development. Single-cell transcriptome data of human and mouse from GEO database were integrated by "Harmony" algorithm. The data was standardized and normalized by using "Seurat" package, and "SingleR" algorithm was used for cell grouping annotation. The "Findmarker" function is used to find differentially expressed genes (DEGs), which were enriched and analyzed by using "clusterProfiler", and a protein interaction network was constructed for DEGs, and four algorithms are used to find the hub genes. The expression of hub genes were analyzed in independent human and mouse single-cell transcriptome data. Bulk RNA data were used to integrate by the "SVA" function, verify the expression levels of hub genes and build a diagnostic model. The L1000FWD platform was used to screen potential drugs. Through exploring the similarities and differences by integrated single-cell atlas, we found that the lung parenchymal cells showed abnormal oxidative stress, cell matrix adhesion and ubiquitination in COPD, corona virus disease 2019 (COVID-19), ILD and IPF. Meanwhile, the lung resident immune cells showed abnormal Toll-like receptor signals, interferon signals and ubiquitination. However, unlike acute pneumonia (COVID-19), chronic pulmonary disease shows enhanced ubiquitination. This phenomenon was confirmed in independent external human single-cell atlas, but unfortunately, it was not confirmed in mouse single-cell atlas of bleomycin-induced pulmonary fibrosis model and influenza virus-infected mouse model, which means that the model needs to be optimized. In addition, the bulk RNA-Seq data of COVID-19, ILD and IPF was integrated, and we found that the immune infiltration of lung tissue was enhanced, consistent with the single-cell level, UBA52, UBB and UBC were low expressed in COVID-19 and high expressed in ILD, and had a strong correlation with the expression of cell matrix adhesion genes. UBA52 and UBB have good diagnostic efficacy, and salermide and SSR-69071 can be used as their candidate drugs. Our study found that the disorder of protein ubiquitination in chronic pulmonary diseases is an important cause of pathological phenotype of pulmonary fibrosis by integrating scRNA-Seq and bulk RNA-Seq, which provides a new horizons for clinicopathology, diagnosis and treatment.


Subject(s)
RNA-Seq , Ubiquitin , Humans , Animals , Mice , Ubiquitin/metabolism , Ubiquitin/genetics , Single-Cell Analysis/methods , Transcriptome , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Gene Expression Profiling , Protein Interaction Maps , Chronic Disease , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , SARS-CoV-2/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Single-Cell Gene Expression Analysis
12.
Proc Natl Acad Sci U S A ; 121(39): e2408697121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39264755

ABSTRACT

An N-degron is a degradation signal whose main determinant is a "destabilizing" N-terminal residue of a protein. Specific N-degrons, discovered in 1986, were the first identified degradation signals in short-lived intracellular proteins. These N-degrons are recognized by a ubiquitin-dependent proteolytic system called the Arg/N-degron pathway. Although bacteria lack the ubiquitin system, they also have N-degron pathways. Studies after 1986 have shown that all 20 amino acids of the genetic code can act, in specific sequence contexts, as destabilizing N-terminal residues. Eukaryotic proteins are targeted for the conditional or constitutive degradation by at least five N-degron systems that differ both functionally and mechanistically: the Arg/N-degron pathway, the Ac/N-degron pathway, the Pro/N-degron pathway, the fMet/N-degron pathway, and the newly named, in this perspective, GASTC/N-degron pathway (GASTC = Gly, Ala, Ser, Thr, Cys). I discuss these systems and the expanded terminology that now encompasses the entire gamut of known N-degron pathways.


Subject(s)
Proteolysis , Humans , Ubiquitin/metabolism , Proteins/metabolism , Proteins/genetics , Proteins/chemistry , Animals , Signal Transduction , Degrons
13.
Virology ; 599: 110199, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39116646

ABSTRACT

Hepatitis B virus is one of the leading causes behind the neoplastic transformation of liver tissue and associated mortality. Despite the availability of many therapies and vaccines, the pathogenic landscape of the virus remains elusive; urging the development of novel strategies based on the fundamental infectious and transformative modalities of the virus-host interactome. Ubiquitination is a widely observed post-translational modification of several proteins, which either regulates the proteins' turnover or impacts their functionalities. In recent years, ample amount of literature has accumulated regarding the ubiquitination dynamics of the HBV proteins as well as the host proteins during HBV infection and carcinogenesis; with direct and detailed characterization of the involvement of HBV in these processes. Interestingly, while many of these ubiquitination events restrict HBV life cycle and carcinogenesis, several others promote the emergence of hepatocarcinoma by putting the virus in an advantageous position. This review sums up the snowballing literature on ubiquitination-mediated regulation of the host-HBV crosstalk, with special emphasis on its influence on the establishment and progression of hepatocellular carcinoma on a molecular level. With the advent of cutting-edge ubiquitination-targeted therapeutic approaches, the findings emanating from this review may potentiate the identification of novel anti-HBV targets for the formulation of novel anticancer strategies to control the HBV-induced hepato-carcinogenic process on a global scale.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B virus , Host-Pathogen Interactions , Liver Neoplasms , Ubiquitin , Ubiquitination , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/metabolism , Humans , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Liver Neoplasms/virology , Liver Neoplasms/metabolism , Ubiquitin/metabolism , Hepatitis B/virology , Hepatitis B/complications , Animals
14.
Nucleic Acids Res ; 52(16): 9978-9995, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39149911

ABSTRACT

Monoubiquitination of histones H2B-K120 (H2BK120ub) and H2A-K119 (H2AK119ub) play opposing roles in regulating transcription and chromatin compaction. H2BK120ub is a hallmark of actively transcribed euchromatin, while H2AK119ub is highly enriched in transcriptionally repressed heterochromatin. Whereas H2BK120ub is known to stimulate the binding or activity of various chromatin-modifying enzymes, this post-translational modification (PTM) also interferes with the binding of several proteins to the nucleosome H2A/H2B acidic patch via an unknown mechanism. Here, we report cryoEM structures of an H2BK120ub nucleosome showing that ubiquitin adopts discrete positions that occlude the acidic patch. Molecular dynamics simulations show that ubiquitin remains stably positioned over this nucleosome region. By contrast, our cryoEM structures of H2AK119ub nucleosomes show ubiquitin adopting discrete positions that minimally occlude the acidic patch. Consistent with these observations, H2BK120ub, but not H2AK119ub, abrogates nucleosome interactions with acidic patch-binding proteins RCC1 and LANA, and single-domain antibodies specific to this region. Our results suggest a mechanism by which H2BK120ub serves as a gatekeeper to the acidic patch and point to distinct roles for histone H2AK119 and H2BK120 ubiquitination in regulating protein binding to nucleosomes.


Subject(s)
Cryoelectron Microscopy , Histones , Molecular Dynamics Simulation , Nucleosomes , Ubiquitin , Ubiquitination , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleosomes/chemistry , Histones/metabolism , Histones/chemistry , Ubiquitin/metabolism , Ubiquitin/chemistry , Ubiquitin/genetics , Humans , Protein Binding , Protein Processing, Post-Translational
15.
Nat Commun ; 15(1): 7479, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214970

ABSTRACT

The Legionella pneumophila Sde family of translocated proteins promotes host tubular endoplasmic reticulum (ER) rearrangements that are tightly linked to phosphoribosyl-ubiquitin (pR-Ub) modification of Reticulon 4 (Rtn4). Sde proteins have two additional activities of unclear relevance to the infection process: K63 linkage-specific deubiquitination and phosphoribosyl modification of polyubiquitin (pR-Ub). We show here that the deubiquitination activity (DUB) stimulates ER rearrangements while pR-Ub protects the replication vacuole from cytosolic surveillance by autophagy. Loss of DUB activity is tightly linked to lowered pR-Ub modification of Rtn4, consistent with the DUB activity fueling the production of pR-Ub-Rtn4. In parallel, phosphoribosyl modification of polyUb, in a region of the protein known as the isoleucine patch, prevents binding by the autophagy adapter p62. An inability of Sde mutants to modify polyUb results in immediate p62 association, a critical precursor to autophagic attack. The ability of Sde WT to block p62 association decays quickly after bacterial infection, as predicted by the presence of previously characterized L. pneumophila effectors that inactivate Sde and remove polyUb. In sum, these results show that the accessory Sde activities act to stimulate ER rearrangements and protect from host innate immune sensing in a temporal fashion.


Subject(s)
Autophagy , Bacterial Proteins , Endoplasmic Reticulum , Legionella pneumophila , Ubiquitin , Ubiquitination , Vacuoles , Legionella pneumophila/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , Ubiquitin/metabolism , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endoplasmic Reticulum/metabolism , Animals , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Polyubiquitin/metabolism , Mice , Membrane Proteins
16.
Sci Rep ; 14(1): 20159, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39215164

ABSTRACT

Capacitation is an essential post-testicular maturation event endowing spermatozoa with fertilizing capacity within the female reproductive tract, significant for fertility, reproductive health, and contraception. By using a human-relevant large animal model, the domestic boar, this study focuses on furthering our understanding of the involvement of the ubiquitin-proteasome system (UPS) in sperm capacitation. The UPS is a universal, evolutionarily conserved, cellular proteome-wide degradation and recycling machinery, that has been shown to play a significant role in reproduction during the past two decades. Herein, we have used a bottom-up proteomic approach to (i) monitor the capacitation-related changes in the sperm protein levels, and (ii) identify the targets of UPS regulation during sperm capacitation. Spermatozoa were capacitated under proteasomal activity-permissive and inhibiting conditions and extracted sperm proteins were subjected to high-resolution mass spectrometry. We report that 401 individual proteins differed at least two-fold in abundance (P < 0.05) after in vitro capacitation (IVC) and 13 proteins were found significantly different (P < 0.05) between capacitated spermatozoa with proteasomal inhibition compared to the vehicle control. These proteins were associated with biological processes including sperm capacitation, sperm motility, metabolism, binding to zona pellucida, and proteasome-mediated catabolism. Changes in RAB2A, CFAP161, and TTR during IVC were phenotyped by immunocytochemistry, image-based flow cytometry, and Western blotting. We conclude that (i) the sperm proteome is subjected to extensive remodeling during sperm capacitation, and (ii) the UPS has a narrow range of distinct protein substrates during capacitation. This knowledge highlights the importance of the UPS in sperm capacitation and offers opportunities to identify novel pharmacological targets to modulate sperm fertilizing ability for the benefit of human reproductive health, assisted reproductive therapy, and contraception, as well as reproductive management in food animal agriculture.


Subject(s)
Proteasome Endopeptidase Complex , Proteomics , Sperm Capacitation , Spermatozoa , Ubiquitin , Sperm Capacitation/physiology , Animals , Male , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Swine , Spermatozoa/metabolism , Spermatozoa/physiology , Proteomics/methods , Proteome/metabolism
17.
Expert Opin Ther Targets ; 28(8): 689-700, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39086205

ABSTRACT

INTRODUCTION: Psoriasis is a chronic immune-mediated disorder affecting over 2-3% of the population worldwide, significantly impacting quality of life. Despite the availability of various therapeutic interventions, concerns persist regarding lesion recurrence and potential alterations in immune surveillance promoting cancer progression. Recent advancements in understanding cellular and molecular pathways have unveiled key factors in psoriasis etiology, including IL-17, 22, 23, TNF-α, PDE-4, JAK-STAT inhibitors, and AhR agonists. This work explores the potential of S-phase kinase-associated protein 2 (Skp2) as a therapeutic target in psoriasis. AREA COVERED: This review covers the current understanding of psoriasis pathophysiology, including immune dysregulation, and the role of keratinocytes and ubiquitin. It also delves into Skp2 role in cell cycle regulation, and its correlation with angiogenesis and ubiquitin in psoriasis. The evolving therapeutic approaches targeting Skp2, including small molecule inhibitors, are also discussed. EXPERT OPINION: Targeting Skp2 holds promise for developing novel therapeutic approaches for psoriasis. By modulating Skp2 activity or expression, it may be possible to intervene in inflammatory and proliferative processes underlying the disease. Further research into Skp2 inhibitors and their efficacy in preclinical and clinical settings is warranted to harness the full potential of Skp2 as a therapeutic target in psoriasis management.


Subject(s)
Molecular Targeted Therapy , Psoriasis , S-Phase Kinase-Associated Proteins , Humans , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , S-Phase Kinase-Associated Proteins/metabolism , Psoriasis/drug therapy , Psoriasis/pathology , Animals , Quality of Life , Keratinocytes/drug effects , Keratinocytes/metabolism , Ubiquitin/metabolism , Drug Development
18.
Bioconjug Chem ; 35(9): 1441-1449, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39167708

ABSTRACT

Protein-protein interactions (PPIs) are some of the most challenging target classes in drug discovery. Highly sensitive detection techniques are required for the identification of chemical modulators of PPIs. Here, we introduce PPI confocal nanoscanning (PPI-CONA), a miniaturized, microbead based high-resolution fluorescence imaging assay. We demonstrate the capabilities of PPI-CONA by detecting low affinity ternary complex formation between the human CDC34A ubiquitin-conjugating (E2) enzyme, ubiquitin, and CC0651, a small molecule enhancer of the CDC34A-ubiquitin interaction. We further exemplify PPI-CONA with an E2 enzyme binding study on CC0651 and a CDC34A binding specificity study of a series of CC0651 analogues. Our results indicate that CC0651 is highly selective toward CDC34A. We further demonstrate how PPI-CONA can be applied to screening very low affinity interactions. PPI-CONA holds potential for high-throughput screening for modulators of PPI targets and characterization of their affinity, specificity, and selectivity.


Subject(s)
Protein Binding , Ubiquitin-Conjugating Enzymes , Ubiquitin , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Ubiquitin/chemistry , Anaphase-Promoting Complex-Cyclosome/metabolism , Microscopy, Confocal , Protein Interaction Mapping/methods
19.
Biomed Pharmacother ; 179: 117127, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191026

ABSTRACT

Neuropathic pain (NP) remains one of the world's most difficult problems, and people suffering from NP have their quality of life affected to a great extent and constantly suffer from pain. Sensitization of injurious receptors, ectopic firing of afferent nerves after nerve injury, and coupling between sympathetic and sensory neurons are involved in the onset or development of NP, but the pathogenesis of NP is still not well understood. We found that the ubiquitin system is involved in the pathogenesis of NP and has a crucial role in it. The ubiquitin system can be involved in the onset or reversal of NP by affecting ion channels, cellular signal transduction, glial cells, and the regulation of non-coding RNAs. This provides new ideas for the treatment of NP. The ubiquitin system may be a new effective target for the treatment of NP. A continued, in-depth understanding of the mechanisms of the ubiquitin system involved in NP could further refine the study of analgesic targets and improve pharmacological studies.


Subject(s)
Neuralgia , Ubiquitin , Neuralgia/metabolism , Neuralgia/drug therapy , Neuralgia/physiopathology , Humans , Ubiquitin/metabolism , Animals , Signal Transduction
20.
Nature ; 633(8028): 216-223, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39143218

ABSTRACT

Transthiolation (also known as transthioesterification) reactions are used in the biosynthesis of acetyl coenzyme A, fatty acids and polyketides, and for post-translational modification by ubiquitin (Ub) and ubiquitin-like (Ubl) proteins1-3. For the Ub pathway, E1 enzymes catalyse transthiolation from an E1~Ub thioester to an E2~Ub thioester. Transthiolation is also required for transfer of Ub from an E2~Ub thioester to HECT (homologous to E6AP C terminus) and RBR (ring-between-ring) E3 ligases to form E3~Ub thioesters4-6. How isoenergetic transfer of thioester bonds is driven forward by enzymes in the Ub pathway remains unclear. Here we isolate mimics of transient transthiolation intermediates for E1-Ub(T)-E2 and E2-Ub(T)-E3HECT complexes (where T denotes Ub in a thioester or Ub undergoing transthiolation) using a chemical strategy with native enzymes and near-native Ub to capture and visualize a continuum of structures determined by single-particle cryo-electron microscopy. These structures and accompanying biochemical experiments illuminate conformational changes in Ub, E1, E2 and E3 that are coordinated with the chemical reactions to facilitate directional transfer of Ub from each enzyme to the next.


Subject(s)
Protein Processing, Post-Translational , Schizosaccharomyces pombe Proteins , Sulfhydryl Compounds , Ubiquitin-Activating Enzymes , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitin , Cryoelectron Microscopy , Esterification , Models, Molecular , Protein Conformation , Schizosaccharomyces/enzymology , Schizosaccharomyces/ultrastructure , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/ultrastructure , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin/ultrastructure , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/ultrastructure , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/ultrastructure , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL