Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.206
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.
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
3.
Sci Rep ; 14(1): 20664, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237639

ABSTRACT

The recA gene, encoding Recombinase A (RecA) is one of three Mycobacterium tuberculosis (Mtb) genes encoding an in-frame intervening protein sequence (intein) that must splice out of precursor host protein to produce functional protein. Ongoing debate about whether inteins function solely as selfish genetic elements or benefit their host cells requires understanding of interplay between inteins and their hosts. We measured environmental effects on native RecA intein splicing within Mtb using a combination of western blots and promoter reporter assays. RecA splicing was stimulated in bacteria exposed to DNA damaging agents or by treatment with copper in hypoxic, but not normoxic, conditions. Spliced RecA was processed by the Mtb proteasome, while free intein was degraded efficiently by other unknown mechanisms. Unspliced precursor protein was not observed within Mtb despite its accumulation during ectopic expression of Mtb recA within E. coli. Surprisingly, Mtb produced free N-extein in some conditions, and ectopic expression of Mtb N-extein activated LexA in E. coli. These results demonstrate that the bacterial environment greatly impacts RecA splicing in Mtb, underscoring the importance of studying intein splicing in native host environments and raising the exciting possibility of intein splicing as a novel regulatory mechanism in Mtb.


Subject(s)
Bacterial Proteins , Escherichia coli , Inteins , Mycobacterium tuberculosis , Protein Splicing , Rec A Recombinases , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Rec A Recombinases/metabolism , Rec A Recombinases/genetics , Inteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Exteins/genetics , DNA Damage , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Serine Endopeptidases
4.
Nat Commun ; 15(1): 8184, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294158

ABSTRACT

New proteasomes are produced to accommodate increases in cellular catabolic demand and prevent the accumulation of cytotoxic proteins. Formation of the proteasomal 20S core complex relies on the function of the five chaperones PAC1-4 and POMP. Here, to understand how these chaperones facilitate proteasome assembly, we tagged the endogenous chaperones using CRISPR/Cas gene editing and examined the chaperone-bound complexes by cryo-EM. We observe an early α-ring intermediate subcomplex that is stabilized by PAC1-4, which transitions to ß-ring assembly upon dissociation of PAC3/PAC4 and rearrangement of the PAC1 N-terminal tail. Completion of the ß-ring and dimerization of half-proteasomes repositions critical lysine K33 to trigger cleavage of the ß pro-peptides, leading to the concerted dissociation of POMP and PAC1/PAC2 to yield mature 20S proteasomes. This study reveals structural insights into critical points along the assembly pathway of the human proteasome and provides a molecular blueprint for 20S biogenesis.


Subject(s)
Cryoelectron Microscopy , Molecular Chaperones , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Humans , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Models, Molecular , HEK293 Cells , CRISPR-Cas Systems
5.
Bioorg Med Chem Lett ; 112: 129945, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39222889

ABSTRACT

Regulatory T (Treg) cells play a central role in immune homeostasis. Forkhead box P3 (Foxp3), a hallmark molecule in Treg cells, is a vital transcription factor for their development and function. Studies have shown that degradation of the Foxp3 could provide therapeutic benefits in achieving effective anti-tumor immunity. In this study, we designed three PROTAC molecules, P60-L1-VHL, P60-L2-VHL, and P60-L3-VHL, based on a 15-mer peptide inhibitor of Foxp3 (P60), and explored their potential in regulating Foxp3 expression and function. Our data show that, among these molecules, P60-L3-VHL can inhibit the expression and nuclear localization of Foxp3 in HEK 293 T and HeLa cells, respectively. Meanwhile, use of proteasome inhibitor in P60-L3-VHL treated cells revealed an increased Foxp3 expression, indicating that P60-L3-VHL mediates the inhibition of Foxp3 through its degradation in the proteasome pathway. We further substantiate that P60-L3-VHL reduces the differentiation and Foxp3 expression in the in-vitro activated Treg cells. Overall, our findings suggest that P60-L3-VHL inhibits the differentiation of Treg cells by degrading the Foxp3, which may have potential implications in cancer immunotherapy.


Subject(s)
Forkhead Transcription Factors , Proteolysis , Humans , Forkhead Transcription Factors/metabolism , Proteolysis/drug effects , HEK293 Cells , HeLa Cells , T-Lymphocytes, Regulatory/drug effects , Structure-Activity Relationship , Molecular Structure , Drug Discovery , Dose-Response Relationship, Drug , Proteasome Endopeptidase Complex/metabolism , Proteolysis Targeting Chimera
6.
Cells ; 13(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273066

ABSTRACT

Many cellular processes are regulated by proteasome-mediated protein degradation, including regulation of signaling pathways and gene expression. Among the pathways regulated by the ubiquitin-proteasome system is the Hedgehog pathway and its downstream effectors, the Gli transcription factors. Here we provide evidence that proteasomal activity is necessary for maintaining the activation of the Hedgehog pathway, and this crucial event takes place at the level of Gli proteins. We undertook extensive work to demonstrate the specificity of the observed phenomenon by ruling out the involvement of primary cilium, impaired nuclear import, failed dissociation from Sufu, microtubule stabilization, and stabilization of Gli repressor forms. Moreover, we showed that proteasomal-inhibition-mediated Hedgehog pathway downregulation is not restricted to the NIH-3T3 cell line. We demonstrated, using CRISPR/Ca9 mutagenesis, that neither Gli1, Gli2, nor Gli3 are solely responsible for the Hedgehog pathway downregulation upon proteasome inhibitor treatment, and that Cul3 KO renders the same phenotype. Finally, we report two novel E3 ubiquitin ligases, Btbd9 and Kctd3, known Cul3 interactors, as positive Hedgehog pathway regulators. Our data pave the way for a better understanding of the regulation of gene expression and the Hedgehog signaling pathway.


Subject(s)
Cullin Proteins , Hedgehog Proteins , Proteasome Endopeptidase Complex , Signal Transduction , Ubiquitination , Animals , Proteasome Endopeptidase Complex/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , NIH 3T3 Cells , Cullin Proteins/metabolism , Cullin Proteins/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Humans , Gene Expression Regulation
7.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273140

ABSTRACT

(1): Atopic dermatitis and psoriasis vulgaris are chronic, inflammatory diseases. Clinical presentation usually leads to a proper diagnosis, but sometimes neither clinical examination nor histopathological evaluation can be conclusive. Therefore, we aimed to build up a novel diagnostic tool and check it for accuracy. The main objective of our work was to differentiate between healthy skin (C), atopic dermatitis (AD) and psoriasis vulgaris (PV) biopsies on the base of involucrin (IVL) and human ß-defensin-2 (hBD-2) concentrations and their mRNA, as well as mRNA for TPP2 and PSMB8. (2): ELISA for IVL and hBD-2 proteins and Real-time PCR for the relative expression of mRNA for: IVL (IVL mRNA), hBD-2 (hBD-2 mRNA), PSMB8 (PSMB8 mRNA) and TPP2 (TPP2 mRNA), isolated from skin biopsies taken from AD and PV patients and healthy volunteers were performed. (3): hBD-2 mRNA and PSMB8 mRNA correlated with some parameters of clinical assessment of inflammatory disease severity. hBD-2 mRNA expression, exclusively, was sufficient to distinguish inflammatory skin biopsies from the healthy ones. (4): hBD-2 mRNA and PSMB8 mRNA analysis were the most valuable parameters in differentiating AD and PV biopsies.


Subject(s)
Dermatitis, Atopic , Psoriasis , RNA, Messenger , Skin , beta-Defensins , Humans , Psoriasis/genetics , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/diagnosis , beta-Defensins/genetics , beta-Defensins/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Dermatitis, Atopic/diagnosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Biopsy , Female , Male , Skin/metabolism , Skin/pathology , Adult , Middle Aged , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Diagnosis, Differential , Young Adult , Adolescent
8.
Autophagy ; 20(10): 2221-2237, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39316746

ABSTRACT

Dysregulation in protein homeostasis results in accumulation of protein aggregates, which are sequestered into dedicated insoluble compartments so-called inclusion bodies or aggresomes, where they are scavenged through different mechanisms to reduce proteotoxicity. The protein aggregates can be selectively scavenged by macroautophagy/autophagy called aggrephagy, which is mediated by the autophagic receptor SQSTM1. In this study, we have identified PLK2 as an important regulator of SQSTM1-mediated aggregation of polyubiquitinated proteins. PLK2 is upregulated following proteasome inhibition, and then associates with and phosphorylates SQSTM1 at S349. The phosphorylation of SQSTM1 S349 strengthens its binding to KEAP1, which is required for formation of large SQSTM1 aggregates/bodies upon proteasome inhibition. Our findings suggest that PLK2-mediated phosphorylation of SQSTM1 S349 represents a critical regulatory mechanism in SQSTM1-mediated aggregation of polyubiquitinated proteins.


Subject(s)
Proteasome Endopeptidase Complex , Protein Aggregates , Protein Serine-Threonine Kinases , Sequestosome-1 Protein , Sequestosome-1 Protein/metabolism , Phosphorylation , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitinated Proteins/metabolism , Autophagy/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , HEK293 Cells , Ubiquitination , Protein Binding
9.
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
10.
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
11.
J Clin Invest ; 134(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39286982

ABSTRACT

Maintaining protein homeostasis (proteostasis) requires precise control of protein folding and degradation. Failure to properly respond to stresses disrupts proteostasis, which is a hallmark of many diseases, including cataracts. Hibernators are natural cold-stress adaptors; however, little is known about how they keep a balanced proteome under conditions of drastic temperature shift. Intriguingly, we identified a reversible lens opacity phenotype in ground squirrels (GSs) associated with their hibernation-rewarming process. To understand this "cataract-reversing" phenomenon, we first established induced lens epithelial cells differentiated from GS-derived induced pluripotent stem cells, which helped us explore the molecular mechanism preventing the accumulation of protein aggregates in GS lenses. We discovered that the ubiquitin-proteasome system (UPS) played a vital role in minimizing the aggregation of the lens protein αA-crystallin (CRYAA) during rewarming. Such function was, for the first time to our knowledge, associated with an E3 ubiquitin ligase, RNF114, which appears to be one of the key mechanisms mediating the turnover and homeostasis of lens proteins. Leveraging this knowledge gained from hibernators, we engineered a deliverable RNF114 complex and successfully reduced lens opacity in rats with cold-induced cataracts and zebrafish with oxidative stress-related cataracts. These data provide new insights into the critical role of the UPS in maintaining proteostasis in cold and possibly other forms of stresses. The newly identified E3 ubiquitin ligase RNF114, related to CRYAA, offers a promising avenue for treating cataracts with protein aggregates.


Subject(s)
Cataract , Hibernation , Lens, Crystalline , Sciuridae , Ubiquitin-Protein Ligases , Animals , Cataract/genetics , Cataract/pathology , Cataract/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Cold Temperature , Proteasome Endopeptidase Complex/metabolism , Proteostasis , Induced Pluripotent Stem Cells/metabolism , Crystallins/metabolism , Crystallins/genetics , Rats
12.
Sci Adv ; 10(38): eadp0334, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292784

ABSTRACT

Lin28, a highly conserved carcinogenic protein, plays an important role in the generation of cancer stem cells, contributing to the unfavorable prognosis of cancer patients. This RNA binding protein specifically binds to pri/pre-microRNA (miRNA) lethal-7 (let-7), impeding its miRNA maturation. The reduced expression of tumor suppressor miRNA let-7 fosters development and progression-related traits such as proliferation, invasion, metastasis, and drug resistance. We report a series of miRNA-based Lin28A-miRNA proteolysis-targeting chimeras (Lin28A-miRNA-PROTACs) designed to efficiently degrade Lin28A through a ubiquitin-proteasome-dependent mechanism, resulting in up-regulation of mature let-7 family. The augmented levels of matured let-7 miRNAs further exert inhibitory effects on cancer cell proliferation and migration, and increase its sensitivity to chemotherapy. In a mouse ectopic tumor model, Lin28A-miRNA-PROTAC demonstrates a substantial efficacy in inhibiting tumor growth. When combined with tamoxifen, the tumors exhibit gradual regression. This study displays an effective miRNA-based PROTACs to degrade Lin28A and inhibit tumor growth, providing a promising therapeutic avenue for cancer treatment with miRNA-based therapy.


Subject(s)
Breast Neoplasms , Cell Proliferation , MicroRNAs , Proteolysis , RNA-Binding Proteins , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Animals , Female , Mice , Proteolysis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis Targeting Chimera
13.
Cells ; 13(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273003

ABSTRACT

TRIM44, a tripartite motif (TRIM) family member, is pivotal in linking the ubiquitin-proteasome system (UPS) to autophagy in multiple myeloma (MM). However, its prognostic impact and therapeutic potential remain underexplored. Here, we report that TRIM44 overexpression is associated with poor prognosis in a Multiple Myeloma Research Foundation (MMRF) cohort of 858 patients, persisting across primary and recurrent MM cases. TRIM44 expression notably increases in advanced MM stages, indicating its potential role in disease progression. Single-cell RNA sequencing across MM stages showed significant TRIM44 upregulation in smoldering MM (SMM) and MM compared to normal bone marrow, especially in patients with t(4;14) cytogenetic abnormalities. This analysis further identified high TRIM44 expression as predictive of lower responsiveness to proteasome inhibitor (PI) treatments, underscoring its critical function in the unfolded protein response (UPR) in TRIM44-high MM cells. Our findings also demonstrate that TRIM44 facilitates SQSTM1 oligomerization under oxidative stress, essential for its phosphorylation and subsequent autophagic degradation. This process supports the survival of PI-resistant MM cells by activating the NRF2 pathway, which is crucial for oxidative stress response and, potentially, other chemotherapy-induced stressors. Additionally, TRIM44 counters the TRIM21-mediated suppression of the antioxidant response, enhancing MM cell survival under oxidative stress. Collectively, our discoveries highlight TRIM44's significant role in MM progression and resistance to therapy, suggesting its potential value as a therapeutic target.


Subject(s)
Multiple Myeloma , Proteasome Endopeptidase Complex , Tripartite Motif Proteins , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/genetics , Humans , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Prognosis , Cell Line, Tumor , Proteasome Endopeptidase Complex/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Autophagy/genetics , Cell Survival/drug effects , Cell Survival/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Proteasome Inhibitors/pharmacology , Drug Resistance, Neoplasm/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Gene Expression Regulation, Neoplastic
14.
Front Immunol ; 15: 1464267, 2024.
Article in English | MEDLINE | ID: mdl-39281672

ABSTRACT

Immunoproteasome is a specialized form of proteasome which plays a crucial role in antigen processing and presentation, and enhances immune responses against malignant cells. This review explores the role of immunoproteasome in the anti-tumor immune responses, including immune surveillance and modulation of the tumor microenvironment, as well as its potential as a target for cancer immunotherapy. Furthermore, we have also discussed the therapeutic potential of immunoproteasome inhibitors, strategies to enhance antigen presentation and combination therapies. The ongoing trials and case studies in urology, melanoma, lung, colorectal, and breast cancers have also been summarized. Finally, the challenges facing clinical translation of immunoproteasome-targeted therapies, such as toxicity and resistance mechanisms, and the future research directions have been addressed. This review underscores the significance of targeting the immunoproteasome in combination with other immunotherapies for solid tumors and its potential broader applications in other diseases.


Subject(s)
Immunotherapy , Neoplasms , Proteasome Endopeptidase Complex , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/immunology , Tumor Microenvironment/immunology , Animals , Proteasome Inhibitors/therapeutic use , Proteasome Inhibitors/pharmacology , Carcinoma/immunology , Carcinoma/therapy
15.
Mol Plant Pathol ; 25(9): e70006, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39267531

ABSTRACT

The pathogen Agrobacterium tumefaciens is known for causing crown gall tumours in plants. However, it has also been harnessed as a valuable tool for plant genetic transformation. Apart from the T-DNA, Agrobacterium also delivers at least five virulence proteins into the host plant cells, which are required for an efficient infection. One of these virulence proteins is VirD5. F-box proteins, encoded in the host plant genome or the Ti plasmid, and the ubiquitin/26S proteasome system (UPS) also play an important role in facilitating Agrobacterium infection. Our study identified two Arabidopsis F-box proteins, D5BF1 and D5BF2, that bind VirD5 and facilitate its degradation via the UPS. Additionally, we found that Agrobacterium partially suppresses the expression of D5BF1 and D5BF2. Lastly, stable transformation and tumorigenesis efficiency assays revealed that D5BF1 and D5BF2 negatively regulate the Agrobacterium infection process, showing that the plant F-box proteins and UPS play a role in defending against Agrobacterium infection.


Subject(s)
Agrobacterium tumefaciens , Arabidopsis Proteins , Arabidopsis , F-Box Proteins , Transformation, Genetic , Arabidopsis/microbiology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/pathogenicity , F-Box Proteins/metabolism , F-Box Proteins/genetics , Carcinogenesis/genetics , Plant Tumors/microbiology , Proteasome Endopeptidase Complex/metabolism , Gene Expression Regulation, Plant
16.
Elife ; 122024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255192

ABSTRACT

Proteotoxic stress impairs cellular homeostasis and underlies the pathogenesis of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). The proteasomal and autophagic degradation of proteins are two major pathways for protein quality control in the cell. Here, we report a genome-wide CRISPR screen uncovering a major regulator of cytotoxicity resulting from the inhibition of the proteasome. Dihydrolipoamide branched chain transacylase E2 (DBT) was found to be a robust suppressor, the loss of which protects against proteasome inhibition-associated cell death through promoting clearance of ubiquitinated proteins. Loss of DBT altered the metabolic and energetic status of the cell and resulted in activation of autophagy in an AMP-activated protein kinase (AMPK)-dependent mechanism in the presence of proteasomal inhibition. Loss of DBT protected against proteotoxicity induced by ALS-linked mutant TDP-43 in Drosophila and mammalian neurons. DBT is upregulated in the tissues of ALS patients. These results demonstrate that DBT is a master switch in the metabolic control of protein quality control with implications in neurodegenerative diseases.


Subject(s)
Proteasome Endopeptidase Complex , Proteostasis , Animals , Proteasome Endopeptidase Complex/metabolism , Humans , Drosophila/metabolism , Autophagy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Neurons/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics
17.
Elife ; 132024 Sep 04.
Article in English | MEDLINE | ID: mdl-39230574

ABSTRACT

Proteasomes are essential molecular machines responsible for the degradation of proteins in eukaryotic cells. Altered proteasome activity has been linked to neurodegeneration, auto-immune disorders and cancer. Despite the relevance for human disease and drug development, no method currently exists to monitor proteasome composition and interactions in vivo in animal models. To fill this gap, we developed a strategy based on tagging of proteasomes with promiscuous biotin ligases and generated a new mouse model enabling the quantification of proteasome interactions by mass spectrometry. We show that biotin ligases can be incorporated in fully assembled proteasomes without negative impact on their activity. We demonstrate the utility of our method by identifying novel proteasome-interacting proteins, charting interactomes across mouse organs, and showing that proximity-labeling enables the identification of both endogenous and small-molecule-induced proteasome substrates.


Subject(s)
Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Animals , Mice , Humans , Mass Spectrometry/methods , Protein Interaction Mapping
18.
J Med Virol ; 96(9): e29891, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39223933

ABSTRACT

The ubiquitin-proteasome system is frequently employed to degrade viral proteins, thereby inhibiting viral replication and pathogenicity. Through an analysis of the degradation kinetics of all the SARS-CoV-2 proteins, our study revealed rapid degradation of several proteins, particularly NSP5. Additionally, we identified FBXO22, an E3 ubiquitin ligase, as the primary regulator of NSP5 ubiquitination. Moreover, we validated the interaction between FBXO22 and NSP5, demonstrating that FBXO22-mediated ubiquitination of NSP5 facilitated its recognition by the proteasome, leading to subsequent degradation. Specifically, FBXO22 catalyzed the formation of K48-linked polyubiquitin chains on NSP5 at lysine residues 5 and 90. Knockdown of FBXO22 resulted in decreased NSP5 ubiquitination levels, increased stability, and enhanced ability to evade the host innate immune response. Notably, the protein level of FBXO22 were negatively correlated with SARS-CoV-2 load, highlighting its importance in inhibiting viral replication. This study elucidates the molecular mechanism by which FBXO22 mediates the degradation of NSP5 and underscores its critical role in limiting viral replication. The identification of FBXO22 as a regulator of NSP5 stability provides new insights and potential avenues for targeting NSP5 in antiviral strategies.


Subject(s)
Proteasome Endopeptidase Complex , SARS-CoV-2 , Ubiquitination , Virus Replication , Humans , Proteasome Endopeptidase Complex/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , COVID-19/virology , COVID-19/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Proteolysis , Coronavirus Papain-Like Proteases/metabolism , Receptors, Cytoplasmic and Nuclear
19.
Cancer Immunol Immunother ; 73(11): 228, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249578

ABSTRACT

BACKGROUND: The antigen processing machinery (APM) plays a critical role in generating tumor-specific antigens that can be recognized and targeted by the immune system. Proper functioning of APM components is essential for presenting these antigens on the surface of tumor cells, enabling immune detection and destruction. In many cancers, defects in APM can lead to immune evasion, contributing to tumor progression and poor clinical outcomes. However, the status of the APM in sarcomas is not well characterized, limiting the development of effective immunotherapeutic strategies for these patients. METHODS: We investigated 126 patients with 8 types of bone and soft tissue sarcoma operated between 2001-2021. Tissue microarrays mapped 11 specific areas in each case. The presence/absence of APM protein was determined through immunohistochemistry. Bayesian networks were used. RESULTS: All investigated sarcomas had some defects in APM. The least damaged component was HLA Class I subunit ß2-microglobulin and HLA Class II. The proteasome LMP10 subunit was defective in leiomyosarcoma (LMS), myxoid liposarcoma (MLPS), and dedifferentiated liposarcoma (DDLPS), while MHC I transporting unit TAP2 was altered in undifferentiated pleomorphic sarcoma (UPS), gastrointestinal stromal tumor (GIST), and chordoma (CH). Among different neoplastic areas, high-grade areas showed different patterns of expression compared to high lymphocytic infiltrate areas. Heterogeneity at the patient level was also observed. Loss of any APM component was prognostic of distant metastasis (DM) for LMS and DDLPS and of overall survival (OS) for LMS. CONCLUSION: Sarcomas exhibit a high degree of defects in APM components, with differences among histotypes and tumoral areas. The most commonly altered APM components were HLA Class I subunit ß2-microglobulin, HLA Class I subunit α (HC10), and MHC I transporting unit TAP2. The loss of APM components was prognostic of DM and OS and clinically relevant for LMS and DDLPS. This study explores sarcoma molecular mechanisms, enriching personalized therapeutic approaches.


Subject(s)
Antigen Presentation , Sarcoma , Humans , Sarcoma/immunology , Sarcoma/pathology , Antigen Presentation/immunology , Male , Female , Middle Aged , Aged , Adult , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Proteasome Endopeptidase Complex/metabolism , beta 2-Microglobulin/metabolism , Prognosis , ATP Binding Cassette Transporter, Subfamily B, Member 3
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL