Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Biochem ; 175(5): 473-480, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38348509

ABSTRACT

Aging is a major risk factor for many diseases. Recent studies have shown that age-related disruption of proteostasis leads to the accumulation of abnormal proteins and that dysfunction of the two major intracellular proteolytic pathways, the ubiquitin-proteasome pathway, and the autophagy-lysosome pathway, is largely responsible for this process. Conversely, it has been shown that activation of these proteolytic pathways may contribute to lifespan extension and suppression of pathological conditions, making it a promising intervention for anti-aging. This review provides an overview of the important role of intracellular protein degradation in aging and summarizes how the disruption of proteostasis is involved in age-related diseases.


Subject(s)
Aging , Autophagy , Cellular Senescence , Proteasome Endopeptidase Complex , Proteolysis , Proteostasis , Humans , Cellular Senescence/physiology , Aging/metabolism , Animals , Autophagy/physiology , Proteostasis/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Lysosomes/metabolism
2.
Cell Rep ; 42(8): 112880, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37541257

ABSTRACT

The proteasome plays a central role in intracellular protein degradation. Age-dependent decline in proteasome activity is associated with cellular senescence and organismal aging; however, the mechanism by which the proteasome plays a role in senescent cells remains elusive. Here, we show that nuclear foci that contain the proteasome and exhibit liquid-like properties are formed in senescent cells. The formation of senescence-associated nuclear proteasome foci (SANPs) is dependent on ubiquitination and RAD23B, similar to previously known nuclear proteasome foci, but also requires proteasome activity. RAD23B knockdown suppresses SANP formation and increases mitochondrial activity, leading to reactive oxygen species production without affecting other senescence traits such as cell-cycle arrest and cell morphology. These findings suggest that SANPs are an important feature of senescent cells and uncover a mechanism by which the proteasome plays a role in senescent cells.


Subject(s)
Cell Nucleus , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Cell Nucleus/metabolism , Ubiquitination , Cellular Senescence
3.
Endocrinology ; 164(6)2023 04 17.
Article in English | MEDLINE | ID: mdl-37103220

ABSTRACT

Muscle atrophy is the cause and consequence of obesity. Proteasome dysfunction mediates obesity-induced endoplasmic reticulum (ER) stress and insulin resistance in the liver and adipose tissues. However, obesity-associated regulation of proteasome function and its role in the skeletal muscles remains underinvestigated. Here, we established skeletal muscle-specific 20S proteasome assembly chaperone-1 (PAC1) knockout (mPAC1KO) mice. A high-fat diet (HFD) activated proteasome function by ∼8-fold in the skeletal muscles, which was reduced by 50% in mPAC1KO mice. mPAC1KO induced unfolded protein responses in the skeletal muscles, which were reduced by HFD. Although the skeletal muscle mass and functions were not different between the genotypes, genes involved in the ubiquitin proteasome complex, immune response, endoplasmic stress, and myogenesis were coordinately upregulated in the skeletal muscles of mPAC1KO mice. Therefore, we introduced an immobilization-induced muscle atrophy model in obesity by combining HFD and immobilization. mPAC1KO downregulated atrogin-1 and MuRF1, together with their upstream Foxo1 and Klf15, and protected against disused skeletal muscle mass reduction. In conclusion, obesity elevates proteasome functions in the skeletal muscles. PAC1 deficiency protects mice from immobilization-induced muscle atrophy in obesity. These findings suggest obesity-induced proteasome activation as a possible therapeutic target for immobilization-induced muscle atrophy.


Subject(s)
Muscular Atrophy , Proteasome Endopeptidase Complex , Mice , Male , Animals , Proteasome Endopeptidase Complex/metabolism , Mice, Obese , Muscular Atrophy/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism
4.
Nat Commun ; 12(1): 6819, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819510

ABSTRACT

Impaired proteasome activity due to genetic variants of certain subunits might lead to proteasome-associated autoinflammatory syndromes (PRAAS). Here we report a de novo heterozygous missense variant of the PSMB9 proteasome subunit gene in two unrelated Japanese infants resulting in amino acid substitution of the glycine (G) by aspartic acid (D) at position 156 of the encoded protein ß1i. In addition to PRAAS-like manifestations, these individuals suffer from pulmonary hypertension and immunodeficiency, which are distinct from typical PRAAS symptoms. The missense variant results in impaired immunoproteasome maturation and activity, yet ubiquitin accumulation is hardly detectable in the patients. A mouse model of the heterozygous human genetic variant (Psmb9G156D/+) recapitulates the proteasome defects and the immunodeficiency phenotype of patients. Structurally, PSMB9 G156D interferes with the ß-ring-ßring interaction of the wild type protein that is necessary for 20S proteasome formation. We propose the term, proteasome-associated autoinflammatory syndrome with immunodeficiency (PRAAS-ID), to indicate a separate category of autoinflammatory diseases, similar to, but distinct from PRAAS, that describes the patients in this study.


Subject(s)
Cysteine Endopeptidases/genetics , Hereditary Autoinflammatory Diseases/genetics , Hypertension, Pulmonary/genetics , Primary Immunodeficiency Diseases/genetics , Proteasome Endopeptidase Complex/metabolism , Animals , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Female , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Heterozygote , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/immunology , Infant, Newborn , Male , Mice , Mice, Transgenic , Mutation, Missense , Pedigree , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Proteasome Endopeptidase Complex/genetics , Syndrome
5.
iScience ; 23(7): 101299, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32634741

ABSTRACT

The proteasome is a therapeutic target in cancer, but resistance to proteasome inhibitors often develops owing to the induction of compensatory pathways. Through a genome-wide siRNA screen combined with RNA sequencing analysis, we identified hexokinase and downstream O-GlcNAcylation as cell survival factors under proteasome impairment. The inhibition of O-GlcNAcylation synergistically induced massive cell death in combination with proteasome inhibition. We further demonstrated that O-GlcNAcylation was indispensable for maintaining proteasome activity by enhancing biogenesis as well as proteasome degradation in a manner independent of Nrf1, a well-known compensatory transcription factor that upregulates proteasome gene expression. Our results identify a pathway that maintains proteasome function under proteasome impairment, providing potential targets for cancer therapy.

6.
Int J Mol Sci ; 21(10)2020 May 23.
Article in English | MEDLINE | ID: mdl-32456207

ABSTRACT

Protein folding is a substantively error prone process, especially when it occurs in the endoplasmic reticulum (ER). The highly exquisite machinery in the ER controls secretory protein folding, recognizes aberrant folding states, and retrotranslocates permanently misfolded proteins from the ER back to the cytosol; these misfolded proteins are then degraded by the ubiquitin-proteasome system termed as the ER-associated degradation (ERAD). The 26S proteasome is a multisubunit protease complex that recognizes and degrades ubiquitinated proteins in an ATP-dependent manner. The complex structure of the 26S proteasome requires exquisite regulation at the transcription, translation, and molecular assembly levels. Nuclear factor erythroid-derived 2-related factor 1 (Nrf1; NFE2L1), an ER-resident transcription factor, has recently been shown to be responsible for the coordinated expression of all the proteasome subunit genes upon proteasome impairment in mammalian cells. In this review, we summarize the current knowledge regarding the transcriptional regulation of the proteasome, as well as recent findings concerning the regulation of Nrf1 transcription activity in ER homeostasis and metabolic processes.


Subject(s)
Nuclear Respiratory Factor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteostasis , Animals , Endoplasmic Reticulum-Associated Degradation , Humans , Nuclear Respiratory Factor 1/genetics , Proteasome Endopeptidase Complex/genetics
7.
Mol Cell Biol ; 40(10)2020 04 28.
Article in English | MEDLINE | ID: mdl-32123008

ABSTRACT

Proteasomes are essential protease complexes that maintain cellular homeostasis, and aberrant proteasomal activity supports cancer development. The regulatory mechanisms and biological function of the ubiquitin-26S proteasome have been studied extensively, while those of the ubiquitin-independent 20S proteasome system remain obscure. Here, we show that the cap 'n' collar (CNC) family transcription factor NRF3 specifically enhances 20S proteasome assembly in cancer cells and that 20S proteasomes contribute to colorectal cancer development through ubiquitin-independent proteolysis of the tumor suppressor p53 and retinoblastoma (Rb) proteins. The NRF3 gene is highly expressed in many cancer tissues and cell lines and is important for cancer cell growth. In cancer cells, NRF3 upregulates the assembly of the 20S proteasome by directly inducing the gene expression of the 20S proteasome maturation protein POMP. Interestingly, NRF3 knockdown not only increases p53 and Rb protein levels but also increases p53 activities for tumor suppression, including cell cycle arrest and induction of apoptosis. Furthermore, protein stability and cell viability assays using two distinct proteasome inhibitor anticancer drugs, the 20S proteasome inhibitor bortezomib and the ubiquitin-activating enzyme E1 inhibitor TAK-243, show that the upregulation of the NRF3-POMP axis leads to ubiquitin-independent proteolysis of p53 and Rb and to impaired sensitivity to bortezomib but not TAK-243. More importantly, the NRF3-POMP axis supports tumorigenesis and metastasis, with higher NRF3/POMP expression levels correlating with poor prognoses in patients with colorectal or rectal adenocarcinoma. These results suggest that the NRF3-POMP-20S proteasome assembly axis is significant for cancer development via ubiquitin-independent proteolysis of tumor suppressor proteins.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Molecular Chaperones/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , HCT116 Cells , HeLa Cells , Humans , Proteolysis , Ubiquitin/metabolism
8.
Cell Rep ; 29(9): 2901-2916.e6, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31775054

ABSTRACT

The thymic function to produce self-protective and self-tolerant T cells is chiefly mediated by cortical thymic epithelial cells (cTECs) and medullary TECs (mTECs). Recent studies including single-cell transcriptomic analyses have highlighted a rich diversity in functional mTEC subpopulations. Because of their limited cellularity, however, the biochemical characterization of TECs, including the proteomic profiling of cTECs and mTECs, has remained unestablished. Utilizing genetically modified mice that carry enlarged but functional thymuses, here we show a combination of proteomic and transcriptomic profiles for cTECs and mTECs, which identified signature molecules that characterize a developmental and functional contrast between cTECs and mTECs. Our results reveal a highly specific impact of the thymoproteasome on proteasome subunit composition in cTECs and provide an integrated trans-omics platform for further exploration of thymus biology.


Subject(s)
Epithelial Cells/metabolism , Proteomics/methods , Thymus Gland/physiopathology , Cell Differentiation , Humans
9.
Genes Cells ; 24(12): 801-813, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31621149

ABSTRACT

The proteasome degradation machinery is essential for a variety of cellular processes including senescence and T-cell immunity. Decreased proteasome activity is associated with the aging process; however, the regulation of the proteasome in CD4+ T cells in relation to aging is unclear. Here, we show that defects in the induction of the proteasome in CD4+ T cells upon T-cell receptor (TCR) stimulation underlie T-cell senescence. Proteasome dysfunction promotes senescence-associated phenotypes, including defective proliferation, cytokine production and increased levels of PD-1+ CD44High CD4+ T cells. Proteasome induction by TCR signaling via MEK-, IKK- and calcineurin-dependent pathways is attenuated with age and decreased in PD-1+ CD44High CD4+ T cells, the proportion of which increases with age. Our results indicate that defective induction of the proteasome is a hallmark of CD4+ T-cell senescence.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cellular Senescence , Proteasome Endopeptidase Complex/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , CD4-Positive T-Lymphocytes/physiology , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/genetics , Signal Transduction
10.
Genes Cells ; 24(8): 559-568, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31210371

ABSTRACT

Maintaining protein homeostasis is central to cell survival. The ubiquitin-proteasome system and autophagy play pivotal roles in protein quality control through protein degradation. Activities of these degradative pathways are carefully orchestrated, and autophagy is up-regulated during proteasome dysfunction for cellular homeostasis. However, the mechanism by which proteasome impairment induces compensatory autophagy has remained largely elusive. Here, we show that FAM48A mediates autophagy induction during proteasome inhibition. FAM48A is degraded by the proteasome and accumulates in cells by proteasome inhibition. Knockdown of FAM48A led to defective induction of autophagy during proteasome inhibition and accompanied by defective localization of Atg9 on recycling endosomes. Our results indicate that FAM48A is a kind of sensor that is required for compensatory autophagy induction upon proteasome impairment.


Subject(s)
Autophagy , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/genetics , Autophagy/genetics , Autophagy-Related Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Humans , Immunohistochemistry , Substrate Specificity , Transcription Factors/metabolism
11.
Cell Microbiol ; 21(3): e12974, 2019 03.
Article in English | MEDLINE | ID: mdl-30414351

ABSTRACT

Subversion of antigen-specific immune responses by intracellular pathogens is pivotal for successful colonisation. Bacterial pathogens, including Shigella, deliver effectors into host cells via the type III secretion system (T3SS) in order to manipulate host innate and adaptive immune responses, thereby promoting infection. However, the strategy for subverting antigen-specific immunity is not well understood. Here, we show that Shigella flexneri invasion plasmid antigen H (IpaH) 4.5, a member of the E3 ubiquitin ligase effector family, targets the proteasome regulatory particle non-ATPase 13 (RPN13) and induces its degradation via the ubiquitin-proteasome system (UPS). IpaH4.5-mediated RPN13 degradation causes dysfunction of the 19S regulatory particle (RP) in the 26S proteasome, inhibiting guidance of ubiquitinated proteins to the proteolytically active 20S core particle (CP) of 26S proteasome and thereby suppressing proteasome-catalysed peptide splicing. This, in turn, reduces antigen cross-presentation to CD8+ T cells via major histocompatibility complex (MHC) class I in vitro. In RPN13 knockout mouse embryonic fibroblasts (MEFs), loss of RPN13 suppressed CD8+ T cell priming during Shigella infection. Our results uncover the unique tactics employed by Shigella to dampen the antigen-specific cytotoxic T lymphocyte (CTL) response.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Immune Evasion , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Proteasome Endopeptidase Complex/metabolism , Shigella flexneri/growth & development , T-Lymphocytes, Cytotoxic/immunology , Animals , Cells, Cultured , Cluster Analysis , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Disease Models, Animal , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Humans , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Phylogeny , RNA, Ribosomal/genetics , Sequence Analysis, DNA , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , T-Lymphocytes, Cytotoxic/microbiology , Virulence Factors/metabolism
12.
Mol Cell Biol ; 39(1)2019 01 01.
Article in English | MEDLINE | ID: mdl-30348842

ABSTRACT

The proteasome is the proteolytic machinery at the center of regulated intracellular protein degradation and participates in various cellular processes. Maintaining the quality of the proteasome is therefore important for proper cell function. It is unclear, however, how proteasomes change over time and how aged proteasomes are disposed. Here, we show that the proteasome undergoes specific biochemical alterations as it ages. We generated Rpn11-Flag/enhanced green fluorescent protein (EGFP) tag-exchangeable knock-in mice and established a method for selective purification of old proteasomes in terms of their molecular age at the time after synthesis. The half-life of proteasomes in mouse embryonic fibroblasts isolated from these knock-in mice was about 16 h. Using this tool, we found increased association of Txnl1, Usp14, and actin with the proteasome and specific phosphorylation of Rpn3 at Ser 6 in 3-day-old proteasomes. We also identified CSNK2A2 encoding the catalytic α' subunit of casein kinase II (CK2α') as a responsible gene that regulates the phosphorylation and turnover of old proteasomes. These findings will provide a basis for understanding the mechanism of molecular aging of the proteasome.


Subject(s)
Aging/physiology , Casein Kinase II/genetics , Proteasome Endopeptidase Complex/genetics , Ubiquitin/metabolism , Animals , Gene Knock-In Techniques/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/genetics
13.
Proc Jpn Acad Ser B Phys Biol Sci ; 94(8): 325-336, 2018.
Article in English | MEDLINE | ID: mdl-30305478

ABSTRACT

The 26S proteasome is a large protease complex that selectively degrades ubiquitinated proteins. It comprises 33 distinct subunits, each of which differ in function and structure, and which cannot be substituted by the other subunits. Owing to its complicated structure, the biogenesis of the 26S proteasome is elaborately regulated at the transcription, translation, and molecular assembly levels. Recent studies revealed that Nrf1 (NFE2L1) is a transcription factor that upregulates the expression of all the proteasome subunit genes in a concerted manner, especially during proteasome impairment in mammalian cells. In this review, we summarize current knowledge regarding the transcriptional regulation of the proteasome and recent findings concerning the regulation of Nrf1 transcription activity.


Subject(s)
Gene Expression Regulation , Nuclear Respiratory Factor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription, Genetic , Humans , Molecular Targeted Therapy , Nuclear Respiratory Factor 1/chemistry , Protein Deficiency
14.
Genes Cells ; 23(10): 839-848, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30133132

ABSTRACT

The proteasome core particle (CP) is a cytoplasmic and nuclear protease complex and is comprised of two α-rings and two ß-rings stacked in order of αßßα. The assembly of CP proceeds by ordered recruitment of ß-subunits on an α-ring with help of assembly chaperones PAC1-PAC2, PAC3-PAC4, and UMP1. However, the mechanism of α-ring formation remains unsolved. Here, we show that α4, α5, α6, and α7 form a core intermediate as the initial process of α-ring assembly, which requires PAC3-PAC4. α1 and α3 can be incorporated independently into the core α4-α7 intermediate, whereas α2 incorporation is dependent on preceding incorporation of α1. Through these processes, PAC1-PAC2 prevents nonproductive dimerization of α-ring assembly intermediates. We also found that PAC1-PAC2 overrides the effect of nuclear localization signals of α-subunits and retains α-ring assembly intermediates in the cytoplasm. Our results first show a detailed assembly pathway of proteasomal α-ring and explain the mechanism by which CP assembly occurs in the cytoplasm.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Cytoplasm , HEK293 Cells , Humans , Models, Biological , Models, Molecular , Molecular Chaperones/metabolism , Protein Binding , Protein Subunits/metabolism , RNA, Small Interfering
15.
Mol Cell Biol ; 38(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29109084

ABSTRACT

Ubiquitin-mediated protein degradation plays essential roles in proteostasis and is involved in the pathogenesis of neurodegenerative diseases in which ubiquitin-positive aberrant proteins accumulate. However, how such aberrant proteins are processed inside cells has not been fully explored. Here, we show that the product of CG5445, a previously uncharacterized Drosophila gene, prevents the accumulation of aggregate-prone ubiquitinated proteins. We found that ubiquitin conjugates were associated with CG5445, the knockdown of which caused the accumulation of detergent-insoluble ubiquitinated proteins. Furthermore, CG5445 rescued eye degeneration caused by the amyotrophic lateral sclerosis (ALS)-linked mutant TAR DNA-binding protein of 43 kDa (TDP-43), which often forms ubiquitin-positive aggregates in cells through the capacity of CG5445 to bind to ubiquitin chains. Biochemically, CG5445 inhibited the accumulation of insoluble forms and promoted their clearance. Our results demonstrate a new possible mechanism by which cells maintain ubiquitinated aggregation-prone proteins in a soluble form to decrease their cytotoxicity until they are degraded.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Eye/pathology , Gene Expression Regulation , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Solubility , Ubiquitin/metabolism , Ubiquitination
16.
J Ovarian Res ; 10(1): 53, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28784174

ABSTRACT

BACKGROUND: Ovarian carcinoma is highly dependent on the ubiquitin proteasome system (UPS), but its clinical response to treatment with the proteasome inhibitor bortezomib has been disappointing. This has driven exploration of alternate approaches to target the UPS in ovarian cancer. Recently, proteasome inhibitors targeting the 19S regulatory particle-associated RPN13 protein have been described, such as RA190. RPN13, which is encoded by ADRM1, facilitates the recognition by the proteasome of its polyubiquinated substrates. Inhibition of RPN13 produces a rapid, toxic accumulation of polyubiquitinated proteins in ovarian and other cancer cells, triggering apoptosis. Here, we sought to determine if RPN13 is available as a target in precursors of ovarian/fallopian tube cancer as well as all advanced cases, and the impact of increased ADRM1 gene copy number on sensitivity of ovarian cancer to RA190. METHODS: ADRM1 mRNA was quantified by RNAscope in situ hybridization and RPN13 protein detected by immunohistochemistry in high grade serous carcinoma (HGSC) of the ovary and serous tubal intraepithelial carcinoma (STIC). Amplification of ADRM1 and sensitivity to RA190 were determined in ovarian cancer cell lines. RESULTS: Here, we demonstrate that expression of ADRM1mRNA is significantly elevated in STIC and HGSC as compared to normal fallopian tube epithelium. ADRM1 mRNA and RPN13 were ubiquitously and robustly expressed in ovarian carcinoma tissue and cell lines. No correlation was found between ADRM1 amplification and sensitivity of ovarian cancer cell lines to RA190, but all were susceptible. CONCLUSIONS: RPN13 can potentially be targeted by RA190 in both in situ and metastatic ovarian carcinoma. Ovarian cancer cell lines are sensitive to RA190 regardless of whether the ADRM1 gene is amplified.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Fallopian Tube Neoplasms/genetics , Membrane Glycoproteins/genetics , Ovarian Neoplasms/genetics , Aged , Benzylidene Compounds/pharmacology , Cell Line, Tumor , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Middle Aged , Ovarian Neoplasms/metabolism , Proteasome Inhibitors/pharmacology , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics
17.
Nat Commun ; 8: 15540, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28598414

ABSTRACT

Proteasome-ubiquitin receptor hRpn13/Adrm1 binds and activates deubiquitinating enzyme Uch37/UCHL5 and is targeted by bis-benzylidine piperidone RA190, which restricts cancer growth in mice xenografts. Here, we solve the structure of hRpn13 with a segment of hRpn2 that serves as its proteasome docking site; a proline-rich C-terminal hRpn2 extension stretches across a narrow canyon of the ubiquitin-binding hRpn13 Pru domain blocking an RA190-binding surface. Biophysical analyses in combination with cell-based assays indicate that hRpn13 binds preferentially to hRpn2 and proteasomes over RA190. hRpn13 also exists outside of proteasomes where it may be RA190 sensitive. RA190 does not affect hRpn13 interaction with Uch37, but rather directly binds and inactivates Uch37. hRpn13 deletion from HCT116 cells abrogates RA190-induced accumulation of substrates at proteasomes. We propose that RA190 targets hRpn13 and Uch37 through parallel mechanisms and at proteasomes, RA190-inactivated Uch37 cannot disassemble hRpn13-bound ubiquitin chains.


Subject(s)
Antineoplastic Agents/chemistry , Benzylidene Compounds/chemistry , Hexosyltransferases/metabolism , Membrane Glycoproteins/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin Thiolesterase/metabolism , Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Biophysics , Drug Screening Assays, Antitumor , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins , Neoplasms/drug therapy , Proline/chemistry , Protein Binding , Protein Domains
18.
Nat Commun ; 8: 14419, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176764

ABSTRACT

The thymus is an organ that produces functionally competent T cells that protect us from pathogens and malignancies. Foxn1 is a transcription factor that is essential for thymus organogenesis; however, the direct target for Foxn1 to actuate thymic T-cell production is unknown. Here we show that a Foxn1-binding cis-regulatory element promotes the transcription of ß5t, which has an essential role in cortical thymic epithelial cells to induce positive selection of functionally competent CD8+ T cells. A point mutation in this genome element results in a defect in ß5t expression and CD8+ T-cell production in mice. The results reveal a Foxn1-ß5t transcriptional axis that governs CD8+ T-cell production in the thymus.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Proteasome Endopeptidase Complex/genetics , Thymus Gland/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Epithelial Cells/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Point Mutation , Regulatory Elements, Transcriptional/physiology , Thymus Gland/cytology , Tissue Culture Techniques
19.
Elife ; 52016 08 16.
Article in English | MEDLINE | ID: mdl-27528193

ABSTRACT

In response to proteasome dysfunction, mammalian cells upregulate proteasome gene expression by activating Nrf1. Nrf1 is an endoplasmic reticulum-resident transcription factor that is continually retrotranslocated and degraded by the proteasome. Upon proteasome inhibition, Nrf1 escapes degradation and is cleaved to become active. However, the processing enzyme for Nrf1 remains obscure. Here we show that the aspartyl protease DNA-damage inducible 1 homolog 2 (DDI2) is required to cleave and activate Nrf1. Deletion of DDI2 reduced the cleaved form of Nrf1 and increased the full-length cytosolic form of Nrf1, resulting in poor upregulation of proteasomes in response to proteasome inhibition. These defects were restored by adding back wild-type DDI2 but not protease-defective DDI2. Our results provide a clue for blocking compensatory proteasome synthesis to improve cancer therapies targeting proteasomes.


Subject(s)
Aspartic Acid Proteases/metabolism , Nuclear Respiratory Factor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Aspartic Acid Proteases/genetics , Cell Line , Gene Deletion , Genetic Complementation Test , Humans
20.
PLoS Genet ; 11(7): e1005401, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26222436

ABSTRACT

Intracellular proteins tagged with ubiquitin chains are targeted to the 26S proteasome for degradation. The two subunits, Rpn10 and Rpn13, function as ubiquitin receptors of the proteasome. However, differences in roles between Rpn10 and Rpn13 in mammals remains to be understood. We analyzed mice deficient for Rpn13 and Rpn10. Liver-specific deletion of either Rpn10 or Rpn13 showed only modest impairment, but simultaneous loss of both caused severe liver injury accompanied by massive accumulation of ubiquitin conjugates, which was recovered by re-expression of either Rpn10 or Rpn13. We also found that mHR23B and ubiquilin/Plic-1 and -4 failed to bind to the proteasome in the absence of both Rpn10 and Rpn13, suggesting that these two subunits are the main receptors for these UBL-UBA proteins that deliver ubiquitinated proteins to the proteasome. Our results indicate that Rpn13 mostly plays a redundant role with Rpn10 in recognition of ubiquitinated proteins and maintaining homeostasis in Mus musculus.


Subject(s)
Carrier Proteins/genetics , Cell Adhesion Molecules/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitinated Proteins/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Autophagy-Related Proteins , DNA-Binding Proteins/metabolism , Homeostasis , Intracellular Signaling Peptides and Proteins , Liver/injuries , Mice , Proteasome Endopeptidase Complex/genetics , Protein Binding/genetics , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins , Ubiquitin/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...