PSME3 forms a homohexameric ring structure that associates with the 20S core of the proteasome, replacing the 19S regulator to form the immunoproteasome . Key features include:
Proteasomal Activation: Enhances trypsin-like activity of the proteasome while inhibiting chymotrypsin-like and postglutamyl-preferring (PGPH) subunits .
Ubiquitin-Independent Degradation: Facilitates ATP-independent degradation of substrates such as p21, p53, and steroid receptor coactivators (SRCs) .
Protein Interactions: Binds P53, MDM2, and UBE2V2, influencing apoptosis, cell cycle regulation, and metabolic pathways .
PSME3 is implicated in tumor progression and therapy resistance across multiple cancer types.
Mechanistic Insights:
PSME3’s functions extend beyond cancer:
PSME3 participates in diverse biological processes:
Biomarker Potential: Elevated PSME3 correlates with advanced tumor stages and poor survival in liver, lung, and ovarian cancers .
Therapeutic Targeting:
Mechanistic Studies: Elucidate PSME3’s role in non-cancer conditions (e.g., preeclampsia).
Therapeutic Development: Optimize inhibitors targeting PSME3’s proteasomal activation domain.
Biomarker Validation: Multi-center trials to confirm PSME3’s prognostic utility in diverse cancers.
PSME3 (Proteasome Activator Complex Subunit 3) is a nuclear, homoheptameric protein that functions as a proteasome activator. In the scientific literature, it is also known by several alternative names including PA28γ, 11S REGγ, REG GAMMA, PA28g, and Ki nuclear autoantigen . The protein has a calculated and observed molecular weight of approximately 30 kDa and is encoded by the PSME3 gene (Gene ID: 10197) in humans .
Key functions include:
Regulation of nuclear organization, including nuclear speckles, Cajal bodies, and promyelocytic leukemia bodies
Interaction with cell cycle regulators (p16, p19, p21) and transcriptional regulators (c-Myc, KLF2, SMURF2, NF-κB, LATS1/2)
Promotion of MDM2-TP53 interaction, leading to ubiquitination-dependent proteasomal degradation of TP53
Regulation of cell migration through interaction with the HSP90 co-chaperone NUDC
Facilitation of myoblast differentiation in a proteasome-independent manner
Cleavage Under Targets & Release Using Nuclease (CUT&RUN) has proven effective for investigating PSME3's association with chromatin. This technique revealed that PSME3 extensively associates with chromatin at over 5,000 distinct regions in cycling myoblasts, with a large majority of these peaks co-positive for the active promoter mark H3K4me3 . When studying dynamic chromatin binding across cell state transitions, researchers should consider:
Performing CUT&RUN against endogenous PSME3 populations
Comparing binding patterns at different stages (e.g., cycling cells vs. differentiated cells)
Using mild formaldehyde fixation to reduce lability of transient chromatin interactions
Analyzing binding preferences for different genomic regions (promoters, gene bodies, intergenic regions)
Research data demonstrated PSME3 shows a stronger preference for promoter-proximal regions (≤1kb) than H3K4me3, primarily at the expense of binding to gene bodies or intergenic regions .
PSME3 can be effectively detected using multiple immunological techniques with properly validated antibodies. Commercial antibodies such as 14907-1-AP have been validated for Western blotting (WB), immunohistochemistry (IHC), immunofluorescence/immunocytochemistry (IF/ICC), immunoprecipitation (IP), and ELISA applications .
Application Method | Validation Status | Species Reactivity | Sample Types |
---|---|---|---|
Western Blotting | Validated in 7+ publications | Human, mouse, rat | Cell lysates, tissue extracts |
Immunohistochemistry | Validated in 6+ publications | Human, mouse, rat | Tissue sections, paraffin-embedded samples |
Immunofluorescence | Validated in 2+ publications | Human, mouse | Fixed cells, tissue sections |
Immunoprecipitation | Validated | Human, mouse, rat | Cell lysates |
ELISA | Validated | Human, mouse, rat | Purified proteins, cell lysates |
When designing experiments, researchers should follow specific protocols for each application to ensure optimal results, considering factors such as antibody dilution, incubation conditions, and appropriate positive and negative controls .
PSME3 plays a critical role in myoblast differentiation through a proteasome-independent mechanism. Research methodologies to investigate this function include:
Genomic binding analysis: CUT&RUN assays revealed PSME3 binds to highly active promoters in cycling myoblasts but becomes undetectable by the second day of myogenic differentiation, indicating dynamic chromatin association during differentiation .
Protein interaction analysis: Techniques such as co-immunoprecipitation followed by mass spectrometry demonstrated that PSME3 interacts with the HSP90 co-chaperone NUDC, through which it may regulate cell migration rates, levels of cell adhesion-related proteins, and ultimately myogenesis .
Functional assays: PSME3 depletion studies showed reduced efficiency of differentiation, likely related to alterations in cell migration. This can be assessed through:
Cell migration assays
Monitoring of cell adhesion-related protein levels
Visualization of myotube formation
Proteasome dependence evaluation: Using proteasome inhibitors or PSME3 mutants incapable of proteasome interaction revealed that PSME3's role in differentiation occurs in a proteasome-independent manner .
The experimental evidence indicates that PSME3 likely influences myogenesis through regulation of cytoskeletal reorganization and cell mobility rather than through transcriptional regulation, as RNA sequencing showed little change in gene expression upon PSME3 depletion .
PSME3 regulates cell migration through interaction with the HSP90 co-chaperone NUDC and by maintaining levels of cell migration-related proteins. This relationship can be experimentally validated through several approaches:
Protein-protein interaction studies: Co-immunoprecipitation experiments followed by western blotting or mass spectrometry can confirm direct interaction between PSME3 and NUDC .
PSME3 depletion studies: Knockdown or knockout of PSME3 using siRNA, shRNA, or CRISPR-Cas9 techniques followed by:
Wound healing assays to measure migration rates
Time-lapse microscopy to track cell movement
Transwell migration assays to quantify migratory capacity
Protein level analysis: Western blotting to monitor levels of migration-related proteins in PSME3-deficient versus control cells .
Research has shown that PSME3 depletion in C2C12 cells results in increased rates of cell migration. This is significant because proper balance of migration regulators is essential for myogenesis, suggesting that the migration alterations are functionally related to the reduced efficiency of differentiation observed in PSME3-depleted cells .
PSME3 is frequently overexpressed in various cancer cell lines and associated with accelerated cell division, metastatic potential, and reduced rates of apoptosis . Research methodologies to investigate its oncogenic mechanisms include:
Expression analysis: Quantitative RT-PCR, western blotting, and immunohistochemistry to compare PSME3 levels between normal and cancerous tissues or cell lines .
Functional studies:
Knockdown/overexpression studies to assess effects on proliferation, migration, and invasion
Cell cycle analysis using flow cytometry
Apoptosis assays to evaluate resistance to cell death
Signaling pathway analysis: Western blotting and reporter assays to investigate how PSME3 affects cancer-related signaling pathways, including:
In vivo studies: Xenograft models using PSME3-modulated cancer cells to assess tumor growth, invasiveness, and metastatic potential.
Published studies have demonstrated that REG γ (PSME3) knockdown suppresses proliferation by inducing apoptosis and cell cycle arrest in osteosarcoma, and that its oncogenic role is exerted by activating the Wnt/β-catenin signaling pathway . Additionally, PSME3 promotes pancreatic cancer growth via the c-Myc-glycolysis signaling axis .
PSME3 plays a crucial role in bone health and healing. Research has shown that suppression of PSME3 expression biases bone marrow stromal cells towards an adipogenic rather than osteogenic fate, and mice lacking PSME3 display corresponding bone-healing defects . Experimental approaches to investigate this function include:
Differentiation assays: Compare osteogenic versus adipogenic differentiation potential of bone marrow stromal cells with normal or reduced PSME3 levels through:
Alkaline phosphatase staining
Alizarin red staining for mineralization
Oil Red O staining for lipid accumulation
Analysis of lineage-specific markers by qRT-PCR
In vivo models: Generate PSME3 knockout or conditional knockout mice to study:
Bone development and homeostasis
Fracture healing processes
Microarchitecture analysis using micro-CT
Histomorphometric analysis
Signaling pathway analysis: Investigate how PSME3 regulates osteogenic versus adipogenic fate decisions through:
Western blotting for key signaling molecules
Chromatin immunoprecipitation to identify regulatory targets
RNA-seq to identify global changes in gene expression profiles
The experimental data suggests that therapeutic strategies targeting PSME3 might be relevant for bone-related disorders, particularly in contexts where the balance between osteogenesis and adipogenesis is disrupted .
PSME3 functions through both proteasome-dependent and proteasome-independent mechanisms, with only a small portion of the PSME3 population associated with the core proteasome . To experimentally distinguish between these functions, researchers can employ several sophisticated approaches:
Proteasome interaction-deficient mutants: Generate PSME3 mutants that cannot interact with the proteasome but retain other functions. Studies have shown that PSME3 can induce mitotic arrest, regulate p53 levels, and maintain global chromatin compaction even when prevented from interacting with the proteasome .
Proteasome inhibition studies: Use specific proteasome inhibitors (e.g., MG132, bortezomib) to block proteasome function while examining PSME3-dependent processes. Indirubin-3'-monoxime has been identified as a proteasome inhibitor with therapeutic application in multiple myeloma .
Protein degradation kinetics: Pulse-chase experiments to track protein turnover rates of PSME3 targets in the presence or absence of proteasome inhibitors.
Structure-function analysis: Create domain-specific mutants to identify regions of PSME3 responsible for proteasome-dependent versus independent functions.
Comparative proteomics: Use stable isotope labeling by amino acids in cell culture (SILAC) followed by mass spectrometry to identify proteins whose levels are affected by PSME3 in a proteasome-dependent or independent manner.
Research has demonstrated that PSME3 facilitates myoblast differentiation in a proteasome-independent manner, highlighting the importance of distinguishing between these dual functionalities when studying PSME3-related processes .
Despite PSME3's association with chromatin, its precise role in transcriptional regulation appears complex and potentially context-dependent. Advanced methodologies to investigate this interaction include:
Genome-wide binding studies: CUT&RUN and ChIP-seq analyses have revealed that PSME3 associates extensively with chromatin at over 5,000 distinct regions in cycling myoblasts, with peaks co-positive for the active promoter mark H3K4me3 . PSME3 shows a stronger preference for promoter-proximal regions than H3K4me3, primarily binding within 1kb of transcription start sites .
Protein-protein interaction network analysis: PSME3 interacts with RPRD1A, a regulator of RNA polymerase II, suggesting a potential mechanism for its chromatin association . Techniques to explore this include:
Proximity labeling (BioID, APEX) to identify chromatin-associated interaction partners
Sequential ChIP to identify co-occupancy with other factors
High-resolution imaging to visualize nuclear localization patterns
Transcriptomic analysis across differentiation: RNA-seq of PSME3-deficient cells at multiple stages of differentiation (cycling, confluent pre-differentiation, and differentiated cells) revealed no global changes in gene expression . This suggests that:
PSME3 may be primed to perform functions not engaged during normal differentiation
PSME3 might respond to specific cellular stresses rather than regulate basal transcription
PSME3's role may be post-transcriptional rather than directly affecting mRNA production
Stress response studies: Investigate PSME3's role under conditions such as:
The apparent paradox that PSME3 occupies highly active promoter regions despite having no apparent role in their regulation under normal conditions suggests sophisticated regulatory mechanisms that might be activated only under specific cellular contexts .
PSME3 interacts with components of the cellular chaperone system, notably the HSP90 co-chaperone NUDC, which mediates cargo transfer between HSP70 and HSP90 . The methodological investigation of this interaction and its significance requires sophisticated approaches:
Protein-protein interaction mapping:
Co-immunoprecipitation followed by mass spectrometry
Proximity labeling techniques (BioID, APEX)
Fluorescence resonance energy transfer (FRET) to detect direct interactions
Yeast two-hybrid screening to identify specific interaction domains
Functional analysis of the PSME3-NUDC axis:
NUDC knockdown/knockout studies to assess effects on PSME3-dependent functions
Double depletion experiments to identify epistatic relationships
Rescue experiments with NUDC mutants to identify critical domains
Chaperone-mediated protein quality control assessment:
Temperature-dependent studies: Research has shown that cold temperature extends longevity and prevents disease-related protein aggregation through PA28γ (PSME3)-induced proteasomes . Methodologies include:
Exposure of cells/organisms to different temperature conditions
Measurement of proteasome activity and protein aggregation
Analysis of lifespan and disease markers in model organisms
The interaction between PSME3 and the chaperone system represents an important intersection between protein quality control pathways and may explain PSME3's diverse cellular functions, particularly under stress conditions .
Single-cell technologies offer unprecedented opportunities to understand PSME3 function in heterogeneous cell populations during development, differentiation, and disease progression. Methodological approaches include:
Single-cell RNA-seq (scRNA-seq):
Profile PSME3 expression across different cell types and states
Identify cell populations where PSME3 is differentially expressed
Correlate PSME3 expression with cell-type specific gene signatures
Single-cell ATAC-seq (scATAC-seq):
Map chromatin accessibility in PSME3-high versus PSME3-low cells
Identify regulatory elements potentially influenced by PSME3
Integrate with scRNA-seq data to correlate accessibility with expression
Single-cell CUT&Tag or CUT&RUN:
Map PSME3 binding at single-cell resolution
Identify cell-type specific binding patterns
Correlate binding with cellular states during differentiation
Spatial transcriptomics:
Map PSME3 expression in tissue contexts
Correlate expression with cellular microenvironments
Identify spatial relationships between PSME3-expressing cells
Given PSME3's role in differentiation of myoblasts and T cells, single-cell approaches could reveal how PSME3 functions in subpopulations during lineage commitment and terminal differentiation processes .
PSME3's involvement in multiple cellular processes, including cancer progression, differentiation, and stress response, makes it a potential therapeutic target. Methodological approaches for preclinical validation include:
Cancer models:
Patient-derived xenografts from cancers with PSME3 overexpression
Genetically engineered mouse models with tissue-specific PSME3 alterations
Cell line panels representing different cancer types to assess specificity
Drug discovery approaches:
High-throughput screening for PSME3 inhibitors
Structure-based drug design targeting PSME3's proteasome-binding site
Peptide inhibitors disrupting specific protein-protein interactions
Combination therapy assessment:
Testing PSME3 inhibitors with proteasome inhibitors
Evaluating synergy with DNA-damaging agents, given PSME3's role in DNA damage response
Combining with HSP90 inhibitors to disrupt the PSME3-NUDC-HSP90 axis
Disease-specific models:
Recent research showing that PSME3 (PA28γ)-induced proteasomes can prevent disease-related protein aggregation suggests potential applications beyond cancer, particularly in neurodegenerative diseases characterized by protein aggregation .
The 26S proteasome is a multicatalytic proteinase complex with a highly ordered structure composed of two complexes: a 20S core and a 19S regulator . The 20S core is made up of four rings of 28 non-identical subunits, with two rings composed of seven alpha subunits and two rings composed of seven beta subunits . The 19S regulator consists of a base, which contains six ATPase subunits and two non-ATPase subunits, and a lid, which contains up to ten non-ATPase subunits .
PSME3 is a subunit of the 11S regulator, which is an alternate regulator of the proteasome . The 11S regulator, also known as PA28, replaces the 19S regulator in the immunoproteasome, a modified form of the proteasome that processes class I MHC peptides . The gamma subunit of the 11S regulator, encoded by the PSME3 gene, forms a homohexameric ring composed of six gamma subunits .
PSME3 plays a significant role in various cellular processes, including the regulation of the cell cycle, apoptosis, and the immune response . It is involved in the degradation of proteins that are ubiquitinated, a process that tags proteins for degradation by the proteasome . The immunoproteasome, which includes the 11S regulator, is essential for the processing of class I MHC peptides, which are crucial for the immune response .
Recombinant PSME3 is used in various research applications to study its structure, function, and role in disease processes . Understanding the mechanisms by which PSME3 regulates the proteasome and its involvement in cellular processes can provide insights into potential therapeutic targets for diseases associated with proteasome dysfunction .