PSMD5 Human

Proteasome 26S Subunit, Non-ATPase 5 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Assembly

PSMD5 (UniProt ID: Q92530) is a 504-amino acid protein encoded by the PSMD5 gene located on chromosome 9p13.3. It functions as a non-ATPase subunit of the 19S regulatory particle (RP) within the 26S proteasome complex, which comprises a 20S catalytic core and a 19S regulatory cap .

Biological Functions

PSMD5 plays a pivotal role in maintaining proteostasis through:

  1. Proteasome Assembly: Facilitates the correct folding and integration of 19S subunits into the 26S proteasome .

  2. Protein Quality Control: Mediates the degradation of ubiquitinated proteins, including cell cycle regulators (e.g., p53, NF-κB) and oncoproteins .

  3. Disease Pathways: Dysregulation contributes to neurodegenerative diseases, cardiovascular disorders, and cancer .

Table 1: PSMD5 Mutations and Associated Phenotypes

MutationModel SystemPhenotypeSource
CRISPR-inducedZebrafishOcular coloboma, vertebral defects
Cys52PheComputational MDReduced bortezomib binding affinity
  • Cancer:

    • PSMD5 suppression via promoter methylation is linked to drug resistance in neuroblastoma, ovarian cancer, and multiple myeloma .

    • Low PSMD5 expression correlates with poor prognosis in bortezomib-treated myeloma patients (hazard ratio = 1.8, P = 0.004) .

  • Neurodegeneration: Accumulation of misfolded proteins due to proteasome dysfunction is implicated in Alzheimer’s and Parkinson’s diseases .

Table 2: PSMD5 Expression and Drug Response in Cancer

Cancer TypePSMD5 StatusDrug SensitivityOutcome
NeuroblastomaSuppressed (IMR32)Bortezomib-resistantReduced 26S assembly
Multiple MyelomaLow expressionShorter time to relapsePoor survival
  • Therapeutic Strategies:

    • Re-expression of PSMD5 in tumor cells restores proteasome inhibitor sensitivity .

    • Targeting PSMD5 methylation may reverse chemoresistance in gliomas and bladder carcinomas .

Recombinant PSMD5 and Experimental Applications

Recombinant human PSMD5 (e.g., ab207131) is produced in E. coli with >90% purity and is used to study proteasome assembly mechanisms . Applications include:

  • In vitro chaperone assays to map 19S assembly intermediates .

  • Structural studies using cryo-EM and X-ray crystallography .

Product Specs

Introduction
Proteasome 26S Subunit, Non-ATPase 5, also known as PSMD5, is a component of the proteasome subunit S5B/HSM3 family. This protein plays a role in the 26S proteasome, a complex responsible for degrading ubiquitinated proteins within eukaryotic cells. PSMD5 functions as a chaperone protein, contributing to the assembly of the 26s proteasome, specifically the base subcomplex of the PA700/19S regulatory complex. It is characterized by numerous dileucine repeats, which are believed to be involved in the trafficking of various transmembrane proteins.
Description
Recombinant human PSMD5, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 529 amino acids (specifically, residues 1-504). With a molecular weight of 58.9 kDa, this protein is fused to a 25 amino acid His-tag at its N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PSMD5 protein solution is provided at a concentration of 1 mg/ml and is prepared in a buffer consisting of Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. To maintain product integrity, avoid repeated cycles of freezing and thawing.
Purity
The purity of the PSMD5 protein is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
PSMD5, Proteasome (Prosome, Macropain) 26S Subunit, Non-ATPase, 526S Protease Subunit S5 Basic,26S Proteasome Subunit S5B,S5B,26S Proteasome Non-ATPase Regulatory Subunit 5,KIAA0072.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFMAAQA LALLREVARL EAPLEELRAL HSVLQAVPLN ELRQQAAELR LGPLFSLLNE NHREKTTLCV SILERLLQAM EPVHVARNLR VDLQRGLIHP DDSVKILTLS QIGRIVENSD AVTEILNNAE LLKQIVYCIG GENLSVAKAA IKSLSRISLT QAGLEALFES NLLDDLKSVM KTNDIVRYRV YELIIEISSV SPESLNYCTT SGLVTQLLRE LTGEDVLVRA TCIEMVTSLA YTHHGRQYLA QEGVIDQISN IIVGADSDPF SSFYLPGFVK FFGNLAVMDS PQQICERYPI FVEKVFEMIE SQDPTMIGVA VDTVGILGSN VEGKQVLQKT GTRFERLLMR IGHQSKNAPV ELKIRCLDAI SSLLYLPPEQ QTDDLLRMTE SWFSSLSRDP LELFRGISSQ PFPELHCAAL KVFTAIANQP WAQKLMFNSP GFVEYVVDRS VEHDKASKDA KYELVKALAN SKTIAEIFGN PNYLRLRTYL SEGPYYVKPV STTAVEGAE.

Q&A

What is PSMD5 and what is its basic function in human cells?

PSMD5 (also known as 26S protease subunit S5 basic) is a non-ATPase subunit of the 19S regulator base of the 26S proteasome. The 26S proteasome is a multicatalytic proteinase complex composed of a 20S core and a 19S regulator, which together function as the primary machinery for non-lysosomal protein degradation in eukaryotic cells . PSMD5 specifically acts as a key orchestrator of proteasome assembly, functioning primarily as an inhibitor of 26S proteasome assembly under normal conditions . The protein is encoded by the PSMD5 gene in humans and has a molecular weight of approximately 58.9 kDa, consisting of 504 amino acids in its mature form . As part of the ubiquitin-proteasome system (UPS), PSMD5 contributes to protein quality control mechanisms that regulate numerous cellular processes including cell cycle progression, differentiation, signal transduction, and apoptosis .

What is the structure of the 26S proteasome and how does PSMD5 fit within this structure?

The 26S proteasome has a highly ordered structure composed of two major subcomplexes:

  • 20S core particle: Consists of 4 stacked rings forming a barrel-like structure with 28 non-identical subunits. The outer rings contain 7 alpha subunits each, while the inner rings contain 7 beta subunits each. The catalytic activities reside within the beta subunits .

  • 19S regulatory particle: Composed of a "base" and a "lid." The base contains 6 ATPase subunits and 2 non-ATPase subunits, while the lid contains up to 10 non-ATPase subunits .

PSMD5 functions as a non-ATPase subunit of the 19S regulator base. Rather than being a constitutive component of the assembled proteasome, PSMD5 serves as a chaperone or regulator during proteasome assembly. It interacts with proteasome components during the assembly process but is not typically found in the final assembled and functional 26S proteasome . This regulatory role allows PSMD5 to control the rate and efficiency of 26S proteasome formation, which becomes particularly significant in cancer contexts where proteasome capacity is often altered.

How is PSMD5 expression regulated in normal human tissues?

Under normal physiological conditions, PSMD5 expression is maintained at levels that allow for appropriate proteasome assembly regulation. In healthy cells and tissues, including peripheral blood mononuclear cells and CD138+ plasma cells from healthy donors, PSMD5 promoter regions are generally unmethylated, allowing for normal gene expression .

Regulation of PSMD5 occurs at multiple levels:

Normal PSMD5 expression is crucial for maintaining appropriate proteasome assembly rates and ensuring cellular protein homeostasis through regulated protein degradation pathways.

What molecular mechanisms explain PSMD5's role in 26S proteasome assembly inhibition?

PSMD5 functions as a key negative regulator of 26S proteasome assembly through several molecular mechanisms:

  • Chaperone-like activity: PSMD5 acts as a chaperone during the early stages of 19S regulatory particle assembly, potentially sequestering certain subunits and controlling the rate of assembly.

  • Competitive binding: Research suggests that PSMD5 may competitively bind to sites that would otherwise facilitate interaction between the 19S and 20S particles, thereby inhibiting complete 26S proteasome formation.

  • Assembly checkpoint function: PSMD5 may serve as a quality control checkpoint, ensuring that only properly formed subunits proceed to full assembly.

When PSMD5 is experimentally re-expressed in tumor cells (where it is often suppressed), it causes decreased 26S proteasome assembly and subsequent accumulation of polyubiquitinated proteins . This suggests that PSMD5 directly interferes with the assembly process, resulting in reduced proteasome capacity and function.

The inhibitory role of PSMD5 in proteasome assembly serves as a regulatory mechanism to control proteasome abundance and activity in response to cellular needs. Under normal conditions, this regulation helps maintain proteostasis, but cancer cells appear to gain advantage by suppressing PSMD5 to enhance proteasome capacity.

How does PSMD5 suppression contribute to cancer progression and proteasome inhibitor resistance?

PSMD5 suppression appears to be a strategic adaptation in cancer cells that contributes to both cancer progression and therapeutic resistance through multiple mechanisms:

  • Enhanced 26S proteasome assembly: Upon tumorigenic transformation of epithelial cells, 26S proteasome assembly is significantly enhanced, despite unchanged levels of individual subunits. This enhancement increases with tumor progression and is observed specifically in transformed cells . PSMD5 suppression facilitates this increased assembly rate.

  • Handling of proteotoxic stress: Cancer cells face considerable protein overload due to high metabolic rates, reprogrammed energy metabolism, and often aneuploidy. Enhanced proteasome assembly enables cancer cells to cope with this increased proteotoxic stress.

  • Resistance to proteasome inhibitors: In proteasome inhibitor-refractory multiple myeloma patients, PSMD5 promoter hypermethylation and subsequent epigenetic gene silencing were identified in approximately 24% of cases. This silencing correlates with increased resistance to proteasome inhibitors such as bortezomib .

  • Altered proteolytic capacity: Downregulation of PSMD5 increases the cell's proteolytic capacity, allowing cancer cells to better manage protein turnover requirements associated with rapid proliferation and altered metabolism .

  • Broad transcriptome remodeling: Proteasome subunit suppression, including PSMD5, serves as a sentinel of more global remodeling of the transcriptome, producing a distinct gene signature that supports cancer progression .

Research has identified that PSMD5 is the most commonly suppressed 19S subunit gene in human tumor resection samples , suggesting an important role in natural tumor evolution rather than merely a response to therapeutic pressure.

What is the relationship between PSMD5 expression and clinical outcomes in cancer patients?

Analysis of clinical data reveals significant correlations between PSMD5 expression patterns and patient outcomes:

  • Poor prognosis in multiple myeloma: Suppression of PSMD5 and other 19S proteasome subunits is associated with poor outcomes in myeloma patients, where proteasome inhibitors are a mainstay of treatment . This association likely stems from the reduced efficacy of proteasome inhibitor therapy in patients with PSMD5 suppression.

  • Resistance biomarker: PSMD5 promoter hypermethylation appears in approximately 24% of proteasome inhibitor-refractory multiple myeloma patients but is generally not observed in newly diagnosed patients . This suggests that PSMD5 silencing may evolve as an adaptive response to proteasome inhibitor therapy.

  • Potential predictive marker: The methylation status of the PSMD5 promoter could potentially serve as a predictive biomarker for proteasome inhibitor therapy response in multiple myeloma. Patients with hypermethylated PSMD5 promoters might benefit from alternative or combination therapies.

  • Association with heritably altered cancer states: PSMD5 suppression is part of a naturally arising imbalance in 19S regulatory complex composition that marks the emergence of a heritably altered and therapeutically relevant state in diverse cancers . This suggests PSMD5 expression patterns may have broader prognostic relevance beyond just predicting proteasome inhibitor response.

These clinical correlations highlight the potential utility of PSMD5 as both a prognostic indicator and a predictive biomarker for guiding therapeutic decisions in cancer treatment.

What are the most effective techniques for measuring PSMD5 expression and methylation status?

Several complementary techniques can be employed to comprehensively evaluate PSMD5 expression and methylation status:

For PSMD5 Expression Analysis:

  • Western Blotting: Standard protein detection method using PSMD5-specific antibodies. Particularly useful for detecting protein levels in cell lines, patient samples, or xenograft tissues.

  • Quantitative RT-PCR (qRT-PCR): For measuring PSMD5 mRNA expression levels. Primers should be designed to specifically amplify PSMD5 transcripts.

  • RNA-Seq: Provides comprehensive transcriptome profiling that can place PSMD5 expression in the context of global gene expression patterns.

  • Immunohistochemistry (IHC): Allows visualization of PSMD5 protein in tissue sections, providing spatial information about expression patterns.

For PSMD5 Methylation Analysis:

  • Deep Bisulfite Sequencing: The gold standard for DNA methylation profiling, as used in the identified research to detect PSMD5 promoter hypermethylation in PI-refractory patients .

  • Methylation-Specific PCR (MSP): A targeted approach to assess the methylation status of specific CpG sites in the PSMD5 promoter.

  • Pyrosequencing: Provides quantitative methylation data for specific genomic regions.

  • Dual-Luciferase Reporter Assay: Used to functionally validate the impact of methylation on PSMD5 promoter activity, as demonstrated in the research where the regulatory impact of this region was confirmed .

Functional Validation:

To establish causality between PSMD5 expression/methylation and phenotypic outcomes, researchers have employed:

  • Lentiviral Gene Transfer: For re-expressing PSMD5 in tumor cells with suppressed endogenous expression .

  • Demethylating Agents: Cell lines such as KMS11 serve as models to test the impact of demethylating agents on PSMD5 expression and proteasome function .

  • Native Gel Electrophoresis: Useful for analyzing intact 26S proteasome complexes and assessing assembly status in relation to PSMD5 manipulation .

These methodological approaches should be selected based on the specific research question and available sample materials.

How can researchers experimentally manipulate PSMD5 expression to study its function?

Researchers can employ several strategies to manipulate PSMD5 expression for functional studies:

For PSMD5 Overexpression:

  • Lentiviral/Retroviral Expression Systems: Stable integration of PSMD5 cDNA under constitutive or inducible promoters. This approach has been successfully used to re-express PSMD5 in tumor cells, resulting in decreased 26S proteasome assembly and accumulation of polyubiquitinated proteins .

  • Transient Transfection: For short-term PSMD5 overexpression studies, plasmid-based transient transfection can be employed using lipid-based or electroporation methods.

  • Recombinant Protein: Purified recombinant PSMD5 protein (such as the commercially available form described in the search results) can be used for biochemical studies or potentially for cell permeabilization experiments .

For PSMD5 Knockdown/Knockout:

  • siRNA/shRNA: RNA interference techniques can be used for transient or stable knockdown of PSMD5 expression.

  • CRISPR-Cas9 Gene Editing: For generating complete PSMD5 knockout cell lines or animal models.

For Manipulating PSMD5 Methylation:

  • Demethylating Agents: Compounds like 5-azacytidine or decitabine can be used to reverse PSMD5 promoter hypermethylation, as demonstrated in the KMS11 cell line model .

  • Epigenetic Editing: Advanced techniques using CRISPR-dCas9 fused to DNA methyltransferases or demethylases can be employed for site-specific manipulation of PSMD5 promoter methylation.

Experimental Models:

  • Cancer Cell Lines: Established cancer cell lines with varying levels of PSMD5 expression serve as important models.

  • Organoids: As described in the research, advanced tumor organoids can be used for PSMD5 functional studies. The methodology involves dissociating organoids, incubating with viral particles in liquid media, re-embedding in Matrigel, and selection with puromycin .

  • In vivo Models: Mouse models of intestinal cancer have been used to study PSMD5 expression changes during tumorigenesis .

These experimental approaches allow researchers to establish causative relationships between PSMD5 expression levels and functional outcomes related to proteasome assembly, protein homeostasis, and cancer biology.

What assays can effectively measure 26S proteasome assembly and how can PSMD5's impact be quantified?

Several specialized assays can be employed to measure 26S proteasome assembly and quantify PSMD5's regulatory impact:

Proteasome Assembly Assays:

  • Native Gel Electrophoresis: This technique separates intact protein complexes based on size and charge, allowing visualization of assembled 26S proteasomes, 20S core particles, and 19S regulatory particles. Following electrophoresis, proteasome complexes can be detected using activity-based probes or western blotting with proteasome subunit-specific antibodies .

  • Glycerol Gradient Centrifugation: Separates protein complexes based on size and density, allowing isolation and quantification of 26S proteasomes versus free 20S and 19S particles.

  • Fluorescence Correlation Spectroscopy (FCS): Can be used with fluorescently tagged proteasome subunits to monitor assembly dynamics in real-time.

  • Proteasome Activity Assays: Fluorogenic peptide substrates specific for the different catalytic activities of the proteasome (chymotrypsin-like, trypsin-like, and caspase-like) can indirectly measure functional 26S proteasome assembly.

Quantifying PSMD5's Impact:

These assays, particularly when used in combination, provide a comprehensive assessment of how PSMD5 influences proteasome assembly and subsequent cellular proteostasis.

How does PSMD5 methylation status evolve during cancer progression and treatment?

The evolution of PSMD5 methylation status during cancer progression and treatment reveals important insights into tumor adaptation mechanisms:

Methylation Changes During Cancer Progression:

  • Initial Tumorigenesis: In early transformation stages, PSMD5 expression is often reduced compared to normal tissue, but the mechanisms may be varied and not necessarily methylation-driven .

  • Progressive Silencing: As tumors progress, PSMD5 expression is further reduced through increasing promoter methylation. This represents a selective advantage for cancer cells by enhancing proteasome assembly capacity .

  • Metastatic Disease: Advanced research suggests that PSMD5 silencing may be more prevalent in aggressive or metastatic disease, though the search results don't provide specific data on this progression.

Methylation Changes During Treatment:

  • Treatment-Naive State: Patients with newly diagnosed multiple myeloma generally show unmethylated PSMD5 promoter profiles, similar to normal cells .

  • Therapy-Induced Selection: Under the selective pressure of proteasome inhibitor treatment, multiple myeloma cells acquire methylation of the PSMD5 promoter, silencing PSMD5 gene expression . This represents an adaptive response to therapy.

  • Resistant Disease: In proteasome inhibitor-refractory patients, approximately 24% show PSMD5 promoter hypermethylation, suggesting this as a clinically significant resistance mechanism .

The temporal dynamics of PSMD5 methylation changes suggest an evolutionary process where cancer cells progressively silence PSMD5 to enhance proteasome capacity, particularly when challenged with proteasome inhibitor therapy. This epigenetic adaptation appears to be more prevalent in response to therapeutic pressure rather than occurring spontaneously in treatment-naive disease, highlighting the importance of considering PSMD5 methylation in treatment planning and resistance monitoring.

What is the relationship between PSMD5 expression and other 19S proteasome subunits in cancer?

The relationship between PSMD5 and other 19S proteasome subunits in cancer reveals coordinated regulatory patterns and unique features of PSMD5:

  • Differential Suppression Patterns: Among 19S proteasome subunits, PSMD5 stands out as the most commonly suppressed subunit gene in human tumor resection samples and across diverse cancer cell line datasets including GDSC and CCLE . This suggests a potentially unique or primary role for PSMD5 suppression in cancer biology.

  • Coordinated Regulation: While PSMD5 is often the most commonly suppressed, multiple other 19S subunits can also show reduced expression in cancers. This suggests a coordinated regulation of the 19S regulatory particle composition as part of a broader adaptation strategy .

  • Imbalanced Subunit Expression: Cancer cells frequently display imbalances in the expression of various proteasome subunits. This imbalance - particularly between 19S regulatory and 20S core components - appears to be a feature of many cancers and contributes to altered proteasome function .

  • Selective Methylation: While several proteasome subunits including PSMB5, PSMC2, PSMC5, PSMC6, PSMD1, and PSMD5 have been associated with proteasome inhibitor resistance, DNA methylation profiling revealed that PSMD5 was particularly subject to epigenetic silencing through promoter hypermethylation . This suggests different mechanisms may regulate various subunits.

  • Effect on Assembly vs. Function: While some proteasome subunits (like PSMB5) directly affect the catalytic function of the proteasome when mutated or suppressed, PSMD5 primarily influences the assembly process. This mechanistic distinction has important implications for how different patterns of subunit suppression affect proteasome biology and drug resistance .

This complex interplay between PSMD5 and other proteasome subunits highlights the sophisticated ways in which cancer cells can modulate the proteasome system to adapt to cellular stresses and therapeutic challenges. The preferential suppression of PSMD5 suggests it may represent a particularly effective target for modulating proteasome function in cancer cells.

What novel therapeutic strategies could target the PSMD5-associated mechanisms in cancer?

Several innovative therapeutic approaches could exploit the PSMD5-associated mechanisms in cancer:

  • Epigenetic Modifiers: Since PSMD5 silencing often occurs through promoter hypermethylation, DNA methyltransferase inhibitors (DNMTi) like 5-azacytidine or decitabine could potentially restore PSMD5 expression. This approach was functionally tested in the KMS11 cell line model . Combining DNMTi with proteasome inhibitors might reverse resistance mechanisms in multiple myeloma and other cancers.

  • PSMD5 Mimetics: Developing small molecules that mimic PSMD5's inhibitory effect on proteasome assembly could potentially recreate the natural regulatory function lost in cancer cells. These compounds would target the proteasome assembly process rather than catalytic function, potentially overcoming existing resistance mechanisms.

  • Targeted Protein Degradation: Proteolysis-targeting chimeras (PROTACs) or molecular glues could be designed to induce degradation of key proteasome assembly factors that become dysregulated when PSMD5 is suppressed.

  • Synthetic Lethality Approaches: Research revealed that PSMD5 suppression creates new vulnerabilities to certain drugs, including the proapoptotic drug ABT-263 . Screening for additional synthetic lethal interactions with PSMD5-low states could identify other therapeutic opportunities.

  • Proteasome Assembly Modulators: Compounds that specifically interfere with enhanced proteasome assembly in cancer cells (rather than targeting catalytic activity like traditional proteasome inhibitors) could represent a novel class of therapeutics with potentially improved specificity for cancer cells.

  • Combination Strategies: The distinct gene signature associated with proteasome subunit suppression suggests unique vulnerabilities that could be exploited through rationally designed combination therapies . For instance, combining proteasome assembly inhibitors with drugs targeting stress response pathways might enhance cancer cell killing.

  • Biomarker-Guided Therapy: PSMD5 promoter methylation status could serve as a biomarker to guide treatment decisions, directing the use of alternative therapies in patients likely to have proteasome inhibitor resistance. This personalized approach could improve outcomes in multiple myeloma and potentially other cancers.

These approaches represent promising directions for therapeutic development that specifically target the altered proteasome biology associated with PSMD5 suppression in cancer.

What are the key challenges in studying proteasome assembly dynamics in vivo?

Investigating proteasome assembly dynamics in living systems presents several significant technical challenges:

  • Complex and Dynamic Process: Proteasome assembly involves multiple intermediate complexes and chaperone proteins. Capturing this dynamic process in vivo requires sophisticated techniques that can monitor protein interactions in real-time without disrupting cellular functions.

  • Tissue-Specific Variations: Proteasome composition and assembly rates can vary significantly between different tissues and cell types. Studies in one system may not translate to others, necessitating tissue-specific approaches .

  • Visualizing Native Complexes: Traditional protein visualization techniques often require conditions that disrupt native protein complexes. Developing methods that preserve proteasome integrity while allowing visualization remains challenging.

  • Distinguishing Assembly vs. Disassembly: Determining whether observed changes represent enhanced assembly or reduced disassembly of proteasomes can be difficult to discern in dynamic cellular systems.

  • Cellular Heterogeneity: Within tumors, significant cell-to-cell variation can exist in proteasome assembly states. Single-cell approaches are needed but technically challenging for studying large protein complexes.

  • Quantitative Assessment: Accurate quantification of proteasome assembly rates in vivo requires standardized methods that account for various confounding factors like cell cycle stage and metabolic state.

  • Temporal Resolution: Assembly occurs over minutes to hours, requiring experimental designs that can capture appropriate time points without excessive manipulation of the biological system.

  • Animal Model Development: Developing animal models that recapitulate the PSMD5 methylation and expression patterns seen in human cancers requires sophisticated genetic engineering approaches.

Researchers have addressed some of these challenges through innovative approaches such as:

  • Using organoid cultures that better preserve the biological characteristics of original tissues compared to traditional cell lines

  • Employing the SUnSET non-radioactive labeling method to measure protein translation rates in both cultured cells and in vivo systems

  • Utilizing Native gel electrophoresis techniques optimized to preserve intact proteasome complexes

Despite these advances, comprehensive in vivo analysis of proteasome assembly dynamics remains technically challenging, particularly in the context of understanding how PSMD5 regulation affects these processes in human tumors.

How can researchers distinguish between cause and effect in PSMD5-related proteasome alterations?

Establishing causality in PSMD5-related proteasome alterations requires rigorous experimental approaches:

  • Temporal Analysis: Sequential sampling during cancer progression or treatment resistance development can help establish whether PSMD5 silencing precedes or follows other proteasome changes. This approach has revealed that in multiple myeloma, PSMD5 promoter methylation emerges specifically under proteasome inhibitor treatment pressure .

  • Genetic Manipulation Experiments:

    • Loss of Function: CRISPR-Cas9 knockout or siRNA knockdown of PSMD5 in normal cells to determine if this recapitulates the cancer phenotype.

    • Gain of Function: Re-expression of PSMD5 in cancer cells with suppressed endogenous expression has demonstrated direct causation by showing decreased 26S assembly and accumulated polyubiquitinated proteins .

  • Epigenetic Modulation: Using demethylating agents specifically on the PSMD5 promoter region (through targeted approaches) can help determine if restoring PSMD5 expression alone is sufficient to reverse proteasome assembly phenotypes.

  • Dose-Response Relationships: Establishing quantitative relationships between PSMD5 expression levels and proteasome assembly rates can help determine threshold effects and strengthen causal inferences.

  • Single-Cell Analyses: Correlating PSMD5 expression/methylation with proteasome assembly status at the single-cell level can help discriminate cause-effect relationships from coincidental associations.

  • Mathematical Modeling: Developing computational models of proteasome assembly that incorporate PSMD5 regulatory effects can generate testable predictions about causality.

  • In vivo Rescue Experiments: Demonstrating that restoring normal PSMD5 function in tumor models reverses cancer phenotypes provides strong evidence for causality.

  • External Perturbation Approaches: Examining how PSMD5 responds to various cellular stresses can help determine whether its suppression is a primary driver or secondary adaptation.

A particularly compelling experimental approach demonstrated in the research is the use of lentiviral infection to express PSMD5 in advanced tumor organoids, followed by functional assays of proteasome assembly and protein homeostasis . This type of intervention experiment provides strong evidence for PSMD5's causal role in regulating proteasome assembly in cancer.

By employing these complementary approaches, researchers can build a strong case for causal relationships between PSMD5 alterations and subsequent changes in proteasome function and cancer biology.

What are the most significant data interpretation challenges when analyzing PSMD5 expression across different cancer datasets?

Researchers face numerous challenges when analyzing PSMD5 expression data across different cancer datasets:

  • Methodology Variations: Different studies employ varying techniques for measuring gene expression (microarray, RNA-seq, qPCR) and protein levels (western blot, mass spectrometry, immunohistochemistry). These methodological differences can introduce systematic biases that complicate direct comparisons.

  • Sample Heterogeneity: Cancer samples often contain varying proportions of tumor cells, stromal cells, and immune infiltrates. PSMD5 expression in bulk tumor samples may not accurately reflect its levels specifically in cancer cells.

  • Reference Selection: The choice of reference genes or normalization methods can significantly impact the interpretation of PSMD5 expression levels, particularly when comparing across different datasets.

  • Treatment History Effects: Patient samples may come from individuals with different treatment histories, which can significantly affect PSMD5 expression and methylation patterns. As demonstrated in multiple myeloma research, PSMD5 methylation evolves under treatment pressure .

  • Threshold Definitions: Defining what constitutes "suppressed" PSMD5 expression varies between studies, making it difficult to establish consistent prevalence rates of PSMD5 suppression across cancer types.

  • Correlative vs. Causative Interpretations: Distinguishing whether altered PSMD5 expression is driving cancer phenotypes or merely correlating with them requires careful interpretation of associative data.

  • Transcript vs. Protein Discordance: PSMD5 mRNA levels may not directly correlate with protein abundance due to post-transcriptional regulation, making it essential to consider both when available.

  • Isoform Considerations: Potential alternative splicing or protein isoforms of PSMD5 might complicate interpretation of expression data that doesn't discriminate between variants.

  • Cancer Subtype Stratification: Different molecular subtypes within a cancer type may show distinct patterns of PSMD5 regulation, requiring careful stratification during analysis.

To address these challenges, researchers analyzing PSMD5 across cancer datasets should:

  • Employ consistent methodologies or computational corrections for batch effects

  • Include cellular deconvolution approaches for bulk tumor data

  • Incorporate multimodal data (genomic, transcriptomic, epigenomic, proteomic) when available

  • Carefully document patient treatment histories

  • Establish biologically meaningful thresholds for PSMD5 suppression

  • Validate key findings across independent datasets

  • Combine computational analyses with experimental validation

These considerations are particularly important when analyzing PSMD5 as a potential biomarker for proteasome inhibitor response or as part of a broader gene signature associated with cancer progression.

Table 1: PSMD5 Characteristics and Functions

FeatureDescriptionReference
Official Name26S proteasome non-ATPase regulatory subunit 5
SynonymsKIAA0072, 26S Protease Subunit S5 Basic
Gene IDEntrez: 5712, UniProt: Q16401
Molecular Weight58.9 kDa (529 amino acids)
Cellular LocalizationCytoplasmic, associated with proteasome components
Primary FunctionInhibitor of 26S proteasome assembly
Role in CancerMost commonly suppressed 19S subunit in tumors
Methylation in DiseaseHypermethylated in 24% of PI-refractory MM patients
Effect When Re-expressedDecreased 26S assembly, accumulated polyubiquitinated proteins

Table 2: Comparison of PSMD5 Methylation Status Across Different Clinical Contexts

Clinical ContextPSMD5 Promoter Methylation StatusExpression LevelClinical SignificanceReference
Healthy Donors (PBMCs, CD138+ cells)UnmethylatedNormalBaseline physiological state
Newly Diagnosed Multiple MyelomaGenerally unmethylatedNormal to slightly reducedTreatment-naive disease state
PI-Refractory Multiple MyelomaHypermethylated in ~24% of casesReducedAssociated with treatment resistance
Advanced Solid TumorsVariable methylationOften suppressedMost commonly suppressed 19S subunit
Transformed Intestinal EpitheliumReduced expressionReducedEnhances 26S proteasome assembly

Product Science Overview

Introduction

Proteasome 26S Subunit, Non-ATPase 5 (PSMD5) is a component of the 26S proteasome, a complex and essential protease machinery in eukaryotic cells. The 26S proteasome is responsible for the ATP-dependent degradation of ubiquitinated proteins, playing a crucial role in maintaining cellular homeostasis by removing misfolded, damaged, or unneeded proteins .

Structure and Function

The 26S proteasome is a large, multi-subunit complex composed of a 20S core particle and a 19S regulatory particle. PSMD5 is one of the non-ATPase subunits of the 19S regulatory particle. This subunit is involved in recognizing and binding ubiquitinated substrates, facilitating their translocation into the 20S core for degradation .

Biological Significance

PSMD5, along with other subunits of the 26S proteasome, is essential for various cellular processes, including cell cycle regulation, signal transduction, and stress responses. The degradation of ubiquitinated proteins by the proteasome is a highly regulated process, ensuring that only proteins tagged for destruction are targeted, thus maintaining protein homeostasis .

Research and Applications

Recombinant human PSMD5 is used in research to study the structure and function of the 26S proteasome. Advances in cryogenic electron microscopy (cryo-EM) have provided detailed insights into the dynamic organization and functional mechanisms of the proteasome, including the role of PSMD5 in substrate recognition and processing .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.