RPN2 Human

Ribophorin II Human Recombinant
Shipped with Ice Packs
In Stock

Description

RPN2 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 539 amino acids (23-540) and having a molecular mass of 59.2kDa.
RPN2 is fused to a 21 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Ribophorin 2 (RPN2) is a subunit of the dolichyl-diphosphooligosaccharide protein glycosyltransferase complex. This complex is responsible for attaching high mannose oligosaccharides to asparagine residues within the Asn-X-Ser/Thr consensus motif of newly formed polypeptide chains. RPN2 shares sequence similarity with the yeast oligosaccharyl transferase subunit SWP1.
Description
Recombinant human RPN2, expressed in E. coli, is a single polypeptide chain lacking glycosylation. It encompasses 539 amino acids (residues 23-540) and has a molecular weight of 59.2 kDa. The protein includes a 21 amino acid His-tag fused at its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The RPN2 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.1 M NaCl.
Stability
For short-term storage (2-4 weeks), the product can be kept at 4°C. For extended storage, freezing at -20°C is recommended. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advisable. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 90.0% as assessed by SDS-PAGE.
Synonyms
Dolichyl-diphosphooligosaccharide--protein glycosyltransferase subunit 2, Dolichyl-diphosphooligosaccharide--protein glycosyltransferase 63 kDa subunit, RIBIIR, Ribophorin II, RPN-II, Ribophorin-2, RPN2, SWP1, RPNII.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MLTPTHYLTK HDVERLKASL DRPFTNLESA FYSIVGLSSL GAQVPDAKKA CTYIRSNLDP SNVDSLFYAA QASQALSGCE ISISNETKDL LLAAVSEDSS VTQIYHAVAA LSGFGLPLAS QEALSALTAR LSKEETVLAT VQALQTASHL SQQADLRSIV
EEIEDLVARL DELGGVYLQF EEGLETTALF VAATYKLMDH VGTEPSIKED QVIQLMNAIF SKKNFESLSE AFSVASAAAV LSHNRYHVPV VVVPEGSASD THEQAILRLQ VTNVLSQPLT QATVKLEHAK SVASRATVLQ KTSFTPVGDV FELNFMNVKF SSGYYDFLVE VEGDNRYIAN
TVELRVKIST EVGITNVDLS TVDKDQSIAP KTTRVTYPAK AKGTFIADSH QNFALFFQLV DVNTGAELTP HQTFVRLHNQ KTGQEVVFVA EPDNKNVYKF ELDTSERKIE FDSASGTYTL YLIIGDATLK NPILWNVADV VIKFPEEEAP STVLSQNLFT PKQEIQHLFR EPEKRPPTV.

Q&A

What is the basic structure and function of RPN2 in normal human cells?

RPN2 (Ribophorin II) is a type I integral membrane glycoprotein found exclusively in the rough endoplasmic reticulum. The human RPN2 gene is located on chromosome 20q11.23 and consists of 19 exons encoding a protein of 631 amino acid residues with a molecular weight of approximately 69,284 Da . RPN2 functions as an essential component of the N-oligosaccharyl transferase (OST) complex that catalyzes the transfer of high-mannose oligosaccharides to asparagine residues within the Asn-X-Ser/Thr consensus motif of nascent polypeptide chains during protein synthesis . This glycosylation process is critical for proper protein folding, stability, and function. RPN2 plays a crucial role in maintaining the structural uniqueness of the rough endoplasmic reticulum and participates in the translocation of nascent proteins . The protein has been highly conserved throughout evolution, suggesting its fundamental importance in cellular processes.

What experimental techniques are most reliable for measuring RPN2 expression?

Multiple complementary techniques should be employed for comprehensive RPN2 expression analysis:

For mRNA quantification:

  • Quantitative Real-Time PCR (qRT-PCR): This technique requires RNA extraction using reagents like Trizol, followed by cDNA synthesis with reverse transcription kits (e.g., Takara), and amplification with SYBR-Green reagent and RPN2-specific primers . Expression is typically normalized using the 2^-ΔΔCt method with housekeeping genes as internal controls.

  • RNA-Seq: For genome-wide expression analysis with absolute quantification capabilities.

For protein detection:

  • Western blotting: Provides semi-quantitative measurement of RPN2 protein levels in cell or tissue lysates. Researchers should optimize antibody concentrations and ensure proper loading controls .

  • Immunohistochemistry (IHC): Allows visualization of RPN2 expression in tissue sections. RPN2 protein is primarily localized in the cytoplasm of cells, and expression can be quantified using scoring systems such as H-scores .

When analyzing clinical samples, researchers should include paired normal tissues as controls whenever possible. Statistical analysis typically involves paired Student's t-tests for comparing two groups or ANOVA for multiple comparisons, with p<0.05 considered statistically significant .

How do researchers effectively modulate RPN2 expression in experimental models?

Several methodological approaches can be employed to modify RPN2 expression for functional studies:

For downregulation:

  • RNA interference (RNAi): Both small interfering RNA (siRNA) for transient knockdown and short hairpin RNA (shRNA) delivered via lentiviral vectors for stable knockdown have been successfully used to reduce RPN2 expression in bladder cancer and laryngeal cancer cell lines .

  • CRISPR-Cas9 gene editing: For complete knockout of RPN2, though this approach must be used cautiously as complete loss may affect cell viability in some contexts.

For overexpression:

  • Plasmid-based overexpression: Using vectors containing the RPN2 cDNA under control of appropriate promoters, as demonstrated in studies with laryngeal cancer cell lines .

Validation considerations:

  • Confirmation of knockdown/overexpression efficiency at both mRNA level (qRT-PCR) and protein level (Western blot)

  • Inclusion of appropriate controls (scrambled siRNA, empty vector)

  • Cell line-specific optimization of transfection conditions

  • Assessment of potential off-target effects

In studies examining RPN2 in bladder cancer, researchers successfully employed shRNA to silence RPN2 expression in 5637 and T24 cell lines, confirming knockdown efficiency before proceeding with functional assays . Similarly, in laryngeal cancer research, both overexpression in AMC-HN-8 cells and knockdown in TU212 cells were utilized to demonstrate consistent functional effects across complementary experimental approaches .

What mechanisms explain RPN2's role in promoting tumor progression?

RPN2 promotes tumor progression through multiple interconnected mechanisms:

Epithelial-Mesenchymal Transition (EMT) regulation:

  • RPN2 overexpression increases expression of mesenchymal markers (e.g., Vimentin) while decreasing epithelial markers (e.g., E-cadherin) .

  • These changes enhance cancer cell migration, invasion, and metastatic potential.

  • Western blotting analysis in bladder cancer models confirmed that RPN2 knockdown suppresses EMT .

PI3K-Akt pathway activation:

  • RPN2 modulates phosphorylation of PI3K and Akt, promoting cell survival and proliferation .

  • In laryngeal cancer cells, treatment with PI3K inhibitor LY294002 reversed the effects of RPN2 overexpression, confirming pathway involvement .

Reactive Oxygen Species (ROS) modulation:

  • In laryngeal cancer, RPN2 increases ROS production, which subsequently activates PI3K/Akt signaling .

  • Treatment with N-acetyl-L-cysteine (NAC), an ROS inhibitor, attenuated RPN2-induced activation of PI3K/Akt .

Metabolic reprogramming:

  • RPN2 promotes aerobic glycolysis (Warburg effect) by increasing expression of key glycolytic enzymes including hexokinase-2 (HK-2), pyruvate dehydrogenase kinase 1 (PDK1), and lactate dehydrogenase A (LDHA) .

  • Enhanced glycolysis provides energy and building blocks for rapidly proliferating cancer cells.

Treatment resistance mechanisms:

  • RPN2 regulates glycosylation of P-glycoprotein, affecting its membrane localization and drug efflux function .

  • This contributes to multi-drug resistance in various cancer types.

These mechanisms have been validated through a combination of in vitro functional assays and in vivo tumor models, providing strong evidence for RPN2's oncogenic role.

How does RPN2 expression correlate with clinical outcomes across different cancer types?

RPN2 serves as a prognostic biomarker across multiple cancer types, with consistent associations between high expression and poor outcomes:

Bladder cancer:

Laryngeal squamous cell carcinoma:

  • RPN2 is highly expressed in LSCC tissues compared to normal controls .

  • Elevated expression correlates with enhanced proliferation, migration, and glycolytic activity .

Other cancer types:

  • Breast cancer: RPN2 is highly expressed in breast cancer stem cells and associated with tumor metastasis and clinically aggressive features .

  • Non-small cell lung cancer (NSCLC): RPN2 silencing represses tumorigenicity and sensitizes tumors to cisplatin treatment, leading to longer survival in mouse models .

  • Osteosarcoma, esophageal squamous cell carcinoma, and colorectal cancer: RPN2 shows involvement in disease progression across these malignancies .

These findings have been established through a combination of approaches including immunohistochemistry of patient samples, qRT-PCR analysis, bioinformatic analysis of public datasets (TCGA, GEPIA), and survival analysis using Kaplan-Meier methods with log-rank tests .

What is the relationship between RPN2 and chemotherapy resistance?

RPN2 plays a significant role in chemotherapy resistance through several mechanisms:

Glycosylation-dependent mechanisms:

  • RPN2 regulates the glycosylation of multi-drug resistance proteins, particularly P-glycoprotein (P-gp) .

  • Proper glycosylation is essential for P-gp membrane localization and drug efflux function.

  • RPN2 inhibition reduces P-gp glycosylation, decreasing its membrane localization and restoring sensitivity to chemotherapeutic agents like docetaxel .

Signaling pathway modulation:

  • RPN2 activates the PI3K-Akt pathway, which promotes cell survival under stress conditions including chemotherapy exposure .

  • Inhibition of this pathway can restore chemosensitivity in cancer cells with high RPN2 expression.

Experimental evidence:

  • In non-small cell lung cancer models, RPN2 silencing sensitized tumors to cisplatin treatment, leading to longer survival of tumor-bearing mice .

  • Similar effects have been observed with various chemotherapeutic agents across multiple cancer types including breast cancer and osteosarcoma .

Clinical relevance:

  • The correlation between RPN2 expression and treatment outcomes suggests its potential utility as a predictive biomarker for chemotherapy response.

  • Patients with high RPN2 expression may benefit from more aggressive treatment approaches or combination therapies targeting RPN2-mediated resistance mechanisms.

These findings highlight RPN2 as a promising target for overcoming chemotherapy resistance, potentially through combination approaches that inhibit RPN2 alongside standard chemotherapeutic regimens.

What experimental design best evaluates RPN2's impact on cancer cell metabolism?

A comprehensive experimental design to evaluate RPN2's impact on cancer cell metabolism should include:

In vitro metabolic profiling:

  • Cell models preparation:

    • Generate paired cell lines with RPN2 knockdown, overexpression, and appropriate controls

    • Validate expression changes at protein and mRNA levels

    • Use multiple cell lines to ensure robust findings

  • Glucose metabolism assessment:

    • Measure glucose uptake using radiolabeled glucose (2-DG) or fluorescent glucose analogs

    • Quantify lactate production in culture media as an indicator of aerobic glycolysis

    • Determine extracellular acidification rate (ECAR) using Seahorse XF analyzer

  • Mitochondrial function analysis:

    • Measure oxygen consumption rate (OCR) using Seahorse XF analyzer

    • Assess mitochondrial membrane potential with fluorescent probes

    • Evaluate TCA cycle activity using isotope-labeled metabolites

  • Glycolytic enzyme analysis:

    • Quantify expression and activity of key glycolytic enzymes including:

      • Hexokinase-2 (HK-2)

      • Pyruvate dehydrogenase kinase 1 (PDK1)

      • Lactate dehydrogenase A (LDHA)

    • Use Western blotting, qRT-PCR, and enzymatic activity assays

  • Metabolomic analysis:

    • Perform targeted and untargeted metabolomics to identify metabolic pathway alterations

    • Use stable isotope labeling to track carbon flux through glycolysis and TCA cycle

Mechanistic investigations:

  • ROS measurement:

    • Quantify ROS levels using fluorescent probes like DCFDA

    • Use ROS inhibitors (e.g., N-acetyl-L-cysteine) to determine causality

  • Signaling pathway analysis:

    • Examine PI3K/Akt pathway activation via Western blotting

    • Use pathway inhibitors to establish causative relationships

  • Transcription factor analysis:

    • Assess activity of metabolism-related transcription factors (HIF-1α, c-Myc)

    • Perform ChIP assays to determine binding to glycolytic gene promoters

In vivo validation:

  • Xenograft models with modified RPN2 expression

  • PET imaging to assess glucose uptake in tumors

  • Ex vivo analysis of tumor tissue for metabolic enzymes and metabolites

  • Correlation with tumor growth and metastasis

This comprehensive approach would establish causal relationships between RPN2 expression and metabolic reprogramming in cancer cells, as demonstrated in the laryngeal cancer study where RPN2 overexpression increased glucose uptake, lactate production, and glycolytic enzyme expression .

How should researchers design experiments to investigate RPN2's role in treatment resistance?

A rigorous experimental design to investigate RPN2's role in treatment resistance should include:

Cell line selection and preparation:

  • Choose multiple cell lines representing the cancer type of interest

  • Include both intrinsically resistant and sensitive lines

  • Generate stable RPN2 knockdown and overexpression models

  • Validate expression changes at mRNA and protein levels

Drug sensitivity testing:

  • Short-term assays:

    • Perform dose-response curves using multiple cytotoxicity assays:

      • MTT or CCK-8 viability assays

      • Apoptosis assessment (Annexin V/PI staining)

      • Cell cycle analysis

    • Calculate IC50 values before and after RPN2 modulation

    • Test multiple drugs with different mechanisms of action

  • Long-term assays:

    • Colony formation assays to assess clonogenic survival

    • Development of resistant cell lines through chronic drug exposure

    • Assessment of RPN2's role in acquired resistance

Mechanistic investigations:

  • Glycosylation analysis:

    • Assess glycosylation status of drug transporters (e.g., P-glycoprotein)

    • Compare before and after RPN2 modulation

    • Use enzymatic deglycosylation (PNGase F) to confirm glycosylation-dependent effects

  • Drug accumulation studies:

    • Measure intracellular drug concentrations

    • Assess drug efflux activity using fluorescent substrates

  • Signaling pathway analysis:

    • Examine PI3K/Akt pathway activation

    • Use pathway inhibitors (e.g., LY294002) to reverse resistance phenotypes

In vivo validation:

  • Establish xenograft models with modified RPN2 expression

  • Initiate treatment when tumors reach defined size

  • Monitor tumor growth, survival, and treatment response

  • Perform ex vivo analysis of tumor tissue

Clinical correlation:

  • Analyze RPN2 expression in patient samples before and after treatment

  • Correlate expression with treatment response and outcomes

  • Stratify analysis by treatment regimen and cancer subtype

This approach has been partially demonstrated in studies showing that RPN2 silencing can sensitize non-small cell lung cancer to cisplatin treatment and restore docetaxel sensitivity . The comprehensive design outlined above would establish both the clinical relevance and mechanistic basis of RPN2-mediated treatment resistance.

What are the most effective methods to validate RPN2 as a potential therapeutic target?

Validating RPN2 as a therapeutic target requires a systematic approach addressing efficacy, specificity, and translational potential:

Target validation pipeline:

  • Expression and clinical correlation:

    • Comprehensive analysis of RPN2 expression across tumor types

    • Association with clinical outcomes in sufficiently powered cohorts

    • Multivariate analysis to establish independent prognostic value

    Current evidence demonstrates RPN2 overexpression in multiple cancers including bladder cancer, laryngeal cancer, breast cancer, and NSCLC, with consistent associations with poor prognosis .

  • In vitro functional validation:

    ApproachKey FindingsTechnical Details
    Genetic silencingReduced proliferation, migration, invasionshRNA/siRNA targeting RPN2, validated by Western blot
    OverexpressionEnhanced malignant phenotypesPlasmid-based expression systems
    Rescue experimentsRestoration of phenotypesRe-expression of RPN2 in knockdown cells
    Pathway analysisEffects on EMT, PI3K/Akt, glycolysisWestern blotting for pathway components
  • In vivo validation:

    • Xenograft models demonstrate:

      • Reduced tumor growth with RPN2 silencing in bladder cancer models

      • Decreased tumor volume, proliferation marker Ki-67, and vimentin expression in laryngeal cancer models

      • Enhanced sensitivity to cisplatin in NSCLC models

    • These findings confirm RPN2's oncogenic role across multiple tumor types

  • Therapeutic window assessment:

    • Comparison of effects on cancer versus normal cells

    • Evaluation of potential toxicities

    • Assessment of compensatory mechanisms

  • Translational development:

    • Development of clinically viable targeting strategies:

      • siRNA delivery systems

      • Small molecule inhibitor screening

      • Potential antibody approaches

    • Companion diagnostic development for patient selection

The evidence from multiple tumor types consistently supports RPN2 as a promising therapeutic target, with knockdown showing significant anti-tumor effects both in vitro and in vivo . The conservation of these effects across cancer types suggests a fundamental role in oncogenesis rather than a tumor-specific phenomenon.

How do researchers optimize statistical approaches for analyzing RPN2 expression in patient cohorts?

Optimizing statistical approaches for RPN2 expression analysis in patient cohorts requires careful consideration of data characteristics and research questions:

Sample size determination:

  • Perform power calculations based on:

    • Expected effect size (derived from preliminary data)

    • Desired statistical power (typically 0.8)

    • Significance level (typically α = 0.05)

  • Ensure sufficient sample sizes for subgroup analyses

Data normalization and quality control:

  • For qRT-PCR: Normalize to stable reference genes

  • For protein quantification: Use appropriate loading controls

  • For RNAseq: Apply FPKM/TPM normalization

  • Assess data distribution to determine appropriate statistical tests

Expression analysis approaches:

Threshold determination:

  • For dichotomizing continuous RPN2 expression:

    • Median split (used in bladder cancer studies)

    • Receiver operating characteristic (ROC) curve analysis

    • Minimal p-value approach

  • Sensitivity analyses with different thresholds to ensure robustness

Reporting standards:

  • Include complete statistical methodology

  • Report exact p-values rather than thresholds

  • Present appropriate measures of effect size and confidence intervals

  • Use standardized visualizations (e.g., Kaplan-Meier curves, box plots)

What are the most promising approaches for developing RPN2-targeted therapies?

Several promising approaches exist for developing RPN2-targeted therapies, each with specific advantages and challenges:

RNA interference-based approaches:

  • siRNA delivery systems targeting RPN2 mRNA

  • Requires optimization of delivery vehicles (liposomes, nanoparticles)

  • Need for tumor-specific targeting to minimize off-target effects

  • Preclinical evidence supports efficacy in non-small cell lung cancer and other models

Small molecule inhibitors:

  • Targeting RPN2's glycosyltransferase activity or protein-protein interactions

  • Requires high-throughput screening campaigns

  • Structure-based drug design using RPN2 protein structure

  • Development of assays to measure RPN2 activity

Combination therapy strategies:

  • Combining RPN2 inhibition with:

    • Conventional chemotherapy to overcome resistance

    • PI3K/Akt pathway inhibitors to enhance efficacy

    • Glycolysis inhibitors to target metabolic vulnerabilities

  • Studies show RPN2 silencing sensitizes cancer cells to docetaxel and cisplatin

Antibody-based approaches:

  • Development of antibodies targeting RPN2 or RPN2-dependent glycoproteins

  • Potential for antibody-drug conjugates for targeted delivery

  • Challenges include limited accessibility of ER-resident proteins

Patient selection strategies:

  • Development of companion diagnostics to identify patients with high RPN2 expression

  • Incorporation of RPN2 testing into clinical decision-making

  • Identification of molecular signatures that predict response to RPN2-targeted therapy

The translational potential of RPN2 as a therapeutic target is supported by consistent findings across multiple cancer types, including bladder cancer, laryngeal cancer, and breast cancer . The relationship between RPN2 and treatment resistance further suggests that RPN2-targeted therapies might be particularly valuable in recurrent or refractory disease settings.

What contradictions in the current understanding of RPN2 function require further investigation?

While research presents a largely consistent picture of RPN2 as an oncogenic factor, several areas require further investigation to resolve contradictions or knowledge gaps:

Tissue-specific effects:

  • Current research shows oncogenic roles in multiple cancers, but the relative importance of different downstream mechanisms may vary between cancer types.

  • Question: Do different cancer types utilize distinct RPN2-dependent pathways, or is there a universal mechanism?

  • Approach: Comparative studies across cancer types using standardized methodologies and comprehensive pathway analysis.

Regulatory mechanisms:

  • Limited understanding of what regulates RPN2 expression in normal and cancer cells.

  • Question: What transcription factors, epigenetic mechanisms, or post-transcriptional regulators control RPN2 expression?

  • Approach: Promoter analysis, ChIP-seq, methylation profiling, and miRNA studies to identify regulatory mechanisms.

Glycosylation-independent functions:

  • Uncertainty about whether RPN2's effects are exclusively mediated through glycosylation.

  • Question: Does RPN2 have functions beyond its established role in the N-oligosaccharyl transferase complex?

  • Approach: Structure-function studies with RPN2 mutants lacking glycosyltransferase activity but retaining other domains.

ROS-RPN2 relationship:

  • The relationship between RPN2 and reactive oxygen species is not fully elucidated.

  • Question: Is ROS production a direct consequence of RPN2 activity or an indirect effect?

  • Approach: Time-course studies examining the sequence of events following RPN2 modulation, combined with specific inhibitors of intermediate pathways.

Normal physiological roles:

  • Limited information on RPN2's normal functions makes it difficult to predict potential toxicities of targeting.

  • Question: What are the consequences of RPN2 inhibition in normal tissues?

  • Approach: Conditional knockout models and tissue-specific analyses to characterize physiological roles.

Patient heterogeneity:

  • Variable RPN2 expression within and between patients may affect therapeutic targeting.

  • Question: How heterogeneous is RPN2 expression within tumors, and does this change during progression?

  • Approach: Single-cell analysis of RPN2 expression in patient samples at different disease stages.

These areas of uncertainty represent important research opportunities that, when addressed, will enhance our understanding of RPN2 biology and inform therapeutic development strategies.

How should researchers design experiments to investigate RPN2's potential non-glycosylation functions?

Investigating RPN2's potential non-glycosylation functions requires carefully designed experiments that distinguish between glycosylation-dependent and independent effects:

Structure-function analysis:

  • Domain mapping:

    • Generate RPN2 mutants lacking specific domains or with point mutations in catalytic residues

    • Express these mutants in RPN2-knockout backgrounds

    • Assess which functions are restored by which mutants

    • Compare glycosylation-deficient mutants with wild-type RPN2

  • Protein interaction studies:

    • Perform immunoprecipitation followed by mass spectrometry to identify RPN2 binding partners

    • Use proximity labeling approaches (BioID, APEX) to identify the RPN2 interactome

    • Validate key interactions using co-immunoprecipitation and FRET/BRET assays

    • Map interaction domains using truncation mutants

Comparative knockdown approaches:

  • RPN2 vs. other OST components:

    • Perform parallel knockdown of RPN2 and other OST complex components (e.g., RPN1, STT3A/B)

    • Compare phenotypic effects and pathway activation

    • Functions unique to RPN2 knockdown may represent glycosylation-independent roles

  • Rescue experiments:

    • Knockdown endogenous RPN2 and rescue with:

      • Wild-type RPN2

      • Glycosylation-deficient RPN2 mutants

      • RPN2 restricted to specific subcellular compartments

    • Assess which phenotypes are rescued by which constructs

Subcellular localization studies:

  • Non-ER functions:

    • Perform detailed subcellular fractionation to identify potential non-ER pools of RPN2

    • Use super-resolution microscopy to visualize RPN2 distribution

    • Identify signals that might regulate RPN2 trafficking

  • Forced mislocalization:

    • Create fusion proteins targeting RPN2 to specific compartments

    • Assess functional consequences of altered localization

Pathway analysis:

  • Temporal studies:

    • Perform time-course analysis after RPN2 modulation

    • Identify early vs. late effects

    • Early effects may represent direct non-glycosylation functions

  • Glycosylation inhibitors:

    • Compare effects of RPN2 knockdown with general N-glycosylation inhibitors (tunicamycin)

    • Effects unique to RPN2 manipulation suggest glycosylation-independent functions

These experimental approaches would help distinguish RPN2's glycosylation-dependent functions from potential novel roles, providing a more complete understanding of its contribution to cancer biology and potentially revealing new therapeutic opportunities.

What experimental designs best investigate the relationship between RPN2 and the tumor microenvironment?

Investigating RPN2's relationship with the tumor microenvironment requires sophisticated experimental designs that capture the complexity of cellular interactions:

Co-culture systems:

  • 2D co-culture models:

    • Culture cancer cells with modified RPN2 expression alongside:

      • Cancer-associated fibroblasts

      • Immune cells (T cells, macrophages)

      • Endothelial cells

    • Assess bidirectional effects on cellular phenotypes

    • Measure secreted factors in conditioned media

  • 3D co-culture models:

    • Establish spheroid or organoid cultures containing multiple cell types

    • Modify RPN2 expression in cancer cells within these systems

    • Evaluate effects on:

      • Spheroid formation and growth

      • Invasion into surrounding matrix

      • Cellular composition and spatial organization

Extracellular vesicle studies:

  • EV isolation and characterization:

    • Isolate extracellular vesicles from cells with modified RPN2 expression

    • Characterize vesicle content (proteins, RNAs, glycans)

    • Assess effects of these EVs on recipient stromal cells

  • Glycosylation analysis:

    • Determine how RPN2 affects glycosylation patterns of secreted proteins and EVs

    • Investigate how these altered glycosylation patterns affect recognition by immune cells

In vivo microenvironment analysis:

  • Syngeneic mouse models:

    • Use immunocompetent mouse models with modified RPN2 expression in cancer cells

    • Analyze tumor-infiltrating immune cells

    • Assess vascularization and stromal composition

  • Spatial transcriptomics/proteomics:

    • Apply spatial profiling technologies to xenograft tumors with modified RPN2

    • Map expression patterns of cancer and stromal markers

    • Identify spatial relationships between RPN2-expressing cells and microenvironment components

Secretome analysis:

  • Conditioned media studies:

    • Collect conditioned media from cells with modified RPN2 expression

    • Perform proteomic analysis of secreted factors

    • Test functional effects on stromal cell types

    • Identify glycosylation changes in secreted proteins

  • Cytokine/chemokine profiling:

    • Measure production of inflammatory mediators

    • Assess effects on immune cell recruitment and activation

Clinical correlation:

  • Multiplex immunohistochemistry:

    • Perform multiplexed staining of patient samples for:

      • RPN2 expression

      • Immune cell markers

      • Vascular markers

      • Extracellular matrix components

    • Analyze spatial relationships and correlations

  • Single-cell analysis of tumor ecosystems:

    • Apply single-cell RNA-seq to dissociated tumors

    • Correlate RPN2 expression with microenvironmental features

These experimental approaches would provide comprehensive insights into how RPN2 expression in cancer cells influences the tumor microenvironment, potentially revealing new therapeutic strategies targeting these interactions.

Product Science Overview

Introduction

Ribophorin II, also known as dolichyl-diphosphooligosaccharide–protein glycosyltransferase subunit 2, is a crucial component of the N-oligosaccharyl transferase complex. This complex is responsible for the attachment of high mannose oligosaccharides to asparagine residues in nascent polypeptide chains, specifically within the Asn-X-Ser/Thr consensus motif .

Structure and Function

Ribophorin II is a non-catalytic subunit of the oligosaccharyltransferase complex. It plays a significant role in the glycosylation process, which is essential for proper protein folding and stability. The human recombinant form of Ribophorin II is produced in Escherichia coli and is a single, non-glycosylated polypeptide chain containing 539 amino acids, with a molecular mass of approximately 59.2 kilodaltons .

Production and Purification

The recombinant Ribophorin II is fused to a 21 amino acid His-tag at the N-terminus, which facilitates its purification through proprietary chromatographic techniques. The protein is typically formulated in a solution containing 20 millimolar Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.1 molar sodium chloride .

Stability and Storage

For optimal stability, Ribophorin II should be stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to keep the protein frozen at -20°C, with the addition of a carrier protein such as human serum albumin or bovine serum albumin to prevent degradation. It is important to avoid multiple freeze-thaw cycles to maintain the protein’s integrity .

Biological Significance

Ribophorin II is highly conserved across various species, indicating its essential role in cellular processes. It is involved in the glycosylation of proteins, a critical post-translational modification that affects protein folding, stability, and function. The protein’s importance is underscored by its association with several diseases, including progressive encephalopathy with brain atrophy and spasticity, and hereditary sensory and autonomic neuropathy type VII .

Research Applications

Recombinant Ribophorin II is widely used in laboratory research to study protein glycosylation and its implications in various biological processes and diseases. Its high purity and stability make it a valuable tool for researchers investigating the mechanisms of protein folding and the role of glycosylation in health and disease .

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.