PDCD5 Human

Programmed Cell Death-5 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Apoptosis Regulation

PDCD5 promotes apoptosis via mitochondrial pathways and p53 activation:

  • Mitochondrial Pathway: Facilitates Bax translocation, cytochrome c release, and caspase-3 activation .

  • p53 Pathway: Stabilizes p53 by sequestering Mdm2, enhancing DNA damage-induced apoptosis .

Endothelial Function and Cardiovascular Health

PDCD5 modulates endothelial nitric oxide (NO) production by disrupting HDAC3–AKT interactions:

  • Mechanism: Competitively inhibits HDAC3 binding to AKT, reducing eNOS phosphorylation and NO synthesis .

  • Clinical Correlation: Serum PDCD5 levels inversely correlate with cardiovascular risk factors (e.g., diabetes, coronary calcium) .

Cancer Biology

PDCD5 acts as a tumor suppressor in multiple cancers:

Cancer TypeExpressionFunctional RoleClinical Outcome
Hepatocellular Carcinoma↓ mRNA/protein Induces apoptosis, cell cycle arrest (G1)Improved survival with PDCD5 expression
Osteosarcoma↓ mRNA/protein Suppresses Ras/Raf/MEK/ERK signalingPrognostic marker for recurrence
Chondrosarcoma↓ mRNA/protein Sensitizes cells to cisplatinEnhanced chemotherapeutic efficacy

Inflammation and Fibrosis

PDCD5 regulates immunological responses and extracellular matrix remodeling:

  • Pulmonary Fibrosis: Elevated in idiopathic pulmonary fibrosis (IPF), promotes TGF-β-induced matricellular gene expression via PDCD5/β-catenin/Smad3 complex formation .

  • Autoimmune Diseases: Inhibits PDCD5-VHL pathway in ischemic brain injury, suggesting neuroprotective roles .

Protein Interactions

PDCD5 chaperones multiple signaling pathways through direct binding:

PathwayInteraction PartnersOutcomeReferences
Apoptosisp53, Mdm2, Tip60, CCTPromotes p53 stabilization and caspase activation
Endothelial FunctionHDAC3, AKT, eNOSInhibits NO production, reduces atherosclerosis
TGF-β Signalingβ-catenin, Smad3Enhances fibrotic gene transcription
Protein FoldingTRiC (CCT1, CCT3, CCT4)Stabilizes TRiC in open conformation, facilitates substrate folding

Key Findings:

  • PDCD5 binds TRiC’s open form, preventing substrate competition and ensuring efficient protein folding .

  • The N-terminal α-helix is critical for HDAC3 binding, as mutant L6R (HDAC3-binding deficient) fails to inhibit AKT signaling .

Post-Translational Modifications

  • Phosphorylation: Serine 118 phosphorylation by CK2 is required for nuclear translocation and apoptosis induction .

Biomarker for Disease

DiseasePDCD5 ExpressionClinical Implication
Atherosclerosis↑ Serum levelsReflects endothelial dysfunction, linked to cardiovascular risk
Hepatocellular Carcinoma↓ Tissue levelsIndependent predictor of poor survival
Pulmonary Fibrosis↑ Lung tissueDrives fibrotic pathogenesis via TGF-β signaling

Therapeutic Strategies

  • Cancer Therapy: Recombinant human PDCD5 (rhPDCD5) enhances chemosensitivity in osteosarcoma, chondrosarcoma, and leukemia .

  • Cardiovascular Diseases: Targeting PDCD5 to restore AKT-eNOS signaling may improve endothelial function in atherosclerosis .

  • Pulmonary Fibrosis: Inhibiting PDCD5/β-catenin/Smad3 complex formation could mitigate fibrotic remodeling .

Future Directions

  • Structural Studies: Elucidating the PDCD5-TRiC interface could inform drug design for protein folding disorders.

  • Translational Research: Validating serum PDCD5 as a non-invasive biomarker for cardiovascular and oncological diseases.

Product Specs

Introduction
PDCD5 is expressed in tumor cells during apoptosis regardless of the stimuli that initiated the process. Before apoptosis induction, PDCD5 is found in both the nucleus and cytoplasm. Once apoptosis begins, PDCD5 levels increase, and the protein relocates from the cytoplasm, accumulating in the nucleus. PDCD5 protein has a stable helical core structure consisting of a triple-helix bundle and two separate terminal regions. PDCD5 is a significant novel protein that regulates both apoptotic and non-apoptotic programmed cell death. It plays a role in the process of apoptosis and is important in regulating apoptotic processes in gastric cancer cells and gastric tumors. Additionally, PDCD5 is involved in the pathogenesis of rheumatoid arthritis. The -27G/-11A SNP is associated with reduced PDCD5 promoter activity and increased susceptibility to chronic myelogenous leukemia. The PDCD5 gene may be a target gene controlled by important apoptosis-related transcriptional factors during cell apoptosis.
Description
Recombinant PDCD5, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 125 amino acids. It has a molecular weight of 14 kDa.
Physical Appearance
Sterile, colorless solution.
Formulation
PDCD5 protein solution is supplied in 1x PBS at pH 7.4.
Stability
For short-term storage (2-4 weeks), store the solution at 4°C. For long-term storage, freeze the solution at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Programmed cell death protein 5, TF-1 cell apoptosis-related protein 19, Protein TFAR19, PDCD5, TFAR19, MGC9294.
Source
Escherichia Coli.
Amino Acid Sequence
MADEELEALR RQRLAELQAK HGDPGDAAQQ EAKHRGAEMR NSILAQVLDQ SARARLSNLA LVKPEKTKAV ENYLIQMARY GQLSEKVSEQ GLIEILKKVS QQTEKTTTVK LNRRKVMDSD EDDDY.

Q&A

What is the primary function of PDCD5 in human cells?

PDCD5 functions primarily as a pro-apoptotic factor with tumor suppressor capabilities. It plays a pivotal role in both apoptotic and paraptotic cell death pathways . At the molecular level, PDCD5 competitively inhibits the interaction between histone deacetylase 3 (HDAC3) and protein kinase B (PKB/AKT), which subsequently affects AKT-eNOS signaling and nitric oxide production in vascular endothelium . This mechanism contributes to its role in regulating cellular homeostasis, particularly in endothelial cells. Additionally, PDCD5 forms a complex with the cytosolic chaperonin CCT and inhibits β-tubulin folding, which may constitute another mechanism through which it exerts its pro-apoptotic effects .

How is PDCD5 structurally characterized, and what domains are crucial for its function?

PDCD5 contains specific functional domains that are critical for its biological activity. Research has identified residues 109-115 as particularly important for its cellular translocation capability and protein cargo internalization . Through deletion mutagenesis studies, these residues have been shown to drive the internalization of large protein cargo, including the Mdm-2 binding domain of human p53 into living cells . The protein also contains regions that enable its remarkable intercellular transport capabilities through a clathrin-independent endocytic pathway originating from heparan sulfate proteoglycan binding and lipid rafts . Mutation studies, particularly with the PDCD5 L6R mutant (an HDAC3-binding–deficient variant), have demonstrated that specific residues are crucial for PDCD5's interaction with HDAC3 and subsequent inhibition of AKT signaling .

What experimental methods are commonly used to detect and measure PDCD5 expression levels?

Several laboratory methods are employed to detect and quantify PDCD5 expression:

  • Quantitative PCR (qPCR): Using specific primer sequences such as:

    • Human PDCD5: (F) 5'-CTTGAGGCGCTGAGGAGAC-3', (R) 5'-GGCCGACTGATCCAGAACTT-3'

    Expression levels are typically calculated using the 2 × 2 −ΔΔCt method, with normalization to housekeeping genes like GAPDH .

  • Protein Detection Methods:

    • Western blotting for protein quantification

    • Immunofluorescence for cellular localization

    • Enzyme-linked immunosorbent assay (ELISA) for measuring serum PDCD5 levels

  • Mass Spectrometry: For protein identification in co-immunoprecipitation studies, which can be performed using LTQ-Orbitrap mass spectrometry interfaced with nanoAcquity UPLC systems .

When analyzing PDCD5 in clinical samples, it's critical to consider cell type distributions, as different expression patterns may exist between whole blood, peripheral blood mononuclear cells (PBMCs), and granulocytes .

How does PDCD5 contribute to atherosclerosis development and vascular function?

PDCD5 significantly influences vascular function through its interaction with the AKT-eNOS signaling pathway, which is crucial for endothelial homeostasis . In endothelial cells, PDCD5 disrupts the HDAC3–AKT interaction, thereby inhibiting AKT and eNOS phosphorylation and reducing nitric oxide (NO) production . This mechanism directly affects vascular health in several ways:

  • Vascular Remodeling: Endothelial-specific PDCD5 knockout mice showed significantly reduced vascular remodeling compared with wild-type mice after partial carotid ligation, demonstrating PDCD5's role in pathological vascular changes .

  • Endothelial Dysfunction: PDCD5 is associated with endothelial dysfunction, a key factor in atherosclerosis development. Serum PDCD5 levels reflect endothelial NO production status, which is crucial for maintaining vascular health .

  • Clinical Correlations: Serum PDCD5 levels correlate with diabetes mellitus, high-density lipoprotein cholesterol, and coronary calcium in cardiovascular high-risk cohorts, suggesting its potential as a biomarker for cardiovascular risk assessment .

Research methodologies for studying PDCD5 in vascular function typically involve endothelial-specific knockout models, vascular remodeling assays following partial carotid ligation, and in vitro studies of PDCD5's effect on AKT-eNOS signaling in human umbilical vein endothelial cells .

What is the relationship between PDCD5 expression and autoimmune conditions like rheumatoid arthritis?

PDCD5 expression is significantly upregulated in various autoimmune conditions, including rheumatoid arthritis (RA) . The relationship between PDCD5 and RA is characterized by:

  • Increased Expression: Serum and synovial levels of PDCD5 protein are significantly higher in RA patients compared to healthy controls .

  • Correlation with Disease Activity: PDCD5 expression positively correlates with several important clinical parameters in RA patients as shown in the following correlations:

    Clinical ParameterCorrelation Coefficient (r)P-value
    ESR0.772<0.001
    CRP0.755<0.001
    RF0.767<0.001
    Anti-CCP0.656<0.001
    DAS28 score0.707<0.001
    IgG0.744<0.001
    IgA0.714<0.001
    IgM0.648<0.001
  • Potential Mechanism: PDCD5 promotes activation-induced cell death (AICD) of auto-reactive inflammatory Th1 and Th17 cells, which secrete TNF-α, IFN-γ, IL-17A, and IL-6 . The increased PDCD5 expression in RA may represent a defense mechanism attempting to eliminate apoptosis-resistant auto-reactive immune cells .

  • Biomarker Potential: PDCD5 shows promise as a novel biomarker for predicting both the incidence and remission of RA, potentially improving therapeutic management strategies .

Experimental approaches for studying PDCD5 in autoimmune conditions include gene expression analysis in whole blood, PBMCs, and granulocytes, correlation analyses with inflammatory markers, and comparative studies between active and remission disease states .

How does PDCD5 expression differ between cancer tissues and normal tissues?

While the search results don't provide specific comparative data on PDCD5 expression in cancer versus normal tissues, they do indicate that the pathological relevance of PDCD5 is mostly found in human cancers . PDCD5 has been proposed to function as a pro-apoptotic factor with tumor suppressor capabilities , suggesting that its expression or function may be altered in cancer tissues.

For researchers investigating PDCD5 in cancer, recommended methodological approaches include:

  • Comparative Expression Analysis: Quantifying PDCD5 mRNA and protein levels in matched tumor and adjacent normal tissues using qPCR, Western blotting, and immunohistochemistry.

  • Functional Studies: Examining the effect of PDCD5 overexpression or knockdown on cancer cell proliferation, apoptosis, and migration.

  • Mechanistic Investigations: Exploring PDCD5's interaction with known cancer-related pathways, particularly its role in inhibiting β-tubulin folding through interaction with the cytosolic chaperonin CCT , which could affect microtubule dynamics and cell division.

  • Clinical Correlation Studies: Analyzing the relationship between PDCD5 expression levels and clinical parameters such as tumor stage, grade, and patient survival.

What are the recommended protocols for purifying recombinant PDCD5 protein for functional studies?

While the search results don't provide a detailed purification protocol, they do mention the use of recombinant PDCD5 added exogenously to culture medium to enhance programmed cell death . Based on standard protein purification practices and the information available, researchers should consider:

  • Expression System Selection: Bacterial expression systems (E. coli) are commonly used for producing recombinant human proteins. For studies requiring post-translational modifications, consider mammalian or insect cell expression systems.

  • Affinity Tag Strategy: Incorporate histidine, GST, or other affinity tags to facilitate purification. Design constructs carefully to ensure tags don't interfere with PDCD5's functional domains, particularly residues 109-115 which are critical for its translocation activity .

  • Purification Steps:

    • Affinity chromatography (Ni-NTA for His-tagged proteins)

    • Ion exchange chromatography to remove impurities

    • Size exclusion chromatography for final polishing

    • Consider tag removal using specific proteases if the tag might interfere with function

  • Quality Control:

    • SDS-PAGE for purity assessment

    • Western blotting for identity confirmation

    • Mass spectrometry for accurate molecular weight determination

    • Circular dichroism for secondary structure verification

    • Functional assays to confirm biological activity, such as testing the purified protein's ability to enhance apoptosis in appropriate cell models

  • Storage Considerations: Determine optimal buffer conditions and storage temperature to maintain stability and activity. Consider adding glycerol or other stabilizing agents.

What cell-based assays are most effective for studying PDCD5's pro-apoptotic function?

Based on the information in the search results, several cell-based assays can be employed to study PDCD5's pro-apoptotic function:

  • Exogenous PDCD5 Treatment Assays: Recombinant PDCD5 can be added exogenously to culture medium to enhance programmed cell death triggered by certain stimuli . This approach allows for dose-response studies and time-course analyses of PDCD5's pro-apoptotic effects.

  • PDCD5 Overexpression and Knockdown Studies:

    • Transfection with PDCD5 expression vectors or siRNA/shRNA

    • Creation of stable cell lines with inducible PDCD5 expression

    • CRISPR-Cas9-mediated genetic modification

  • Apoptosis Detection Methods:

    • Annexin V/PI staining followed by flow cytometry

    • TUNEL assay for DNA fragmentation

    • Caspase activity assays (particularly caspase-3)

    • Mitochondrial membrane potential measurement

    • Western blotting for apoptotic markers (cleaved PARP, cleaved caspases)

  • Mechanism-Specific Assays:

    • AKT phosphorylation status assessment (given PDCD5's role in disrupting HDAC3-AKT interaction)

    • β-tubulin folding assays (as PDCD5 inhibits β-tubulin folding through interaction with CCT)

    • Co-immunoprecipitation studies to examine protein-protein interactions

    • Subcellular localization studies using immunofluorescence microscopy

  • Cell Type Considerations: Based on existing research, human umbilical vein endothelial cells , cancer cell lines, and cells involved in autoimmune responses (such as T-cells) are relevant models for studying PDCD5 function.

How can researchers effectively design experiments to study PDCD5's intercellular transport mechanism?

PDCD5 exhibits a remarkable role in intercellular transport through a clathrin-independent endocytic pathway . To effectively study this mechanism, researchers should consider the following experimental design approaches:

  • Fluorescent Labeling Techniques:

    • Label recombinant PDCD5 with fluorescent tags (e.g., FITC as mentioned in the search results)

    • Use confocal microscopy for real-time visualization of PDCD5 internalization

    • Perform pulse-chase experiments to track the kinetics of PDCD5 uptake and intracellular trafficking

  • Pathway Dissection Strategies:

    • Employ specific inhibitors of different endocytic pathways (clathrin-dependent, caveolae-mediated, lipid raft-dependent)

    • Use drugs that disrupt lipid rafts or compete with cell membrane heparan sulfate proteoglycans

    • Overexpress dominant negative mutants of pathway components (as mentioned, overexpression of clathrin dominant negative mutant form did not block PDCD5-FITC uptake)

  • Structural Studies:

    • Perform deletion mutagenesis to map critical domains for internalization (residues 109-115 have been identified as important)

    • Create fusion proteins to test PDCD5's ability to drive internalization of cargo proteins

  • Biochemical Characterization:

    • Study the resistance of PDCD5-containing endosomes to nonionic detergents

    • Use sucrose density centrifugation to analyze PDCD5 localization in lipid rafts

    • Employ electron microscopy to visualize PDCD5 in membrane microdomains

  • Cell Type Considerations:

    • Compare PDCD5 uptake in caveolin-1-positive and caveolin-1-negative cells

    • Investigate potential cell-type specific differences in internalization mechanisms

By combining these approaches, researchers can comprehensively characterize PDCD5's intercellular transport mechanism and potentially leverage this function for therapeutic protein delivery applications, as suggested by PDCD5's ability to introduce the Mdm-2 binding domain of human p53 into living cells .

How might PDCD5's role in β-tubulin folding connect to its pro-apoptotic function?

PDCD5 forms a complex with the cytosolic chaperonin CCT and inhibits β-tubulin folding . This finding presents an intriguing mechanistic link between PDCD5's chaperonin-modulating activity and its pro-apoptotic function. Researchers exploring this connection should consider:

  • Mechanistic Investigations:

    • Determine whether PDCD5-mediated inhibition of β-tubulin folding leads to microtubule destabilization, which could trigger apoptotic signaling

    • Investigate if PDCD5 selectively inhibits β-tubulin folding under specific cellular conditions (e.g., during apoptotic stimuli)

    • Examine whether PDCD5 affects the folding of other CCT substrates besides β-tubulin

  • Structural Studies:

    • Use cryo-electron microscopy to determine the structure of the PDCD5-CCT complex

    • Identify the specific binding interface and critical residues involved in the interaction

    • Design mutants that selectively disrupt the PDCD5-CCT interaction without affecting other functions

  • Functional Correlation Studies:

    • Compare the kinetics of PDCD5-mediated inhibition of β-tubulin folding with the timing of apoptotic events

    • Determine if restoration of proper β-tubulin folding can rescue cells from PDCD5-induced apoptosis

    • Investigate the relationship between PDCD5 levels, CCT activity, and cell susceptibility to apoptosis

  • Cancer Relevance:

    • Examine whether cancer cells with altered tubulin dynamics show differential sensitivity to PDCD5-induced apoptosis

    • Explore potential synergies between PDCD5 and microtubule-targeting chemotherapeutic agents

The connection between PDCD5's role in β-tubulin folding and its pro-apoptotic function represents an exciting frontier in PDCD5 research with potential implications for cancer therapy.

What are the most promising therapeutic applications of PDCD5 research in treating autoimmune diseases?

PDCD5 research shows significant potential for therapeutic applications in autoimmune diseases, particularly in rheumatoid arthritis (RA). Based on the search results, promising directions include:

  • Biomarker Development:

    • PDCD5 expression shows good efficacy for predicting disease status and clinical outcomes in RA

    • Its expression correlates strongly with multiple clinical parameters (ESR, CRP, RF, anti-CCP, etc.)

    • Longitudinal monitoring of PDCD5 expression could help predict disease flares and remission

  • Therapeutic Target Exploration:

    • Given PDCD5's role in promoting activation-induced cell death (AICD) of auto-reactive inflammatory Th1 and Th17 cells , enhancing PDCD5 function could potentially help eliminate pathogenic immune cells

    • The TIP60-FOXP3-Treg axis mediated by PDCD5 presents another potential intervention point

  • Protein Delivery Technology:

    • PDCD5's ability to drive the internalization of large protein cargo could be leveraged to deliver therapeutic proteins to specific cell types

    • This property could enable targeted delivery of immunomodulatory proteins to diseased tissues

  • Combined Biomarker Approaches:

    • Integrating PDCD5 measurement with other autoimmune markers could improve diagnostic and prognostic accuracy

    • Researchers should investigate potential synergistic biomarker panels including PDCD5, FOXP3, and inflammatory cytokines

For researchers pursuing these therapeutic applications, experimental approaches should include in vivo autoimmune disease models, patient-derived cell studies, and longitudinal clinical investigations tracking PDCD5 expression in relation to disease progression and treatment response.

How can contradictory findings regarding PDCD5's role in different disease contexts be reconciled?

The search results reveal potentially contradictory aspects of PDCD5 function across different disease contexts. Researchers attempting to reconcile these apparent contradictions should consider:

  • Context-Dependent Functions:

    • PDCD5 appears to have different roles in cancer (pro-apoptotic, tumor suppressor) versus autoimmune conditions (upregulated, potentially as a compensatory mechanism)

    • These seemingly contradictory functions may reflect tissue-specific or disease-specific modulation of PDCD5 activity

  • Methodological Approaches for Resolution:

    • Compare PDCD5 post-translational modifications across disease contexts

    • Identify disease-specific PDCD5 binding partners through proteomics approaches

    • Perform domain-specific functional studies using truncated proteins or point mutants

    • Use tissue-specific or cell-type-specific knockout models to clarify context-dependent functions

  • Integrated Signaling Pathway Analysis:

    • Map PDCD5's involvement in multiple signaling pathways (AKT-eNOS , CCT-tubulin , etc.)

    • Identify pathway nodes where disease-specific regulation might occur

    • Develop computational models to predict how pathway perturbations might alter PDCD5 function

  • Threshold Effects and Temporal Dynamics:

    • Investigate whether different PDCD5 expression levels trigger distinct cellular responses

    • Examine the temporal dynamics of PDCD5 expression and function during disease progression

    • Consider biphasic effects where moderate increases might be compensatory while extreme changes become pathological

By systematically addressing these factors, researchers can develop a more unified understanding of PDCD5's complex role across different pathophysiological contexts.

What are the best molecular techniques for studying PDCD5 interactions with binding partners like HDAC3 and CCT?

To effectively study PDCD5's interactions with binding partners such as HDAC3 and the cytosolic chaperonin CCT, researchers should employ a comprehensive set of molecular techniques:

  • Co-Immunoprecipitation and Mass Spectrometry:

    • As described in the search results, co-immunoprecipitation followed by tandem mass spectrometry (MS/MS) is effective for identifying PDCD5 binding partners

    • Sample preparation should include DTT reduction, iodoacetamide alkylation, and trypsin digestion

    • MS/MS can be performed using an LTQ-Orbitrap mass spectrometer interfaced with nanoAcquity UPLC systems

    • Database search engines like Sequest, SequestHT, and Mascot can be used for protein identification

  • Structural Analysis Techniques:

    • X-ray crystallography of PDCD5 complexes with binding partners

    • Cryo-electron microscopy for visualizing larger complexes like PDCD5-CCT

    • Nuclear magnetic resonance (NMR) for studying dynamic interactions

  • Protein-Protein Interaction Validation Methods:

    • FRET (Förster resonance energy transfer) or BRET (bioluminescence resonance energy transfer) to confirm direct interactions in living cells

    • Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) to determine binding affinity and kinetics

    • Yeast two-hybrid or mammalian two-hybrid assays as complementary approaches

  • Functional Validation Approaches:

    • Mutagenesis studies targeting specific interaction domains

    • Competition assays with peptides derived from interaction interfaces

    • Cell-based functional assays to assess the biological significance of specific interactions

  • Data Analysis and Integration:

    • Use bioinformatics tools to predict interaction interfaces

    • Integrate data from multiple techniques to build comprehensive interaction models

    • Apply network analysis to place PDCD5 interactions in broader signaling contexts

The search results specifically mention that wild-type PDCD5 competitively inhibits interaction between HDAC3 and AKT, but the PDCD5 L6R mutant (HDAC3-binding–deficient) does not , highlighting the value of mutational analysis in studying specific interactions.

How should researchers account for cell-specific variations when studying PDCD5 expression in clinical samples?

When analyzing PDCD5 expression in clinical samples, researchers must carefully account for cell-specific variations to ensure accurate and interpretable results:

  • Cell Type Considerations:

    • The search results indicate that there may be differences in PDCD5 expression between whole blood, peripheral blood mononuclear cells (PBMCs), and granulocytes

    • A comprehensive analysis should examine expression in specific cell populations rather than just whole blood

  • Standardized Isolation Protocols:

    • Develop and adhere to standardized protocols for isolating specific cell populations

    • Consider density gradient centrifugation for separating PBMCs from granulocytes

    • Use cell sorting techniques (FACS or MACS) for isolating specific lymphocyte subsets

  • Analytical Approaches:

    • Use flow cytometry for single-cell analysis of PDCD5 expression in different immune cell subsets

    • Consider single-cell RNA sequencing to capture heterogeneity within cell populations

    • Employ immunohistochemistry or immunofluorescence for tissue samples to visualize cell-specific expression patterns

  • Normalization Strategies:

    • Account for variations in cell counts/percentages between patient samples

    • The search results mention differences in WBC, LYMPH, and HCT between RA patients and healthy controls

    • Consider normalized expression calculations or include cell counts as covariates in statistical analyses

  • Validation Across Multiple Techniques:

    • Compare results from different methodologies (qPCR, Western blotting, flow cytometry)

    • Validate findings in independent cohorts

    • Consider both mRNA and protein expression analyses

The search results note that while there were differences in cell counts between RA patients and healthy controls, further examination found that the expression trend of PDCD5 in PMBCs and granulocytes was consistent with that in whole blood . This suggests that for some conditions, whole blood analysis may be sufficient, but validation in specific cell populations remains important.

What are the critical considerations for developing PDCD5 as a clinical biomarker for disease prediction?

Developing PDCD5 as a clinical biomarker, particularly for conditions like rheumatoid arthritis where it shows promise , requires careful attention to several critical factors:

  • Analytical Validation:

    • Establish standardized measurement protocols with validated reagents

    • Determine assay precision, accuracy, reproducibility, and limits of detection

    • Develop reference standards for calibration across laboratories

    • Consider platform-specific variations (ELISA, qPCR, etc.)

  • Clinical Validation:

    • Conduct large-scale studies across diverse patient populations

    • Establish reference ranges in healthy populations of different ages, sexes, and ethnicities

    • Determine sensitivity, specificity, and predictive values for specific clinical endpoints

    • Compare performance against established biomarkers (e.g., CRP, ESR, RF for RA)

  • Biological Variability Assessment:

    • Characterize diurnal variations in PDCD5 expression

    • Assess the impact of non-disease factors (medication, exercise, stress)

    • Evaluate stability in samples under different storage conditions

    • Determine the effects of common comorbidities

  • Integration with Clinical Decision Making:

    • Develop clear thresholds for clinical interpretation

    • Create algorithms that incorporate PDCD5 with other clinical and laboratory parameters

    • Design prospective studies to evaluate the impact of PDCD5-guided management on patient outcomes

    • Establish cost-effectiveness of PDCD5 testing in clinical practice

  • Technical Implementation Considerations:

    • Develop point-of-care testing options if appropriate

    • Ensure test accessibility in various healthcare settings

    • Create quality control programs for clinical laboratories

    • Develop appropriate documentation and training materials

The search results indicate that PDCD5 expression shows good efficacy for predicting disease status and clinical outcomes in RA, with significant correlations to multiple established disease markers . This provides a strong foundation for further development, but the steps outlined above remain essential for translation into clinical practice.

Product Science Overview

Role in Apoptosis

PDCD5 is primarily known for its role in promoting apoptosis, a form of programmed cell death crucial for maintaining tissue homeostasis and eliminating damaged or infected cells . Apoptosis is a highly regulated process, and PDCD5 has been shown to enhance this process in various cancer cells . The protein is widely expressed in various cell lines, indicating its regulatory role in both pathological and physiological processes .

Expression in Cancer

Decreased expression of PDCD5 has been observed in several human tumors, including breast cancer, gastric cancer, astrocytic glioma, chronic myelogenous leukemia, and hepatocellular carcinoma . This suggests that PDCD5 may act as a tumor suppressor, and its reduced expression could contribute to tumorigenesis .

Mechanisms and Functions

Recent studies have highlighted several functions and mechanisms of PDCD5 in cancer cells:

  • Paraptosis: PDCD5 is involved in paraptosis, a type of programmed cell death distinct from apoptosis .
  • Cell Cycle Regulation: PDCD5 plays a role in regulating the cell cycle, ensuring that cells do not proliferate uncontrollably .
  • Immunoregulation: PDCD5 has been implicated in the immune response, potentially influencing how the body detects and responds to cancer cells .
  • Viral Infection: PDCD5 may also play a role in the body’s response to viral infections .
Therapeutic Potential

Given its role in promoting apoptosis and its decreased expression in various cancers, PDCD5 is being studied as a potential therapeutic target. Enhancing PDCD5 expression or function could provide a new avenue for cancer treatment by promoting the death of cancer cells .

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.