PCSK1N Human

Proprotein Convertase Subtilisin/Kexin Type 1 Inhibitor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to PCSK1N Human

PCSK1N, encoded by the PCSK1N gene (OMIM: 300399), is a neuroendocrine protein primarily expressed in cells with regulated secretory pathways, such as endocrine cells, neurons, and neuroendocrine tumor cells. It is also known as proSAAS (prohormone convertase 1-inhibiting protein) and functions as an endogenous inhibitor of proprotein convertase subtilisin/kexin type 1 (PCSK1), a key enzyme in hormone precursor processing . PCSK1N is processed into bioactive peptides, including Big PEN-LEN, PEN, and LEN, which modulate feeding behavior and neuroendocrine signaling .

Structure

PCSK1N contains:

  • N-terminal domain: Involved in chaperone activity.

  • C-terminal inhibitory domain: Contains the hexapeptide LLRVKR, critical for PCSK1 inhibition .

  • Cleavage sites: Processed into peptides (e.g., PEN, LEN) and a central chaperone domain .

Primary Functions

FunctionMechanismKey Findings
PCSK1 InhibitionBinds to PCSK1 (84 kDa form), blocking its activity in the ER/Golgi .Inhibits processing of proopiomelanocortin (POMC) and proenkephalin .
Chaperone ActivityPrevents β-amyloid and α-synuclein aggregation .Reduces oligomer cytotoxicity in neuronal models .
Feeding RegulationPEN and LEN bind GPR171, modulating appetite .PEN-LEN administration reduces food intake in rodents .

Corticotroph Pituitary Adenomas

PCSK1N expression distinguishes silent corticotroph adenomas (SCA) from functioning corticotroph adenomas (FCA):

ParameterSCAFCA
PCSK1N ExpressionElevated Reduced
POMC ExpressionLow High
Tumor SizeLarger (macroadenomas) Smaller (microadenomas)
ACTH/Cortisol LevelsLower Higher

Key Correlations:

  • Negative correlation between PCSK1N and POMC/TBX19 in adenomas .

  • Positive correlation with tumor size, suggesting a role in tumor growth .

ER Stress Response

In AtT-20 corticotroph cells, ER stress induction (via 17-AAG) upregulates PCSK1N expression, potentially conferring survival advantages through:

  • Downregulation of POMC.

  • Suppression of apoptosis and senescence pathways .

Tissue Expression and Localization

PCSK1N exhibits neuroendocrine-specific expression (Table 2):

Tissue/OrganExpression LevelProtein Localization
Pituitary GlandHighCorticotroph cells, adenoma cells
BrainModerateHippocampus, amygdala, hypothalamic nuclei
PancreasLowIslet cells
Adipose TissueLowNot significant in obesity

Transcriptional Control

Pax6 directly represses PCSK1N expression by binding to its promoter . In Pax6 mutant mice, PCSK1N levels rise, impairing insulin processing and glucose metabolism .

Post-Translational Processing

PCSK1N undergoes N- and C-terminal cleavage to generate:

  • Chaperone domain: Blocks protein aggregation.

  • Bioactive peptides: PEN, LEN, and Big PEN-LEN (GPR171 ligands) .

Protein Interactions

PCSK1N interacts with key neuroendocrine proteins (Table 3):

Interacting ProteinRoleFunctional Impact
PCSK1Prohormone convertaseInhibits PCSK1 activity
CPECarboxypeptidase ERegulates secretory granule trafficking
GPR171G-protein coupled receptorMediates feeding behavior via PEN/LEN
SCG2Secretogranin-2Regulates secretory granule biogenesis

Research Implications

  1. Biomarker Potential: PCSK1N may serve as a diagnostic marker for SCA vs. FCA .

  2. Therapeutic Target: Modulating PCSK1N could improve tumor management in corticotroph adenomas .

  3. Neurodegenerative Disease: Its chaperone activity may mitigate protein aggregation in Parkinson’s/Alzheimer’s .

Product Specs

Introduction
Proprotein Convertase Subtilisin/Kexin Type 1 Inhibitor (PCSK1N) is involved in regulating the neuroendocrine secretory pathway. It acts as an inhibitor of prohormone convertase 1, an enzyme responsible for the proteolytic cleavage of precursors to neuroendocrine peptides. PCSK1N slows down the convertase-mediated processing of proopiomelanocortin and proenkephalin. Additionally, it plays a role in controlling the intracellular timing of PCSK1.
Description
Recombinant human PCSK1N, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 251 amino acids (residues 34-260). It has a molecular weight of 26.6 kDa. The protein includes a 23 amino acid His-Tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The PCSK1N solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
ProSAAS precursor, Proprotein convertase subtilisin/kexin type 1 inhibitor, PROSAAS; SAAS, PCSK1N.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMARPVKE PRGLSAASPP LAETGAPRRF RRSVPRGEAA GAVQELARAL AHLLEAERQE RARAEAQEAE DQQARVLAQL LRVWGAPRNS DPALGLDDDP DAPAAQLARA LLRARLDPAA LAAQLVPAPV PAAALRPRPP VYDDGPAGPD AEEAGDETPD VDPELLRYLL GRILAGSADS EGVAAPRRLR RAADHDVGSE LPPEGVLGAL LRVKRLETPA PQVPARRLLP P.

Q&A

What is PCSK1N and what is its primary function in human physiology?

PCSK1N functions primarily as an endogenous inhibitor of proprotein convertase subtilisin/kexin type 1 (PCSK1 or PC1/3), a crucial enzyme involved in prohormone processing. By inhibiting PCSK1, PCSK1N affects the proteolytic cleavage of pro-opiomelanocortin (POMC) into adrenocorticotropic hormone (ACTH) . Beyond this inhibitory role, recent research has identified PCSK1N as a stress-responsive protein with potent anti-aggregation chaperone properties, capable of preventing protein aggregation in a dose-dependent manner . This dual functionality positions PCSK1N as a multifunctional protein involved in both hormonal regulation and cellular stress responses.

How is PCSK1N expression regulated at the transcriptional level?

PCSK1N expression is regulated through several well-characterized mechanisms, with transcriptional control being particularly important. Research has identified Pax6 (paired box 6) as a direct negative regulator that binds to the PCSK1N promoter region to down-regulate its expression . This regulatory relationship has been demonstrated through multiple experimental approaches including luciferase-reporter analysis, chromatin immunoprecipitation, and electrophoretic mobility shift assays . Additionally, PCSK1N expression appears to be responsive to endoplasmic reticulum (ER) stress conditions, with expression levels increasing as part of the cellular adaptive response to stress . This suggests a complex regulatory network that may vary across different tissue types and pathological states.

What methodological approaches are most reliable for measuring PCSK1N expression in human samples?

Several complementary methodologies are recommended for robust quantification of PCSK1N in human samples:

  • RT-qPCR (Reverse Transcription Quantitative PCR): For measuring PCSK1N mRNA expression, researchers should normalize to validated housekeeping genes such as GAPDH and ALAS1, which have been specifically validated for pituitary tumor samples . The typical Ct value range for PCSK1N in corticotroph adenoma samples has been reported as 18.1-29.9 cycles .

  • Western Blot Analysis: This protein detection method can determine PCSK1N protein levels in tissue extracts or cell cultures, requiring careful validation of antibody specificity and appropriate loading controls.

  • Immunohistochemistry (IHC): This technique provides valuable information about spatial distribution of PCSK1N within tissues, which is particularly important given its subcellular localization in the secretory pathway.

  • Mass Spectrometry-based Proteomics: For unbiased quantification, LC-MS/MS approaches can detect and quantify PCSK1N along with thousands of other proteins, providing a comprehensive view of PCSK1N in relation to the broader proteome .

Statistical analyses should employ appropriate non-parametric tests for comparing expression between groups, such as the Mann-Whitney U test, and Spearman correlation for examining relationships with clinical parameters .

What is the relationship between PCSK1N expression and pituitary adenomas?

Research has established significant correlations between PCSK1N expression and corticotroph adenomas. Silent corticotroph adenomas (SCAs) exhibit higher PCSK1N expression compared to functioning corticotroph adenomas (FCAs) . Furthermore, PCSK1N expression negatively correlates with corticotroph cell markers including POMC (r_s = -0.514; P < .001), TPIT (r_s = -0.386; P = .005), and PCSK1 (r_s = -0.3691; P = .008) .

Particularly noteworthy is the strong positive correlation between PCSK1N expression and tumor size (r_s = 0.645; P < .001) in all corticotroph adenomas . This correlation is especially robust in functioning corticotroph adenomas (r_s = 0.655; P < .001) . The relationship appears functionally significant: higher PCSK1N levels may reduce POMC processing into ACTH through inhibition of PCSK1, potentially explaining the reduced hormone production in SCAs .

Additionally, PCSK1N's involvement in ER stress responses suggests it may contribute to tumor cell survival mechanisms, potentially facilitating tumor growth over time . A negative correlation between PCSK1N and plasma cortisol levels (r_s = -0.356; P = .026) was identified, though no significant correlation with ACTH levels was found .

How does PCSK1N function in endoplasmic reticulum (ER) stress responses?

PCSK1N has been identified as a stress-responsive protein that increases in expression during ER stress conditions . Current research suggests that PCSK1N functions as a chaperone with potent anti-aggregation properties, preventing protein aggregation that would otherwise exacerbate ER stress . Following ER stress induction, cellular levels of PCSK1N increase, potentially rescuing cells from toxicity and providing a survival advantage .

Experimental studies using ER stress inducers like tanespimycin (17-AAG, an HSP90 chaperone inhibitor) have demonstrated that ER stress activation leads to increased PCSK1N expression along with downregulation of apoptosis, cell cycle, and senescence signaling pathways . In the experimental model using AtT-20 cells (a mouse pituitary corticotroph tumor cell line), induction of ER stress by 17-AAG led to a decrease of Pomc and an increase of Pcsk1n gene expression at 24 hours .

This combination of effects—increased PCSK1N expression and altered cellular pathways—could confer a survival advantage to cells under stress conditions . In the context of tumor biology, this mechanism may explain the observed correlation between PCSK1N expression and tumor size, as enhanced stress adaptation could contribute to sustained tumor growth .

What evidence suggests PCSK1N could serve as a biomarker for tumor aggressiveness?

Several lines of evidence support PCSK1N's potential as a biomarker for tumor aggressiveness:

  • Correlation with Tumor Size: There is a strong positive correlation between PCSK1N expression and tumor diameter (r_s = 0.645; P < .001) in corticotroph adenomas, with this correlation being particularly strong in functioning adenomas (r_s = 0.655; P < .001) .

  • Association with Tumor Subtype: PCSK1N expression is higher in silent corticotroph adenomas (SCAs) compared to functioning adenomas (FCAs) . SCAs are generally recognized to exhibit more aggressive clinical behavior, with rapid and invasive growth, tumor heterogeneity, and higher recurrence rates .

  • Molecular Mechanism Support: PCSK1N's role in ER stress responses may confer survival advantages to tumor cells by downregulating apoptosis, cell cycle, and senescence pathways . This biological mechanism provides a plausible explanation for how increased PCSK1N could contribute to more aggressive tumor behavior.

  • Correlation with Other Molecular Markers: PCSK1N expression negatively correlates with ER processing genes (CALR, GRP78, GRP94, and UGGT2) (-0.57 < r_s < -0.35; P < .05) , providing a molecular context for its role in tumor biology.

These findings collectively suggest that PCSK1N quantification could potentially serve as a valuable prognostic tool in the assessment of pituitary adenomas, particularly corticotroph tumors .

What experimental models are most appropriate for studying PCSK1N function?

Several experimental models have proven valuable for investigating PCSK1N function:

  • Cell Culture Systems:

    • AtT-20 mouse pituitary corticotroph tumor cell line: Widely used to study PCSK1N effects on POMC processing and ER stress responses

    • MIN6 pancreatic β-cell line: Valuable for studying PCSK1N's role in proinsulin processing

    • Human corticotroph tumor primary cultures: Provide the most physiologically relevant cellular context for human studies

  • Animal Models:

    • Pax6 R266Stop mutant mice: Display altered PCSK1N expression and PC1/3 activity, useful for studying regulatory mechanisms

    • PCSK1N knockout mice: Essential for understanding physiological roles of PCSK1N in vivo

  • Human Sample Analysis:

    • Corticotroph adenoma specimens: Enable correlation of PCSK1N expression with clinical parameters such as tumor size and hormonal status

    • Categorized samples: Comparing functioning corticotroph adenomas (FCAs) versus silent corticotroph adenomas (SCAs) provides insights into PCSK1N's role in tumor functionality

When designing experiments, researchers should consider combining multiple models to overcome limitations of each system . For instance, findings from cell lines should be validated in primary cultures or tissue samples whenever possible. Additionally, time-course studies are important when investigating ER stress responses, as the research shows distinct temporal patterns in PCSK1N expression following stress induction .

What techniques can accurately measure PC1/3 activity in relation to PCSK1N inhibition?

Accurately measuring PC1/3 activity under PCSK1N inhibition requires specialized methodologies:

  • Enzyme Activity Assays:

    • Fluorogenic substrate assays using synthetic peptides containing PC1/3 cleavage sites

    • In vitro reconstitution assays with purified components to measure direct inhibition kinetics

    • Inclusion of appropriate controls (PC1/3 inhibitors like dec-RVKR-cmk) to confirm specificity

  • C-Terminal Processing Assessment:

    • Monitor the conversion of the 87kDa form of PC1/3 to the more active 66kDa form via western blot analysis

    • Quantify the ratio of these forms as an indicator of PC1/3 activation status

  • Prohormone Processing Analysis:

    • Western blot analysis of prohormone to processed hormone ratios

    • ELISA or RIA methods to quantify processed hormones (ACTH, insulin)

    • RT-qPCR measurement of relevant processing genes including POMC, TPIT, and PCSK1

  • RNA Interference Approaches:

    • siRNA targeting of PCSK1N to assess the direct impact on PC1/3 activity

    • Monitoring changes in PC1/3 C-terminal cleavage and enzyme activity following PCSK1N knockdown

When implementing these assays, researchers should consider cell type-specific differences in PC1/3 and PCSK1N expression, account for potential compensatory mechanisms with prolonged PCSK1N manipulation, and include dose-response studies with varying PCSK1N concentrations to establish inhibition parameters .

How can researchers effectively separate PCSK1N's enzyme inhibitory function from its chaperone activities?

Distinguishing between PCSK1N's dual roles as a PC1/3 inhibitor and a molecular chaperone requires specialized experimental approaches:

  • Structure-Function Analysis:

    • Generate truncation or point mutants of PCSK1N that selectively impair either inhibitory or chaperone functions

    • Test these variants in parallel assays measuring PC1/3 inhibition and anti-aggregation activities

    • Map functional domains responsible for each activity

  • Specific Activity Assays:

    • Enzyme inhibition: In vitro assays with purified PC1/3 and PCSK1N, measuring POMC or proinsulin processing

    • Chaperone activity: Protein aggregation assays using model substrates prone to misfolding (e.g., β-amyloid)

    • Comparative analysis: Assessment of both activities under identical experimental conditions

  • Temporal Dynamics Analysis:

    • Time-course studies following ER stress induction to determine the sequence of PCSK1N's different activities

    • Comparison of PCSK1N expression timing with changes in PC1/3 activity and cellular stress markers

    • Correlation of temporal patterns with cellular outcomes (survival, hormone processing)

  • Genetic Manipulation Strategies:

    • Domain-specific mutations that selectively alter one function while preserving the other

    • Complementation experiments with domain-specific constructs in PCSK1N-depleted cells

    • Expression of competing peptides that selectively interfere with either function

Through these complementary approaches, researchers can dissect the relative contributions of PCSK1N's distinct functional activities in different cellular contexts and disease states . This distinction is particularly important when considering PCSK1N as a potential therapeutic target or biomarker.

How does PCSK1N expression affect prohormone processing in different cellular contexts?

The impact of PCSK1N on prohormone processing varies significantly across cellular contexts:

  • Corticotroph Cells:

    • High PCSK1N expression correlates with reduced POMC processing to ACTH

    • In silent corticotroph adenomas (SCAs), elevated PCSK1N likely contributes to diminished functional ACTH production

    • A negative correlation exists between PCSK1N expression and plasma cortisol levels (r_s = -0.356; P = .026), but not with ACTH levels, suggesting effects on ACTH bioactivity rather than total quantity

  • Pancreatic β-Cells:

    • PCSK1N inhibits PC1/3-mediated proinsulin processing

    • Pax6 mutation-induced PCSK1N overexpression compromises PC1/3 C-terminal cleavage and activity

    • This mechanism contributes to defective proinsulin processing observed in Pax6-deficient models

The cellular microenvironment, including ER stress levels, significantly influences PCSK1N's impact on prohormone processing . Under stress conditions, increased PCSK1N expression may initially impair prohormone processing but ultimately contribute to cellular adaptation and survival . This complex relationship highlights the importance of considering the broader cellular context when studying PCSK1N's functional effects on hormone processing pathways.

What molecular mechanisms link PCSK1N to tumor growth in pituitary adenomas?

Research has revealed a significant positive correlation between PCSK1N expression and tumor size in corticotroph adenomas (r_s = 0.645; P < .001), suggesting a potential role in tumor growth . This relationship appears to be mechanistically linked to several functions of PCSK1N:

  • ER Stress Response Modulation:

    • PCSK1N increases during ER stress, potentially as part of an adaptive response

    • This adaptation may confer survival advantages to corticotroph tumor cells

    • Experimental induction of ER stress (e.g., with 17-AAG) leads to increased PCSK1N expression and downregulation of apoptosis, cell cycle, and senescence pathways

  • Anti-aggregation Chaperone Function:

    • PCSK1N prevents protein aggregation in a dose-dependent manner

    • This function may protect tumor cells from proteotoxic stress

    • Enhanced proteostasis could support continued tumor cell proliferation

  • Hormone Processing Modulation:

    • Inhibition of PC1/3 by PCSK1N alters the hormone production profile of tumor cells

    • Changed hormonal environment may influence tumor cell behavior and surrounding tissue

The correlation between PCSK1N and tumor size is particularly strong in functioning corticotroph adenomas (FCAs) (r_s = 0.655; P < .001) . PCSK1N's negative correlation with ER processing genes (CALR, GRP78, GRP94, and UGGT2) further suggests its integration into broader cellular stress response pathways that collectively influence tumor behavior .

How do post-translational modifications affect PCSK1N function?

While the research on post-translational modifications (PTMs) of PCSK1N is still emerging, several important aspects have been identified:

  • Proteolytic Processing:

    • PCSK1N itself can undergo proteolytic processing, generating smaller peptides with potentially distinct functions

    • These cleaved forms may have different activities regarding PC1/3 inhibition and chaperone function

    • The balance between full-length and processed forms may be tissue-specific and condition-dependent

  • Subcellular Localization Effects:

    • PTMs can affect PCSK1N's trafficking through the secretory pathway

    • Proper localization is crucial for its interaction with PC1/3 and other binding partners

    • Stress conditions may alter PCSK1N's subcellular distribution pattern

  • Functional Modulation:

    • PTMs potentially alter PCSK1N's binding affinity for PC1/3

    • Modifications may differently affect enzyme inhibitory versus chaperone functions

    • Understanding these differential effects is crucial for accurately assessing PCSK1N's varied roles

Future research should employ mass spectrometry-based approaches to comprehensively map PCSK1N's PTM landscape across different tissues and disease states . Site-directed mutagenesis of potential modification sites could help establish the functional significance of specific PTMs.

What transcription factors regulate PCSK1N expression beyond Pax6?

While Pax6 has been established as a direct negative regulator of PCSK1N expression , several other transcription factors and regulatory elements likely contribute to its tissue-specific and condition-dependent expression:

  • ER Stress Response Factors:

    • Transcription factors activated during the unfolded protein response (UPR) may regulate PCSK1N

    • These include ATF6, XBP1, and ATF4, which are activated in response to different arms of the ER stress pathway

    • PCSK1N's role in stress response suggests its transcriptional regulation is integrated with cellular stress adaptation mechanisms

  • Neuroendocrine-Specific Factors:

    • Given PCSK1N's prominent expression in neuroendocrine tissues, transcription factors involved in neuroendocrine differentiation likely regulate its expression

    • These may include additional homeodomain factors related to Pax6

    • The strong negative correlation between TPIT and PCSK1N expression (r_s = -0.386; P = .005) suggests potential regulatory relationships

  • Developmental Regulators:

    • Transcription factors controlling pituitary and pancreatic development may influence PCSK1N expression

    • These regulatory relationships could persist in adult tissues and become dysregulated in disease states

Understanding these additional regulatory mechanisms would provide potential targets for experimental manipulation of PCSK1N expression and could explain tissue-specific expression patterns observed in both normal physiology and pathological conditions .

How can researchers effectively manipulate PCSK1N expression in experimental systems?

Several approaches can be employed to modulate PCSK1N expression in experimental systems:

  • RNA Interference (RNAi):

    • siRNA targeting PCSK1N mRNA for transient knockdown has been successfully used in MIN6 cells

    • shRNA constructs can provide more stable knockdown for long-term studies

    • Validated siRNA sequences are available in the literature for both human and mouse PCSK1N

  • CRISPR-Cas9 Gene Editing:

    • Complete knockout of PCSK1N for loss-of-function studies

    • Knock-in of specific mutations to study structure-function relationships

    • Conditional knockout systems for tissue-specific or inducible deletion

  • Overexpression Systems:

    • Plasmid-based overexpression of wild-type or mutant PCSK1N

    • Viral vectors for efficient delivery to various cell types

    • Inducible expression systems for temporal control

  • Indirect Approaches:

    • Manipulation of Pax6 expression to indirectly affect PCSK1N levels

    • ER stress induction (e.g., with 17-AAG) to increase PCSK1N expression

    • Small molecules targeting stress pathways to modify PCSK1N expression

When implementing these approaches, researchers should confirm knockdown/overexpression efficiency at both mRNA and protein levels, test for potential off-target effects, and include appropriate controls in all experiments . For cell-type specific studies, optimization of delivery methods based on transfection efficiency and cell viability is essential.

How does the genomic context of PCSK1N influence its expression patterns?

The genomic context of PCSK1N plays a crucial role in determining its expression patterns across tissues and in response to various stimuli:

  • Promoter Architecture:

    • The PCSK1N promoter contains binding sites for Pax6, as demonstrated through chromatin immunoprecipitation and electrophoretic mobility shift assays

    • Luciferase reporter analysis has confirmed the functional significance of these binding sites in regulating PCSK1N expression

    • Additional regulatory elements likely contribute to tissue-specific and stress-responsive expression patterns

  • Chromatin Environment:

    • The accessibility of the PCSK1N locus is influenced by the surrounding chromatin state

    • Tissue-specific chromatin marks may explain differential expression between cell types

    • Alterations in chromatin structure during cellular stress could contribute to stress-induced PCSK1N upregulation

  • Long-Range Interactions:

    • Enhancer-promoter interactions can regulate PCSK1N expression over considerable genomic distances

    • These interactions may be tissue-specific and developmentally regulated

    • Disruption of these interactions could contribute to dysregulated expression in disease states

  • Genomic Neighborhood:

    • Nearby genes may share regulatory elements with PCSK1N

    • Coordinated regulation with functionally related genes could occur through shared enhancers or insulator elements

    • Understanding this broader genomic context could reveal co-regulated gene networks

These genomic context factors collectively determine the baseline expression of PCSK1N in different tissues and its responsiveness to various physiological and pathological stimuli, including ER stress conditions .

How might PCSK1N be targeted therapeutically in pituitary tumors?

Given PCSK1N's association with tumor size and potential role in tumor cell survival, several therapeutic strategies could be developed:

  • RNA-based Approaches:

    • siRNA or antisense oligonucleotides targeting PCSK1N mRNA

    • Delivery systems specifically targeting pituitary tumor cells

    • Combination with conventional therapies to enhance treatment efficacy

  • Small Molecule Inhibitors:

    • Compounds that block PCSK1N-PC1/3 interaction

    • Molecules that selectively interfere with PCSK1N's anti-aggregation function while preserving normal prohormone processing

    • Structure-based drug design targeting specific functional domains

  • Stress Response Modulation:

    • Therapeutic agents that normalize ER stress responses in tumor cells

    • Combination approaches targeting both PCSK1N and other stress adaptation pathways

    • Drugs that convert cytoprotective stress responses to cytotoxic ones in tumor cells

  • Indirect Targeting Strategies:

    • Restoring normal Pax6 function to downregulate PCSK1N expression

    • Modulating upstream regulatory pathways that control PCSK1N expression

    • Targeting downstream effectors of PCSK1N's action in tumor cells

Development of such therapeutic approaches would require careful consideration of potential effects on normal tissues expressing PCSK1N and should be guided by comprehensive preclinical studies in relevant model systems . The strong correlation between PCSK1N expression and tumor size suggests that successful targeting could potentially reduce tumor growth by removing survival advantages conferred by elevated PCSK1N .

What methodological challenges exist in studying PCSK1N in human samples?

Several methodological challenges must be addressed when studying PCSK1N in human samples:

  • Sample Heterogeneity:

    • Pituitary tumors show considerable heterogeneity in cell composition and molecular profiles

    • SCAs and FCAs represent a continuum rather than entirely separate entities

    • Microdissection techniques may be needed to isolate specific cell populations

  • Reference Standards:

    • Lack of standardized protocols for PCSK1N quantification

    • Need for validated reference materials and controls

    • Importance of appropriate housekeeping genes for normalization (e.g., GAPDH and ALAS1)

  • Temporal Considerations:

    • ER stress responses show complex temporal dynamics

    • Single time-point measurements may not capture the full biological significance

    • Limited ability to perform time-course studies in human samples

  • Clinical Correlation Challenges:

    • Lack of standardized magnetic resonance imaging–based measurement of tumor volume

    • Variable assessment of hormone overproduction, particularly in SCAs

    • Need for longer-term follow-up to correlate PCSK1N levels with clinical outcomes

  • Technical Limitations:

    • Sample preservation affecting RNA and protein quality

    • Need for highly specific antibodies for PCSK1N detection

    • Challenges in measuring enzymatic activities in fixed or frozen tissues

Researchers should implement rigorous standardization practices, employ multiple complementary methodologies, and carefully document all relevant clinical parameters to address these challenges . Multi-center collaborative studies with standardized protocols could help overcome some of these limitations.

How can PCSK1N research findings be translated into clinical applications?

Translating PCSK1N research findings into clinical applications requires several strategic approaches:

  • Biomarker Development:

    • Standardized assays for PCSK1N quantification in tumor samples

    • Establishment of clinically relevant thresholds based on correlation with outcomes

    • Integration of PCSK1N into multi-marker panels for improved prognostic accuracy

    • Development of algorithms combining PCSK1N levels with clinical and radiological features

  • Therapeutic Target Validation:

    • Confirmation of PCSK1N's role in tumor growth through interventional studies

    • Preclinical testing of PCSK1N-targeting approaches in patient-derived tumor models

    • Identification of patient subgroups most likely to benefit from PCSK1N-targeted therapies

    • Development of companion diagnostics to guide treatment selection

  • Clinical Trial Design:

    • Incorporation of PCSK1N measurement in clinical trials of pituitary tumor therapies

    • Stratification of patients based on PCSK1N expression levels

    • Assessment of PCSK1N as a predictive marker for treatment response

    • Monitoring of PCSK1N levels during treatment as a potential response marker

  • Personalized Medicine Applications:

    • Integration of PCSK1N status into clinical decision-making algorithms

    • Development of treatment strategies tailored to PCSK1N expression levels

    • Combination approaches targeting PCSK1N alongside other molecular abnormalities

Product Science Overview

Introduction

Proprotein Convertase Subtilisin/Kexin Type 1 Inhibitor, commonly referred to as PCSK1N or proSAAS, is a protein encoded by the PCSK1N gene in humans . This protein plays a crucial role in the regulation of the neuroendocrine secretory pathway and is predominantly expressed in cells with a regulated secretory pathway, such as endocrine and neuroendocrine cells .

Gene and Protein Structure

The PCSK1N gene is located on the X chromosome (Xp11.23) in humans . The gene encodes a protein that undergoes further processing into multiple short peptides . The intact proSAAS protein, as well as its carboxy-terminal peptide containing the inhibitory hexapeptide LLRVKR, functions as an inhibitor of prohormone convertase 1/3 (PC1/3), which is responsible for the initial proteolytic cleavage of peptide precursors .

Function and Mechanism

ProSAAS is expressed largely in cells possessing a regulated secretory pathway, such as endocrine/neuroendocrine cells and neurons . The protein functions as an inhibitor of prohormone convertase 1/3 (PC1/3), which is crucial for the proteolytic cleavage of peptide precursors . This inhibition is significant because it regulates the processing of neuroendocrine peptide precursors, thereby influencing various physiological processes .

The central, unprocessed portion of the proSAAS protein may function as a neural- and endocrine-specific chaperone due to its potent ability to block the aggregation of beta-amyloid and alpha-synuclein in vitro, and to block oligomer cytotoxicity in cells . Recent data suggest that nigral proSAAS expression can block the deterioration of the striatonigral pathway in a synuclein rat model of Parkinson’s disease .

Clinical Significance

Differential expression of the PCSK1N gene has been associated with obesity . Additionally, the protein’s ability to inhibit the aggregation of beta-amyloid and alpha-synuclein suggests potential therapeutic applications in neurodegenerative diseases such as Alzheimer’s and Parkinson’s 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.