STAMBPL1 Human

STAM Binding Protein Like 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in Cancer Biology

STAMBPL1 has been implicated in two major oncogenic pathways: epithelial-to-mesenchymal transition (EMT) and angiogenesis.

Epithelial-to-Mesenchymal Transition (EMT)

STAMBPL1 promotes EMT by stabilizing the transcription factor SNAI1, a master regulator of mesenchymal traits. Key findings include:

  • Protein Stability: STAMBPL1 depletion shortens SNAI1’s half-life via proteasomal degradation .

  • Phenotypic Effects: Overexpression induces:

    • ↑ Vimentin (mesenchymal marker)

    • ↓ E-cadherin (epithelial marker)

    • Enhanced migratory capacity in cancer cells (e.g., A549, MCF-7) .

  • Prognostic Relevance: High STAMBPL1 expression correlates with poor survival in lung cancer patients .

Angiogenesis in Triple-Negative Breast Cancer (TNBC)

STAMBPL1 activates the GRHL3/HIF1A/VEGFA axis, driving hypoxia-independent angiogenesis:

  • Mechanism: Binds FOXO1 to enhance GRHL3 transcription, which directly activates HIF1A promoter .

  • Functional Impact:

    • ↑ HIF1α and VEGFA expression under normoxic conditions .

    • ↑ Proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) .

  • Therapeutic Targeting: Combined inhibition of FOXO1 (AS1842856) and VEGFR (apatinib) suppresses TNBC xenograft growth .

Antibody Validations

Polyclonal rabbit antibodies (e.g., 27315-1-AP) demonstrate broad utility:

ApplicationDetails
Western BlotDetects 50 kDa band in A549, HeLa, MCF-7, and human testis/liver/colon .
ImmunohistochemistryValidated in human liver and colon tissues; requires antigen retrieval (TE buffer pH 9.0) .
ELISANot explicitly tested but inferred from antibody class .

Experimental Models

  • Cell Lines: A549, HCC1806, HCC1937 (TNBC), MCF-7, HeLa .

  • In Vivo: Xenograft models show reduced tumor angiogenesis upon STAMBPL1 knockdown .

Regulatory Factors and Interactions

STAMBPL1 expression is modulated by environmental and genetic factors:

  • Chemical Interactions:

    • ↑ By aflatoxin B1, benzo[a]pyrene, ethanol .

    • ↓ By bisphenol A, doxorubicin, entinostat .

  • Genetic Links: Mutant p53 upregulates STAMBPL1, linking it to oncogenic signaling .

Product Specs

Introduction
STAM Binding Protein Like 1 (STAMBPL1) is a zinc metalloprotease that specifically cleaves 'Lys-63'-linked polyubiquitin chains. However, it does not cleave 'Lys-48'-linked polyubiquitin chains. Each subunit of STAMBPL1 binds to two zinc ions. The JAMM motif is essential for its protease activity. STAMBPL1 belongs to the peptidase M67C family and contains one MPN (JAB/Mov34) domain.
Description
Recombinant human STAMBPL1, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 458 amino acids (residues 1-436) and has a molecular weight of 52 kDa. The STAMBPL1 protein is fused to a 22 amino acid His-tag at its N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The STAMBPL1 solution has a concentration of 0.5 mg/ml and is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
ALMalpha, AMSH-FP, AMSH-LP, bA399O19.2, AMSH-like protease, STAM Binding Protein Like 1, STAMBPL1, AMSHLP, KIAA1373.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSQ SHMDQPFTVN SLKKLAAMPD HTDVSLSPEE RVRALSKLGC NITISEDITP RRYFRSGVEM ERMASVYLEE GNLENAFVLY NKFITLFVEK LPNHRDYQQC AVPEKQDIMK KLKEIAFPRT DELKNDLLKK YNVEYQEYLQ SKNKYKAEIL KKLEHQRLIE AERKRIAQMR QQQLESEQFL FFEDQLKKQE LARGQMRSQQ TSGLSEQIDG SALSCFSTHQ NNSLLNVFAD QPNKSDATNY ASHSPPVNRA LTPAATLSAV QNLVVEGLRC VVLPEDLCHK FLQLAESNTV RGIETCGILC GKLTHNEFTI THVIVPKQSA GPDYCDMENV EELFNVQDQH DLLTLGWIHT HPTQTAFLSS VDLHTHCSYQ LMLPEAIAIV CSPKHKDTGI FRLTNAGMLE VSACKKKGFH PHTKEPRLFS ICKHVLVKDI KIIVLDLR.

Q&A

What is STAMBPL1 and what is its primary function in human cells?

STAMBPL1, also known as AMSH-LP, belongs to the AMSH family of deubiquitinating enzymes. It functions as a zinc metalloprotease that specifically cleaves K63-linked polyubiquitin chains . Initially discovered as an AMSH family protein, STAMBPL1 can enhance interleukin-2-mediated induction of the oncogene c-myc . While originally identified as an endosomally associated protein that indirectly activates NF-κB signaling, recent research has revealed its broader roles in cancer progression through various molecular mechanisms including EMT regulation and protein stabilization .

How is STAMBPL1 expression regulated in normal versus cancer cells?

STAMBPL1 expression appears to be tightly regulated in normal cells but is frequently dysregulated in cancer. Research demonstrates that mutant p53 can regulate STAMBPL1 expression, providing a novel concept of oncogenic regulation of this deubiquitinase . In hepatocellular carcinoma (HCC), STAMBPL1 is significantly upregulated compared to normal tissues . The differential expression between normal and cancer tissues suggests complex transcriptional regulation mechanisms that may be exploited by cancer cells during tumor progression .

What are the major cellular pathways influenced by STAMBPL1 activity?

STAMBPL1 influences several critical cellular pathways:

  • The HIF1α/VEGFA axis through enhanced transcription of HIF1A

  • EMT program in multiple carcinomas, particularly through SNAI1 stabilization

  • WNT/PI3K/NF-κB signaling pathway via TRAF2 deubiquitination

  • Protein degradation pathways (both proteasomal and lysosomal) through its deubiquitinating activity

These pathways collectively contribute to cellular processes including angiogenesis, metastasis, and cancer cell survival, highlighting STAMBPL1's multifaceted role in cellular homeostasis and disease .

What evidence links STAMBPL1 to cancer progression and metastasis?

Multiple lines of evidence connect STAMBPL1 to cancer progression:

  • Expression correlation: STAMBPL1 is highly expressed in metastatic tissues compared to matched primary tumors from the same lung cancer patients .

  • Survival analysis: Kaplan-Meier analyses of TCGA data reveal that high STAMBPL1 expression predicts poor prognosis in cancer patients .

  • Functional studies: CRISPR-mediated gene knockout of STAMBPL1 leads to marked recovery of epithelial markers, SNAI1 destabilization, and impaired migratory capacity of cancer cells .

  • Clinical correlation: A significant STAMBPL1-SNAI1 co-signature was observed across multiple tumor types, further supporting its role in metastasis via EMT regulation .

These findings collectively demonstrate that STAMBPL1 contributes to cancer progression through multiple mechanisms, particularly by promoting EMT and metastatic potential .

How does STAMBPL1 contribute to the epithelial-mesenchymal transition (EMT) in cancer cells?

STAMBPL1 contributes to EMT through several interconnected mechanisms:

  • Stabilization of SNAI1 (Snail): STAMBPL1 affects the stability of this key EMT-inducing transcription factor by protecting it from proteasomal degradation. Cycloheximide chase experiments revealed that STAMBPL1 depletion was associated with shortened Snail protein half-life .

  • EMT marker regulation: STAMBPL1 expression reprograms cells toward a mesenchymal phenotype. Genetic depletion leads to recovery of epithelial markers while reducing mesenchymal characteristics .

  • Pathway modulation: STAMBPL1-mediated deubiquitination activates signaling pathways (including WNT/PI3K/NF-κB) that further promote EMT program activation .

  • Cancer cell-specific effects: While Snail plays a crucial role in triggering EMT, the correlation between STAMBPL1 expression and SNAIL increase was not observed in all EMT conditions, suggesting context-dependent mechanisms .

These findings establish STAMBPL1 as a critical regulator of the EMT program, predominantly through its effect on Snail stability and related signaling pathways .

What is the prognostic value of STAMBPL1 expression in cancer patients?

STAMBPL1 has demonstrated significant prognostic value across multiple cancer types:

  • Survival prediction: According to TCGA clinical data analysis for lung adenocarcinoma (LUAD), high STAMBPL1 expression predicts poor patient prognosis. Kaplan-Meier analyses using optimized cut-off values between upper and lower quantiles showed significant survival differences between low and high STAMBPL1 expression groups .

  • Metastatic potential indicator: STAMBPL1 is highly expressed in metastatic tissues compared to matched primary tumors, suggesting its potential as a biomarker for metastatic progression .

  • Multi-cancer relevance: A significant STAMBPL1-SNAI1 co-signature was observed across multiple tumor types beyond lung and breast cancers, indicating broader prognostic applications .

  • HCC-specific findings: In hepatocellular carcinoma, STAMBPL1 upregulation showed strong prognostic value, correlating with more aggressive disease .

These findings position STAMBPL1 as a valuable prognostic biomarker that could be incorporated into clinical decision-making for risk stratification and treatment planning .

What are the recommended techniques for studying STAMBPL1 expression in patient samples?

For studying STAMBPL1 expression in patient samples, researchers have successfully employed several complementary techniques:

  • Immunohistochemistry (IHC):

    • Protocols using anti-STAMBPL1 antibodies (e.g., HPA040202, Atlas Antibodies AB, diluted 1:600) with standard deparaffinization and antigen retrieval procedures

    • Automated staining using instruments like Autostainer 480 (Thermo Fisher Scientific) with 30-minute antibody incubation

    • Visualization with diaminobenzidine and counterstaining with Mayers hematoxylin

    • Digital slide scanning using systems like Aperio AT2

  • TCGA RNA-seq data analysis:

    • Retrieval of Illumina HiSeqV2, RSEM normalized RNA-seq gene expression data from databases like Broad Firebrowse TCGA

    • Log2 transformation of expression data

    • Correlation analysis with EMT markers such as vimentin (VIM) and E-cadherin (CDH1)

  • Correlation screening:

    • Computing Pearson correlation between STAMBPL1 and EMT markers using R packages like Hmisc

    • Visualization of correlation matrices via corrplot package

    • Selection of significant correlations using appropriate thresholds (e.g., correlation coefficient >0.2)

These approaches provide complementary data on STAMBPL1 expression at both protein and mRNA levels, enabling comprehensive assessment in patient samples .

What experimental approaches are effective for investigating the molecular mechanisms of STAMBPL1?

Several experimental approaches have proven effective for investigating STAMBPL1's molecular mechanisms:

  • Protein interaction studies:

    • Immunoprecipitation to identify binding partners (e.g., TRAF2, SNAI1)

    • Domain mapping to identify specific interaction regions (e.g., the 251-436 sites of STAMBPL1 interact with the 294-496 sites of TRAF2)

    • Proximity ligation assays to confirm interactions in cellular contexts

  • Functional depletion/overexpression:

    • CRISPR-mediated gene knockout for complete STAMBPL1 elimination

    • siRNA knockdown for transient depletion

    • Overexpression systems using plasmid constructs with wild-type or catalytically inactive STAMBPL1 mutants

  • Protein stability assays:

    • Cycloheximide chase experiments to assess protein half-life changes upon STAMBPL1 manipulation

    • Ubiquitination assays to detect K63-linked polyubiquitin chains on potential substrates

  • Transcriptional analysis:

    • RT-qPCR to measure effects on target gene expression

    • ChIP assays to investigate transcription factor binding at relevant promoters

These methodologies collectively enable detailed investigation of STAMBPL1's role in deubiquitination, protein stabilization, and pathway activation .

How can researchers effectively analyze STAMBPL1 data from public cancer databases?

Effective analysis of STAMBPL1 data from public cancer databases requires a systematic approach:

  • Data retrieval:

    • Access normalized RNA-seq data from databases like The Cancer Genome Atlas (TCGA) using platforms such as Broad Firebrowse or specialized R packages like TCGA2STAT

    • Focus on specific cancer types (e.g., LUAD for lung adenocarcinoma, BRCA for breast cancer) or analyze across multiple cancer types

  • Expression correlation analysis:

    • Calculate Pearson correlations between STAMBPL1 and markers of interest (e.g., EMT markers like VIM and CDH1)

    • Apply appropriate thresholds for significance (correlation coefficient >0.2)

    • Adjust p-values for multiple testing using methods like Benjamini-Hochberg adjustment

  • Survival analysis:

    • Perform Kaplan-Meier analyses using survival packages in R

    • Test multiple cut-off values between upper and lower quantiles based on log-rank tests

    • Select the best-performing threshold to determine low- and high-risk groups

  • Multi-cancer analysis:

    • Log2 transform expression data for comparability

    • Filter data based on TCGA barcodes to select only tumor samples

    • Assess co-expression patterns across different cancer types

These approaches enable robust, statistically sound analyses that can identify clinically and biologically relevant patterns of STAMBPL1 expression and correlation with disease outcomes .

How does STAMBPL1 regulate the stability and function of its target proteins?

STAMBPL1 regulates target proteins through its deubiquitinating activity with several distinct mechanisms:

  • K63-linked polyubiquitin chain cleavage:

    • STAMBPL1 specifically cleaves K63-linked polyubiquitin chains on target proteins

    • This deubiquitination prevents proteins from degradation by either proteasomal or lysosomal pathways

  • Substrate-specific stabilization:

    • TRAF2: STAMBPL1 interacts with TRAF2 and stabilizes it by removing K63-linked ubiquitin chains, resulting in enhanced WNT/PI3K/NF-κB signaling

    • SNAI1 (Snail): STAMBPL1 indirectly affects Snail stability, as demonstrated by cycloheximide chase experiments showing shortened Snail half-life upon STAMBPL1 depletion

  • Indirect degradation regulation:

    • STAMBPL1 can indirectly affect protein stability by targeting substrates to either proteasomal (e.g., HTLV-1 Tax oncoprotein) or lysosomal degradation (e.g., XIAP)

  • Transcriptional regulation:

    • STAMBPL1 can enhance transcription of certain genes, as demonstrated by its ability to promote HIF1A transcription, which subsequently activates the HIF1α/VEGFA axis

These diverse mechanisms allow STAMBPL1 to exert precise control over multiple cellular pathways through post-translational and transcriptional regulation of key signaling proteins .

What is the relationship between STAMBPL1 and the HIF1α/VEGFA signaling axis?

STAMBPL1 exhibits a crucial regulatory relationship with the HIF1α/VEGFA signaling axis:

  • Transcriptional upregulation:

    • STAMBPL1 promotes HIF1A transcription, as evidenced by experiments showing that STAMBPL1 knockdown significantly inhibits HIF1A transcription

    • Conversely, STAMBPL1 overexpression increases HIF1A transcription levels

  • Pathway activation:

    • Through its upregulation of HIF1α, STAMBPL1 subsequently enhances transcription of VEGFA, a key downstream target of HIF1α

    • STAMBPL1 overexpression directly increases VEGFA transcription, but this effect is blocked by HIF1α knockdown, confirming the pathway dependency

  • Functional consequences:

    • The STAMBPL1-mediated activation of the HIF1α/VEGFA axis likely contributes to enhanced angiogenesis in tumors

    • This pathway activation represents one of several mechanisms through which STAMBPL1 may promote cancer progression

These findings establish STAMBPL1 as an important upstream regulator of the HIF1α/VEGFA signaling axis, primarily through enhancement of HIF1A transcription rather than post-translational modification .

How does STAMBPL1 interact with TRAF2 and what are the downstream consequences?

STAMBPL1 interacts with TRAF2 through a specific binding mechanism with important downstream consequences:

  • Physical interaction:

    • STAMBPL1 directly interacts with TRAF2 protein

    • The interaction occurs specifically between the 251-436 amino acid sites of STAMBPL1 and the 294-496 sites of TRAF2

  • Deubiquitination activity:

    • STAMBPL1 functions as a deubiquitinase for TRAF2, removing K63-linked ubiquitin chains

    • This deubiquitination leads to stabilization of TRAF2 protein

  • Signaling activation:

    • Stabilized TRAF2 promotes translocation of P65 protein into the nucleus

    • This nuclear translocation activates the WNT/PI3K/NF-κB signaling pathway

  • Cancer-promoting effects:

    • The activated signaling cascade significantly promotes proliferation and metastasis in hepatocellular carcinoma cells

    • This mechanism provides one explanation for how STAMBPL1 upregulation contributes to aggressive cancer phenotypes

This STAMBPL1-TRAF2 interaction represents a critical molecular mechanism through which STAMBPL1 exerts its pro-oncogenic effects, linking deubiquitination activity directly to activation of cancer-promoting signaling pathways .

What are the methodological challenges in studying STAMBPL1 substrate specificity?

Studying STAMBPL1 substrate specificity presents several methodological challenges:

  • Distinguishing direct vs. indirect targets:

    • STAMBPL1 appears to have both direct deubiquitination targets (e.g., TRAF2) and indirectly affected proteins (e.g., SNAI1)

    • Separating direct enzymatic activity from secondary effects requires careful experimental design using catalytically inactive mutants as controls

  • Context-dependent activity:

    • Research indicates that STAMBPL1's effects on targets like Snail may be context-dependent and vary by EMT stimuli

    • This necessitates testing across multiple cell types and under various EMT-inducing conditions to fully characterize specificity patterns

  • Ubiquitin chain linkage recognition:

    • As STAMBPL1 specifically cleaves K63-linked polyubiquitin chains, distinguishing these from other ubiquitin linkages requires specialized antibodies or mass spectrometry approaches

    • Development of linkage-specific detection methods is critical for accurate substrate identification

  • Substrate validation:

    • Computational prediction of substrates must be validated through multiple orthogonal techniques

    • Confirmation requires both in vitro deubiquitination assays and cellular studies with careful controls

These challenges highlight the complexity of DUB substrate identification and emphasize the need for integrated approaches combining biochemical, cellular, and computational methods to accurately characterize STAMBPL1 substrate specificity .

How do different cancer genetic backgrounds influence STAMBPL1 function?

The influence of genetic backgrounds on STAMBPL1 function reveals complex interactions:

  • p53 mutational status:

    • Research suggests that mutant p53 can regulate STAMBPL1 expression, indicating that p53 status may significantly impact STAMBPL1 levels and activity

    • This relationship provides a novel concept of oncogenic regulation of a DUB and may explain elevated STAMBPL1 levels in p53-mutant cancers

  • Cancer-type specific effects:

    • While STAMBPL1-SNAI1 correlation is observed across multiple cancer types, the strength and clinical implications vary

    • In systematic analysis across 37 different TCGA cancer cohorts, significant cancer-type variations were observed in STAMBPL1-SNAI1 co-expression patterns

  • EMT context dependency:

    • Different cancer cells exhibit varying susceptibility to EMT stimuli

    • The relationship between STAMBPL1 expression and Snail increase was not observed in all EMT conditions, suggesting genetic background impacts the STAMBPL1-EMT relationship

  • Pathway integration:

    • The integration of STAMBPL1 into signaling networks appears to vary by cancer type and genetic background

    • HCC shows strong STAMBPL1-TRAF2-WNT/PI3K/NF-κB axis activation, while other cancers may utilize different STAMBPL1-dependent pathways

These observations highlight the importance of considering genetic context when studying STAMBPL1 function and suggest potential for targeted therapeutic approaches based on specific cancer genetic backgrounds .

What are the conflicting findings in STAMBPL1 research and how might they be reconciled?

Several apparent conflicts exist in STAMBPL1 research that require careful consideration:

  • Substrate specificity contradictions:

    • While STAMBPL1 is reported to specifically cleave K63-linked polyubiquitin chains, its effects on protein stability suggest potential involvement in K48-linked chain regulation as well

    • This apparent contradiction might be reconciled by considering indirect effects on E3 ligases or other DUBs that regulate K48-linked chains

  • Mechanism of Snail regulation:

    • Studies indicate STAMBPL1 affects Snail stability, but the exact mechanism remains unclear

    • Some data suggest direct deubiquitination, while other findings point to indirect regulation through intermediate factors

    • These conflicting observations may reflect cell-type specific regulatory mechanisms or technical differences in experimental approaches

  • EMT stimuli-dependent effects:

    • STAMBPL1's correlation with Snail expression varies depending on EMT triggers

    • This variability might be explained by the existence of multiple parallel pathways for EMT induction, with STAMBPL1 playing a dominant role only in specific contexts

  • Tissue-specific functions:

    • While STAMBPL1 promotes EMT in lung and breast cancers and proliferation in HCC, its role in other tissues requires further clarification

    • These apparent differences may reflect tissue-specific cofactor availability or alternative pathway wiring

Reconciliation of these conflicts requires integrated approaches combining quantitative proteomics, structural biology, and systems-level pathway analysis to build a comprehensive model of STAMBPL1 function across different cellular contexts .

What are the methodological considerations for developing STAMBPL1 as a prognostic biomarker?

Developing STAMBPL1 as a prognostic biomarker requires addressing several methodological considerations:

  • Threshold determination:

    • Current research shows that while STAMBPL1 expression has prognostic value, determining optimal cut-off thresholds is critical

    • Comprehensive analysis testing all possible thresholds between upper and lower quantiles based on log-rank tests is recommended to identify the most clinically relevant cut-points

  • Multi-marker approaches:

    • Evidence suggests that STAMBPL1-SNAI1 co-signature may have stronger prognostic value than STAMBPL1 alone

    • Development of multi-marker panels including STAMBPL1 and related proteins (e.g., EMT markers) may improve prognostic accuracy

  • Detection methodologies:

    • Standardization of detection protocols for both immunohistochemistry and mRNA expression analysis is essential

    • For IHC, detailed protocols using specific antibodies (e.g., HPA040202, Atlas Antibodies AB, diluted 1:600) with appropriate controls should be established

  • Cancer-specific validation:

    • While STAMBPL1 shows prognostic value across multiple cancer types, cancer-specific validation cohorts are needed

    • Different cut-off values and interpretation guidelines may be required for different cancer types

These considerations highlight the importance of rigorous statistical validation and standardized detection methodologies to establish STAMBPL1 as a reliable prognostic biomarker that can inform clinical decision-making .

How might targeting STAMBPL1 be used in combination with existing cancer therapies?

Targeting STAMBPL1 in combination therapy approaches presents several promising strategies:

  • Sensitization to traditional therapies:

    • EMT is associated with therapy resistance, and STAMBPL1 promotes EMT

    • Inhibiting STAMBPL1 could potentially reverse EMT, re-sensitizing cancer cells to conventional chemotherapies that are typically less effective against mesenchymal-like cells

  • Anti-angiogenic therapy enhancement:

    • STAMBPL1 activates the HIF1α/VEGFA axis, promoting angiogenesis

    • Combining STAMBPL1 inhibition with existing anti-angiogenic therapies (e.g., bevacizumab) might provide synergistic effects by targeting the pathway at multiple points

  • Pathway-specific combinations:

    • For cancers where STAMBPL1 activates WNT/PI3K/NF-κB signaling via TRAF2, combining STAMBPL1 inhibition with existing PI3K or NF-κB pathway inhibitors could enhance therapeutic efficacy

  • p53 status-guided approaches:

    • Given the relationship between mutant p53 and STAMBPL1 regulation, combination approaches could be tailored based on p53 status

    • In p53-mutant cancers, dual targeting of both mutant p53 and STAMBPL1 pathways might provide enhanced therapeutic benefit

These strategic combinations require preclinical validation but represent promising approaches to leverage STAMBPL1 biology for improved cancer treatment outcomes .

What statistical approaches are most appropriate for analyzing STAMBPL1 expression data in clinical cohorts?

Several statistical approaches have proven valuable for analyzing STAMBPL1 expression data in clinical cohorts:

  • Correlation analysis:

    • Pearson correlation analysis is effective for assessing relationships between STAMBPL1 and other markers (e.g., VIM, CDH1, SNAI1)

    • For non-normally distributed data, Spearman rank correlation may be more appropriate

    • Significance thresholds should be adjusted for multiple testing using methods like Benjamini-Hochberg adjustment

  • Survival analysis:

    • Kaplan-Meier analysis with log-rank tests remains the standard for evaluating prognostic significance

    • Testing multiple cut-off values between upper and lower quantiles helps identify optimal thresholds for stratifying patients

    • Cox proportional hazards models allow adjustment for clinical covariates

  • Multi-cancer analysis:

    • For comparing STAMBPL1 effects across cancer types, stratified analysis with cancer-specific adjustments is recommended

    • Meta-analytic approaches can synthesize findings across cohorts while accounting for between-cancer heterogeneity

  • Expression comparison:

    • For matched samples (e.g., primary vs. metastatic), paired statistical tests are more powerful

    • One-way analysis of variance with least significant differences is appropriate for multiple group comparisons

    • Statistical significance can be indicated at various levels (p < 0.05, 0.01, or 0.001) using asterisk notation (* for p < 0.05, ** for p < 0.01, and *** for p < 0.001)

These statistical approaches, when applied rigorously, enable robust analysis of STAMBPL1's clinical significance while accounting for the complexities of cancer datasets .

Product Science Overview

Structure and Function

STAMBPL1 encodes a zinc metalloprotease that specifically cleaves Lys63-linked polyubiquitin chains . This deubiquitinating enzyme (DUB) is involved in the removal of ubiquitin moieties from target proteins, a process crucial for maintaining protein homeostasis within the cell . Unlike other DUBs, STAMBPL1 does not cleave Lys48-linked polyubiquitin chains .

One of the primary functions of STAMBPL1 is its role as a positive regulator of the TORC1 signaling pathway. It mediates the deubiquitination of SESN2, thereby inhibiting SESN2’s interaction with the GATOR2 complex . This regulation is essential for cellular growth and metabolism.

Biological Significance

STAMBPL1 is involved in several critical cellular processes, including:

  1. Protein Degradation: By cleaving Lys63-linked polyubiquitin chains, STAMBPL1 helps in the degradation of misfolded or damaged proteins, thus maintaining cellular protein quality control .
  2. Cell Signaling: It plays a role in the TORC1 signaling pathway, which is vital for cell growth, proliferation, and metabolism .
  3. Epithelial-Mesenchymal Transition (EMT): Recent studies have shown that STAMBPL1 is involved in the EMT process, which is crucial for cancer progression and metastasis . It regulates the transcription factor SNAI1, affecting the migratory capacity of cancer cells .
Clinical Significance

STAMBPL1 has been associated with various diseases and conditions:

  1. Cancer: Overexpression of STAMBPL1 has been linked to the progression and dissemination of multiple carcinomas, including lung and breast cancers . Its expression is regulated by mutant p53, and its catalytic activity is required for the EMT process .
  2. Aortic Aneurysm: Mutations in the STAMBPL1 gene have been associated with aortic aneurysm, a condition characterized by the abnormal bulging of the aorta .
  3. Hermansky-Pudlak Syndrome 2: This rare genetic disorder, which affects multiple systems in the body, has also been linked to mutations in the STAMBPL1 gene .
Research and Therapeutic Potential

Given its role in critical cellular processes and disease progression, STAMBPL1 is a potential target for therapeutic interventions. Inhibitors or modulators of STAMBPL1 activity could be developed to treat cancers and other diseases associated with its dysregulation.

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 2025 TheBiotek. All Rights Reserved.