CAB39L Human

Calcium Binding Protein 39 Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Functional Mechanisms

CAB39L functions as a scaffold protein that stabilizes the LKB1-STRAD complex, enabling phosphorylation and activation of AMP-activated protein kinase (AMPK). This activation triggers downstream effects on:

  • PGC1α phosphorylation, enhancing mitochondrial biogenesis and oxidative phosphorylation

  • Metabolic reprogramming, reversing the Warburg effect by increasing oxygen consumption rate (OCR) and reducing extracellular acidification rate (ECAR)

  • Cell cycle regulation via p21/p27 upregulation and caspase-mediated apoptosis

Epigenetic silencing through promoter hypermethylation is a primary mechanism of CAB39L inactivation in cancers, correlating with advanced tumor stages and poor prognosis .

Gastric Cancer (GC)

FindingExperimental EvidenceOutcome
Growth suppressionOverexpression in AGS/BGC823/MKN45 cells46% reduction in cell viability (P<0.01)
Apoptosis inductionIncreased cleaved caspase-3/-7/-8 and PARP3.2-fold apoptosis increase vs controls
Metastasis inhibitionWound healing/Matrigel assays58% reduced migration in AGS cells (P<0.05)
In vivo efficacyOrthotopic mouse models62% tumor weight reduction (P<0.001)

Kidney Renal Clear Cell Carcinoma (KIRC)

Clinical ParameterLow CAB39L Expression (n=269)High CAB39L Expression (n=270)P-value
T3/T4 Stage73.6%25.9%<0.001
5-Year Survival48.3%82.1%<0.001
Metastasis Rate31.2%12.6%0.003

Data from TCGA analysis demonstrates CAB39L's diagnostic power (AUC=0.89) and prognostic value (HR=0.6, P=0.034) .

Therapeutic Implications

  • Metformin synergy: Enhances LKB1/CAB39L complex formation, restoring AMPK activation in CAB39L-deficient cells

  • Epigenetic targeting: Demethylating agents could reactivate CAB39L expression in 92.9% of methylated GC cell lines

  • Biomarker potential: CAB39L promoter methylation status predicts chemotherapy response in GC (OR=2.4, P=0.008)

Key Research Findings

  1. Methylation Analysis: 13/14 GC cell lines showed CAB39L hypermethylation, inversely correlated with mRNA expression (r=-0.81, P<0.001)

  2. Survival Impact: GC patients with CAB39L methylation had 3.2x higher mortality risk (95% CI 1.8-5.7)

  3. Metabolic Reprogramming: CAB39L overexpression increases mitochondrial complex I-IV expression by 2.1-3.8 fold

  4. Therapeutic Response: KIRC cells overexpressing CAB39L show 67% reduction in proliferation (P<0.01)

Product Specs

Introduction
Calcium-binding protein 39-like (CAB39L) shares similarities with MO25 and is present in the blood of approximately 50% of patients with a blood cancer affecting a specific type of white blood cell. CAB39L is believed to be involved in the development of cancer. Additionally, a protein called LKB1 becomes more active when it binds to a group of proteins that includes CAB39L.
Description
This version of CAB39L is made in bacteria and is a single chain of protein building blocks. It is not modified with sugars and contains 360 building blocks, including a tag for purification. It has a size of 41.5 kDa.
Physical Appearance
A clear and colorless liquid free from bacteria.
Formulation
This CAB39L protein solution is provided at a concentration of 0.5mg/ml in a specific buffer containing Tris-HCl (pH 8.0), NaCl, glycerol, and DTT.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
More than 95% pure based on SDS-PAGE analysis.
Synonyms
MO25-BETA, MO2L, MLAA-34, Mo25-like protein.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMKKMPLF SKSHKNPAEI VKILKDNLAI LEKQDKKTDK ASEEVSKSLQ AMKEILCGTN EKEPPTEAVA QLAQELYSSG LLVTLIADLQ LIDFEGKKDV TQIFNNILRR QIGTRSPTVE YISAHPHILF MLLKGYEAPQ IALRCGIMLR ECIRHEPLAK IILFSNQFRD FFKYVELSTF DIASDAFATF KDLLTRHKVL VADFLEQNYD TIFEDYEKLL QSENYVTKRQ SLKLLGELIL DRHNFAIMTK YISKPENLKL MMNLLRDKSP NIQFEAFHVF KVFVASPHKT QPIVEILLKN QPKLIEFLSS FQKERTDDEQ FADEKNYLIK QIRDLKKTAP.

Q&A

What is CAB39L and what is its primary function in human cells?

CAB39L (Calcium Binding Protein 39-Like) is a protein located on chromosome 13q14.2 that functions as the β isoform of CAB39. It serves as a scaffold protein involved in the activation of kinases, particularly through interaction with the LKB1-STRAD complex . This interaction leads to phosphorylation and activation of AMPKα/β, a critical regulator of cellular energy homeostasis and metabolism . In normal cells, CAB39L maintains proper metabolic function by promoting oxidative phosphorylation and mitochondrial biogenesis . Additionally, CAB39L has been reported to be involved in the reproductive cycle, though its primary role appears to be in metabolic regulation .

How is CAB39L expression regulated in normal human tissues?

CAB39L expression is primarily regulated at the epigenetic level through promoter methylation . In normal tissues such as gastric epithelium, the CAB39L promoter remains largely unmethylated, allowing for normal expression of the gene . Bisulfite genomic sequencing (BGS) analysis reveals that normal gastric tissues demonstrate low methylation levels at CpG sites in the CAB39L promoter and first exon . Analysis from various tissue samples shows that normal stomach tissue exhibits high CAB39L mRNA and protein expression compared to cancerous tissues . The regulatory mechanisms that control tissue-specific expression patterns of CAB39L in different normal human tissues remain an area requiring further investigation.

What methodologies are most effective for detecting CAB39L expression in human tissue samples?

Based on published research, several complementary methodologies have proven effective for detecting CAB39L expression:

  • mRNA detection: RT-PCR provides a sensitive approach for quantifying CAB39L transcript levels in cell lines and tissue samples

  • Protein detection: Western blot analysis using anti-CAB39L antibodies can confirm protein expression in both cell lysates and tissue homogenates

  • Tissue localization: Immunohistochemistry allows for cellular and subcellular localization of CAB39L protein in tissue sections, with particularly effective results in paired tumor/normal samples

  • Methylation analysis: For understanding regulatory mechanisms, methylation-specific PCR (MSP) and bisulfite genomic sequencing (BGS) provide insights into the epigenetic regulation of CAB39L expression

  • Protein interaction studies: Co-immunoprecipitation with anti-CAB39L or with tagged versions (e.g., Flag-tagged CAB39L) enables identification of binding partners

When studying ectopically expressed CAB39L, epitope-tagged versions facilitate detection and functional analysis through tag-specific antibodies .

What mechanisms lead to CAB39L silencing in various cancer types?

The predominant mechanism for CAB39L silencing in cancer is promoter hypermethylation . This epigenetic modification has been thoroughly documented through multiple methodologies:

  • Genome-wide screening: Infinium Human Methylation 450 BeadChip identified CpG sites in the CAB39L promoter region that were differentially methylated by over 45% (Δβ-value = 0.46) in gastric cancer cell lines compared to normal controls

  • Cell line verification: Methylation-specific PCR (MSP) demonstrated that 13 out of 14 (92.9%) gastric cancer cell lines exhibited promoter hypermethylation, with corresponding low mRNA expression

  • Detailed methylation mapping: Bisulfite genomic sequencing (BGS) confirmed dense methylation (average CpG methylation > 50%) in 8 out of 9 gastric cancer cell lines but not in normal tissues

  • Functional validation: Treatment with 5-Aza-2′-deoxycytidine (a DNA methyltransferase inhibitor) restored CAB39L expression in all 6 tested gastric cancer cell lines, confirming methylation as the silencing mechanism

  • Clinical correlation: Integrative analyses of The Cancer Genome Atlas (TCGA) data revealed an inverse correlation between CAB39L promoter methylation and mRNA expression in gastric cancer patients

Similar patterns of hypermethylation-induced silencing have been observed in other cancer types, including kidney renal clear cell carcinoma (KIRC) , establishing promoter hypermethylation as the primary mechanism of CAB39L inactivation across multiple cancer types.

How does CAB39L function as a tumor suppressor at the molecular level?

CAB39L exerts its tumor suppressive effects through multiple interconnected mechanisms :

  • Metabolic reprogramming: CAB39L reverses the Warburg effect in cancer cells by promoting oxidative phosphorylation over glycolysis, as evidenced by enhanced oxygen consumption rate and reduced extracellular acidification rate

  • Signaling pathway activation: CAB39L interacts with the LKB1-STRAD complex, leading to LKB1 phosphorylation and subsequent activation of AMPKα/β . This activation was confirmed by Phospho-Kinase Arrays, which identified AMPKα as the top kinase activated by CAB39L

  • Mitochondrial function enhancement: CAB39L-induced p-AMPK triggers PGC1α phosphorylation and increases expression of genes involved in mitochondrial respiration complexes

  • Cell cycle regulation: CAB39L expression induces cell cycle arrest, with knockdown studies showing CAB39L silencing promotes cell cycle progression from G1 to S phase

  • Apoptosis induction: Overexpression of CAB39L increases both early and late phase apoptosis, with corresponding increases in cleaved caspase-3, -7, -8, and PARP

  • Inhibition of migration and invasion: As a member of the calcium binding protein family, CAB39L influences cytoskeletal rearrangement and cell motility, with functional studies confirming its inhibitory effect on migration and invasion

These mechanisms collectively establish CAB39L as a multifunctional tumor suppressor that acts primarily through metabolic regulation via the LKB1-AMPK-PGC1α signaling axis .

What is the role of CAB39L in regulating the Warburg effect in cancer cells?

CAB39L functions as a critical regulator that reverses the Warburg effect in cancer cells . The Warburg effect represents a metabolic shift where cancer cells predominantly produce energy through aerobic glycolysis rather than the more efficient oxidative phosphorylation, even in the presence of oxygen.

CAB39L counters this metabolic reprogramming through several mechanisms:

  • Activation of energy sensing pathways: CAB39L interacts with the LKB1-STRAD complex to activate AMPK, the master regulator of cellular energy homeostasis

  • Promotion of mitochondrial function: RNAseq and gene set enrichment analysis revealed that CAB39L expression strongly correlates with oxidative phosphorylation and mitochondrial biogenesis pathways

  • Metabolic enzyme regulation: CAB39L-induced p-AMPK triggers PGC1α phosphorylation, a transcriptional coactivator that regulates genes involved in energy metabolism and mitochondrial function

  • Direct metabolic shift: Functional metabolic studies demonstrated that CAB39L overexpression enhanced oxygen consumption rate (OCR) and reduced extracellular acidification rate (ECAR), directly confirming a shift from glycolysis toward oxidative phosphorylation

  • Reversal of metabolic phenotype: CAB39L knockdown experiments showed the opposite effect, promoting a metabolic shift toward the Warburg phenotype

This anti-Warburg effect represents a key mechanism by which CAB39L exerts its tumor suppressive function, particularly in gastric cancer where metabolic dysfunction is a hallmark .

What experimental models best demonstrate the tumor suppressive effects of CAB39L?

Based on published research, several complementary experimental models effectively demonstrate CAB39L's tumor suppressive functions :

  • In vitro cell line models:

    • Gain-of-function: Stable CAB39L overexpression in gastric cancer cell lines (AGS, BGC823, MKN45) demonstrates reduced cell viability (via MTT assay), decreased colony formation capacity, increased apoptosis (flow cytometry), and reduced migration (wound healing assay)

    • Loss-of-function: siRNA knockdown in CAB39L-expressing MKN74 cells shows increased cell viability, enhanced colony formation, suppressed apoptosis, and cell cycle progression from G1 to S phase

    • Rescue experiments: Re-expression of CAB39L in MKN74-shCAB39L cells restores tumor-suppressive effects, confirming specificity

  • Orthotopic xenograft model:

    • This physiologically relevant in vivo model involves subcutaneous implantation of BGC823 cells stably expressing CAB39L or empty vector, followed by transplantation of tumor fragments into the stomach lining

    • This approach better reproduces the organ microenvironment compared to standard subcutaneous xenograft models

    • Results show significant reduction in tumor size and weight with CAB39L overexpression

    • Immunohistochemical analysis demonstrates reduced cell proliferation (Ki-67 staining) and increased apoptosis (TUNEL staining) in CAB39L-expressing tumors

  • Pharmacological validation models:

    • Treatment with metformin (AMPK activator) in CAB39L-silenced cells mimics CAB39L function by activating downstream AMPK signaling

    • Compound C (AMPK inhibitor) rescues the growth inhibitory effect of CAB39L overexpression

    • These models confirm the dependence of CAB39L's tumor suppressive effects on AMPK activation

The orthotopic xenograft model provides particularly compelling evidence due to its physiological relevance and demonstration of CAB39L's in vivo effects.

How do researchers effectively study the CAB39L-LKB1-AMPK signaling axis?

Studying the CAB39L-LKB1-AMPK signaling axis requires a comprehensive experimental approach :

  • Protein interaction analysis:

    • Co-immunoprecipitation (co-IP) to confirm physical interactions between CAB39L, LKB1, and STRAD components

    • Both forward (anti-CAB39L pulldown) and reverse (anti-LKB1 pulldown) co-IP validates the interaction specificity

    • Flag-tagged CAB39L constructs facilitate detection in cells with low endogenous expression

  • Signaling pathway activation assessment:

    • Phospho-Kinase Arrays profile the phosphorylation status of multiple cancer-related kinases simultaneously (43 in the referenced study)

    • Western blot analysis for phosphorylated forms of pathway components: p-LKB1, p-AMPKα, p-AMPKβ, and p-PGC1α

    • Immunohistochemical staining for phosphorylated proteins in xenograft tissues

  • Genetic manipulation approaches:

    • Gain-of-function: Stable overexpression of CAB39L to assess downstream pathway activation

    • Loss-of-function: siRNA or shRNA-mediated knockdown of CAB39L

    • Epistasis analysis: Knockdown of LKB1 in CAB39L-overexpressing cells demonstrates dependency of tumor suppressive effects on LKB1-AMPK pathway

  • Pharmacological intervention:

    • AMPK activators (metformin) selectively target cells with CAB39L silencing

    • AMPK inhibitors (compound C) rescue phenotypes induced by CAB39L overexpression

    • These approaches confirm the functional relevance of AMPK in mediating CAB39L effects

  • Downstream functional readouts:

    • Metabolic assays: Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) measurements

    • Transcriptional profiling: RNAseq followed by gene set enrichment analysis (GSEA)

    • Expression analysis of mitochondrial respiration complex genes

This multi-layered approach provides comprehensive insights into the mechanism and functional significance of the CAB39L-LKB1-AMPK signaling axis.

What assays best measure the metabolic effects of CAB39L modulation?

To comprehensively assess the metabolic effects of CAB39L modulation, researchers should employ complementary methodologies that measure different aspects of cellular metabolism :

The combination of OCR and ECAR measurements provides particularly valuable direct assessment of the metabolic shift between oxidative phosphorylation and glycolysis, which is central to understanding CAB39L's role in countering the Warburg effect .

How does CAB39L promoter methylation status correlate with patient prognosis?

CAB39L promoter hypermethylation has significant prognostic implications in cancer patients :

In gastric cancer, CAB39L promoter hypermethylation strongly correlates with poor clinical outcomes . This association likely reflects the functional consequences of CAB39L silencing, including metabolic reprogramming toward the Warburg phenotype, enhanced proliferation, reduced apoptosis, and increased migration/invasion capacity .

The clinical data supports CAB39L methylation as a potential prognostic biomarker for gastric cancer patients . This epigenetic alteration represents a promising addition to the molecular classification of gastric cancer, potentially helping to identify patients with more aggressive disease who might benefit from more intensive therapeutic approaches .

Similar prognostic associations have been observed in other cancer types, including kidney renal clear cell carcinoma (KIRC), where CAB39L has also been reported to possess diagnostic and prognostic value . This suggests that CAB39L methylation may serve as a broadly applicable biomarker across multiple cancer types.

Future studies focusing on larger patient cohorts with comprehensive clinical follow-up data will be important to validate and refine the prognostic utility of CAB39L methylation in various cancer types and stages.

What potential therapeutic strategies could target the CAB39L pathway in cancer?

Based on understanding CAB39L's tumor suppressive function, several therapeutic approaches show promise :

  • Epigenetic therapy to restore CAB39L expression:

    • DNA methyltransferase inhibitors (DNMTi) such as 5-Aza-2′-deoxycytidine can reactivate silenced CAB39L

    • Experimental evidence shows that 5-Aza treatment successfully restores CAB39L expression in multiple gastric cancer cell lines with promoter hypermethylation

    • Combination of DNMTi with histone deacetylase inhibitors might enhance reactivation efficiency

  • Metabolic therapy targeting the downstream AMPK pathway:

    • Metformin (an AMPK activator commonly used for type-2 diabetes) demonstrates selective efficacy against gastric cancer cells with CAB39L silencing

    • Research shows that "re-activation of AMPK using metformin can selectively target GC with transcriptional silencing of CAB39L"

    • This approach effectively bypasses the need for CAB39L expression by directly activating the downstream AMPK pathway

  • Combination therapies:

    • Pairing AMPK activators with conventional chemotherapeutics may enhance efficacy in tumors with CAB39L silencing

    • Targeting both the epigenetic silencing (with DNMTi) and metabolic consequences (with AMPK activators) could provide synergistic benefits

  • Stratified treatment approaches:

    • CAB39L methylation status could serve as a biomarker to identify patients likely to respond to AMPK-activating therapies

    • Experimental data demonstrates that control cells (with CAB39L silencing) were more sensitive to metformin compared to CAB39L-overexpressing cells

These approaches highlight the potential for both reactivating the silenced tumor suppressor and exploiting the consequences of its loss through targeting downstream pathways.

What are the optimal methodologies for analyzing CAB39L methylation in clinical samples?

For clinical implementation of CAB39L methylation analysis, researchers should consider these optimized methodological approaches :

  • Sample collection and preservation:

    • Fresh frozen tissue samples provide optimal DNA quality for methylation analysis

    • Paired tumor and adjacent normal tissue samples enable comparative analysis and normalization

    • Blood samples may provide less invasive options for methylation detection in circulating tumor DNA

  • Methylation analysis techniques:

    • Bisulfite genomic sequencing (BGS): Provides comprehensive coverage of CpG sites across the CAB39L promoter and first exon, revealing methylation density patterns

    • Methylation-specific PCR (MSP): Offers a rapid and sensitive approach for targeted CpG site analysis in larger sample cohorts

    • Quantitative methylation-specific PCR (qMSP): Enables quantitative assessment of methylation levels with higher throughput

    • Array-based approaches: Infinium Human Methylation arrays provide broader coverage and context within genome-wide methylation patterns

  • Analytical considerations:

    • Focus analysis on the functionally relevant CpG sites in the CAB39L promoter region and first exon

    • Consider methylation density (percentage of methylated CpG sites) rather than binary classification

    • Establish appropriate cut-off values based on correlation with expression and clinical outcomes

  • Validation approaches:

    • Correlate methylation results with CAB39L mRNA expression

    • Perform immunohistochemistry to confirm protein-level changes

    • Integrate methylation data with other clinical and molecular parameters for comprehensive analysis

  • Data interpretation:

    • Calculate differential methylation between tumor and normal tissues

    • Consider the inverse correlation between CAB39L promoter methylation and mRNA expression

    • Assess methylation in context of clinical parameters and patient outcomes

These methodologies enable robust assessment of CAB39L methylation status for potential use as a prognostic biomarker in clinical practice.

What genetic and epigenetic factors regulate CAB39L expression beyond promoter methylation?

While promoter methylation is the predominant mechanism regulating CAB39L expression, several additional regulatory mechanisms warrant investigation :

  • Histone modifications:

    • Research should investigate the role of histone marks (H3K4me3, H3K27me3, H3K9ac) at the CAB39L promoter

    • The interplay between DNA methylation and histone modifications in regulating CAB39L expression remains unexplored

    • Chromatin immunoprecipitation (ChIP) assays could identify specific histone marks associated with CAB39L activation or repression

  • Transcription factor networks:

    • Identification of transcription factors that regulate CAB39L expression under normal and pathological conditions

    • Analysis of transcription factor binding sites in the CAB39L promoter and their functional significance

    • Investigation of whether specific oncogenic signaling pathways suppress CAB39L transcription

  • microRNA regulation:

    • Prediction and validation of microRNAs that target CAB39L mRNA

    • Assessment of microRNA expression patterns that correlate with CAB39L downregulation in cancer

    • Functional studies to demonstrate direct microRNA-mediated regulation of CAB39L

  • Genomic alterations:

    • Comprehensive analysis of copy number variations affecting the CAB39L locus (13q14.2)

    • Screening for mutations in CAB39L coding sequences or regulatory regions

    • Evaluation of chromosomal rearrangements affecting CAB39L expression

  • Long non-coding RNAs:

    • Identification of lncRNAs that regulate CAB39L expression through cis or trans mechanisms

    • Investigation of chromatin remodeling complexes that may be recruited by lncRNAs to the CAB39L locus

Understanding these additional regulatory mechanisms could provide new insights into CAB39L dysregulation in cancer and potential therapeutic targets beyond DNA methylation.

How does CAB39L function across different cancer types and cellular contexts?

Current research has primarily established CAB39L's role in gastric cancer, with limited exploration in other cancer types . Future research should address these key questions:

  • Pan-cancer analysis:

    • Systematic evaluation of CAB39L expression, methylation, and mutation status across multiple cancer types

    • Comparison of CAB39L silencing mechanisms between different cancers

    • Assessment of whether CAB39L's tumor suppressive function is universal or context-dependent

  • Tissue-specific effects:

    • Investigation of CAB39L's normal physiological role in different tissues

    • Comparative analysis of CAB39L-regulated pathways in different cell types

    • Determination of tissue-specific binding partners that may modulate CAB39L function

  • Microenvironmental influences:

    • Examination of how tumor microenvironment factors (hypoxia, nutrient availability, pH) affect CAB39L expression and function

    • Analysis of CAB39L's role in mediating cancer cell interactions with stromal components

    • Investigation of CAB39L's impact on immune cell function within the tumor microenvironment

  • Metabolic diversity:

    • Characterization of CAB39L's metabolic effects across cancer types with diverse metabolic profiles

    • Analysis of how genetic background influences CAB39L-mediated metabolic reprogramming

    • Investigation of CAB39L's role in cancers with mutations in core metabolic enzymes

  • Metastatic progression:

    • Assessment of CAB39L's role in different stages of metastatic cascade

    • Comparison of CAB39L methylation between primary tumors and metastatic lesions

    • Evaluation of CAB39L as a biomarker for metastatic potential

Expanding research across these dimensions would provide a more comprehensive understanding of CAB39L's biological functions and clinical relevance in human cancer.

What novel therapeutic approaches could restore CAB39L function or target downstream pathways?

Based on understanding the CAB39L-LKB1-AMPK-PGC1α axis, several innovative therapeutic strategies could be explored :

  • Targeted epigenetic editing:

    • CRISPR-based approaches using catalytically inactive Cas9 (dCas9) fused to DNA demethylases to specifically demethylate the CAB39L promoter

    • Targeted delivery of epigenetic editing components using nanoparticles or other delivery systems

    • Combination of targeted demethylation with histone modification modifiers for enhanced reactivation

  • Novel AMPK activators:

    • Development of next-generation AMPK activators with improved specificity and pharmacokinetic properties compared to metformin

    • Screening for natural compounds that selectively activate AMPK in CAB39L-deficient cells

    • Design of activators targeting specific AMPK subunits most relevant to CAB39L's tumor suppressive functions

  • Metabolic vulnerability exploitation:

    • Identification of synthetic lethal interactions with CAB39L loss

    • Development of therapies targeting metabolic dependencies created by CAB39L silencing

    • Combination of glycolysis inhibitors with mitochondrial function modulators

  • LKB1-mimetic approaches:

    • Design of peptides or small molecules that mimic CAB39L's interaction with LKB1-STRAD complex

    • Development of strategies to stabilize or enhance LKB1 activity independent of CAB39L

    • Targeting of negative regulators of the LKB1 pathway

  • PGC1α-directed therapies:

    • Development of compounds that directly activate PGC1α, bypassing the need for upstream CAB39L-LKB1-AMPK activation

    • Targeting of PGC1α coactivator function to enhance mitochondrial biogenesis and oxidative phosphorylation

    • Modulation of post-translational modifications that regulate PGC1α activity

  • Combination approaches:

    • Pairing of CAB39L pathway-targeting agents with conventional chemotherapeutics

    • Development of rational combinations based on synthetic lethality principles

    • Integration with immunotherapeutic approaches to enhance anti-tumor immune responses

These innovative strategies could open new avenues for targeting the metabolic vulnerabilities associated with CAB39L loss in cancer.

Product Science Overview

Gene and Protein Structure

The CAB39L gene is located on chromosome 13 and encodes a protein that consists of 341 amino acids with a molecular mass of approximately 42 kDa . The recombinant form of this protein is typically produced in E. coli and includes a 20 amino acid His-Tag at the N-terminus to facilitate purification .

Biological Functions

CAB39L is involved in several key biological pathways:

  • Signal Transduction: It acts as a component of a complex that binds and activates the serine/threonine kinase STK11/LKB1. This complex is essential for stabilizing the interaction between CAB39/MO25 (CAB39/MO25alpha or CAB39L/MO25beta) and STK11/LKB1 .
  • Kinase Activation: CAB39L enables protein serine/threonine kinase activator activity, which is crucial for intracellular signal transduction and the regulation of various cellular processes .
Tissue Distribution and Expression

CAB39L is expressed in various tissues, with significant roles in the innate immune system and endochondral ossification pathways . It is also implicated in diseases such as monocytic leukemia and immunodeficiency 10 .

Clinical Significance

Mutations or dysregulation of the CAB39L gene have been associated with several diseases, including monocytic leukemia and immunodeficiency 10 . As a biomarker, it holds potential for diagnostic and therapeutic applications in these conditions.

Recombinant Protein Production

The recombinant form of CAB39L is produced using E. coli expression systems. The protein is purified using chromatographic techniques to ensure high purity and functionality . This recombinant protein is valuable for research purposes, including studies on protein-protein interactions, kinase activation, and signal transduction pathways.

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.