STK11 Human

Serine/Threonine Kinase 11 Human Recombinant
Shipped with Ice Packs
In Stock

Description

STK11 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 456 amino acids (1-433) and having a molecular mass of 51kDa.
STK11 is fused to a 23 amino acid His-Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
STK11 is a serine/threonine kinase responsible for regulating cell polarity and acting as a tumor suppressor. Mutations in the STK11 gene are associated with Peutz-Jeghers syndrome, an autosomal dominant disorder characterized by gastrointestinal polyps, pigmented macules on the skin and mouth, and an increased risk of developing various cancers.
Description
STK11 Human Recombinant is a non-glycosylated polypeptide chain produced in E. coli. This single chain contains 456 amino acids (1-433) with a molecular weight of 51kDa. For purification, the protein is tagged with a 23 amino acid His-Tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile filtered.
Formulation
The STK11 solution is supplied at a concentration of 0.5mg/ml in a buffer containing 10% glycerol and 20mM Tris-HCl at pH 8.0.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For long-term 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

Serine/threonine-protein kinase STK11, Liver kinase B1, LKB1, hLKB1, Renal carcinoma antigen NY-REN-19, STK11, LKB1, PJS

Source

Escherichia Coli.

Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMEVVDPQ QLGMFTEGEL MSVGMDTFIH RIDSTEVIYQ PRRKRAKLIG KYLMGDLLGE GSYGKVKEVL DSETLCRRAV KILKKKKLRR IPNGEANVKK EIQLLRRLRH KNVIQLVDVL YNEEKQKMYM VMEYCVCGMQ EMLDSVPEKR FPVCQAHGYF CQLIDGLEYL HSQGIVHKDI KPGNLLLTTG GTLKISDLGV AEALHPFAAD DTCRTSQGSP AFQPPEIANG LDTFSGFKVD IWSAGVTLYN ITTGLYPFEG DNIYKLFENI GKGSYAIPGD CGPPLSDLLK GMLEYEPAKR FSIRQIRQHS WFRKKHPPAE APVPIPPSPD TKDRWRSMTV VPYLEDLHGA DEDEDLFDIE DDIIYTQDFT VPGQVPEEEA SHNGQRRGLP KAVCMNGTEA AQLSTKSRAE GRAPNPARKA CSASSKIRRL SACKQQ

Q&A

What is STK11 and what is its role in human cells?

STK11, also known as LKB1 (Liver Kinase B1), functions as a critical tumor suppressor in human cells. It encodes a serine/threonine kinase that regulates various cellular processes including energy metabolism, cell polarity, and cell growth. The gene is located on chromosome 19 and consists of multiple exons that encode a protein with distinct functional domains. In normal cells, STK11 helps maintain cellular homeostasis by activating AMP-activated protein kinase (AMPK) and other related kinases when cellular energy levels are low . This activation leads to inhibition of energy-consuming processes and promotion of energy-producing ones, effectively preventing uncontrolled cell growth that could lead to cancer development.

How are STK11 mutations classified in cancer research?

STK11 mutations in cancer research are classified based on their functional impact and location within the gene. They are typically categorized as splice-site variants, missense variants, nonsense variants, or deletion/insertion variants. Splice-site variants, such as the four described in the provided research (V1: c.598-1G>A, V2: c.464+1G>T, V3: c.862+1G>A, and V4: c.465-2A>C), affect the normal splicing process of STK11 mRNA, often leading to aberrant protein production . Missense variants result in amino acid substitutions and may affect protein function to varying degrees. The nomenclature for these variants follows standard conventions, using c.SYNTAX for nucleotide changes, p.SYNTAX for protein changes, and g.SYNTAX for genomic coordinates, as illustrated in the comprehensive table of 28 missense variants assessed in the research study .

What experimental evidence confirms STK11's role as a tumor suppressor?

Experimental evidence confirming STK11's role as a tumor suppressor comes from multiple approaches. In functional studies, loss of STK11 expression or activity has been shown to promote cancer cell growth and survival. For instance, in NSCLC cell lines, restoration of functional STK11 in cells with STK11 mutations can inhibit tumor growth. The research on STK11 variants provides further evidence - when functional STK11 is absent due to mutations, cells show altered signaling pathways that contribute to cancer progression . Additionally, clinical observations reveal that patients with STK11 mutations tend to have poorer responses to certain therapies, particularly immunotherapies like anti-PD-1 treatment in KRAS-driven NSCLC, further supporting its tumor suppressor role in the context of cancer treatment .

What is the prevalence of STK11 mutations across different cancer types?

STK11 mutations are found across various cancer types, though their prevalence varies significantly. They are most frequently observed in lung adenocarcinoma, particularly in the context of KRAS-driven cancers. Approximately 15,000 patients are diagnosed annually with KRAS-driven, STK11-mutated NSCLC, representing a significant subset of lung cancer cases . Beyond lung cancer, STK11 mutations are also found in cervical, pancreatic, and melanoma tumors, albeit at lower frequencies. In the germline context, STK11 mutations are responsible for Peutz-Jeghers Syndrome, an inherited condition characterized by intestinal polyps and increased cancer risk. The ClinVar database documents numerous germline STK11 variants associated with this syndrome, some of which overlap with somatic variants found in cancers, as noted in the research where certain variants like R297S, D194Y, and G163R were flagged as pathogenic or likely pathogenic in germline studies .

How do specific STK11 splice-site variants affect mRNA processing and protein function?

STK11 splice-site variants significantly disrupt normal mRNA processing through diverse mechanisms, each with profound consequences for protein function. The research demonstrates that four different splice-site variants (V1-V4) all compromised STK11 function but through distinct molecular processes . For instance, variant V1 (c.598-1G>A) and V3 (c.862+1G>A) resulted in intron read-through and exon skipping, respectively, while variants V2 (c.464+1G>T) and V4 (c.465-2A>C) led to cryptic splice-site utilization . Particularly interesting was the V4 variant, which was extensively characterized in an engineered cell line. While quantitative RT-PCR revealed that the amount of STK11 mRNA produced by the V4 variant locus was essentially equivalent to that produced by the wild-type locus, western blot analysis showed no detectable STK11 protein in the V4 variant expressing cell line . This finding suggests that the 14 amino acid deletion resulting from utilization of the STK11 exon 4 cryptic splice-site likely renders the protein unstable, demonstrating how splice variants can affect not only mRNA processing but subsequent protein stability as well.

What methodologies are most effective for characterizing novel STK11 variants?

Characterizing novel STK11 variants requires a multi-faceted approach combining genomic, transcriptomic, and functional analyses. The research demonstrates the importance of integrating complementary methods rather than relying on any single approach. For genomic analysis, next-generation sequencing identifies the variants, while transcriptomic analysis using RT-PCR with strategic primer placement can detect aberrant splicing events, as shown with the four splice-site variants examined . Functional impact assessment requires protein expression studies (western blotting) to determine if stable protein is produced, as well as kinase activity assays to assess protein function if expressed. The researchers utilized complementary methods focusing first on STK11 kinase activity and second on a well-characterized STK11-dependent signaling pathway, namely p53-mediated transcriptional activation . Advanced techniques like CRISPR-Cas9 base editing to generate cell lines with specific STK11 variants provide powerful experimental systems, as demonstrated with the NCI-H441 cells engineered to harbor the V4 variant . Critically, the research highlights that in silico predictive algorithms alone are insufficient, as they often yield discordant results—only 6 of 28 variants showed uniform agreement across all prediction platforms .

How do STK11 mutations affect response to immunotherapy in KRAS-driven NSCLC?

STK11 mutations significantly impact immunotherapy response in KRAS-driven NSCLC, creating a distinct clinical subtype with therapeutic implications. The research explicitly states that "clinical data demonstrating patients with KRAS-driven NSCLC lacking functional STK11 respond poorly to anti-PD-1 mono-therapy" . This reduced efficacy of immunotherapy in STK11-mutated tumors occurs through several mechanisms. STK11 loss alters the tumor microenvironment, creating an immunosuppressive milieu characterized by reduced T-cell infiltration and function. Additionally, STK11 mutations affect cancer cell metabolism and stress responses, potentially rendering them less susceptible to immune-mediated killing. The research emphasizes that understanding the functional status of STK11 variants is critical for guiding oncologic care for the approximately 15,000 patients diagnosed annually with KRAS-driven, STK11-mutated NSCLC . This underscores the importance of accurate functional classification of STK11 variants beyond mere identification of mutations, as different variants may have variable effects on protein function and consequently on treatment response.

What are the limitations of current in silico prediction algorithms for STK11 variant assessment?

Current in silico prediction algorithms for STK11 variant assessment face significant limitations in accuracy and consistency. The research directly addresses this issue, noting that "application of these tools reveals they rarely yield concordant results with respect to pathogenicity predictions, evidenced by the 28 STK11 missense variants we evaluate herein" . This discordance creates substantial challenges for clinical interpretation. Among the 28 variants studied, only 6 demonstrated uniform agreement across all predictive platforms (G56W, G163R, P179R, D194Y, G242V and A397S) . While 15 variants showed majority agreement between predictions and experimental results, three variants resulted in an approximately equal split among predictive algorithms (R104G, H202R and R211Q), and two variants were predicted to behave exactly opposite to experimental findings (S31F and P275L) . These inconsistencies highlight the inherent limitations of algorithms that rely primarily on sequence conservation, structural predictions, or evolutionary constraints without accounting for protein-specific functional domains and interactions. Additionally, in silico tools are particularly challenged when predicting the effects of variants on splicing, as demonstrated by the research finding that "predicting splicing outcomes for splice-site variants based solely on primary sequence data remains an unconquered challenge" .

How can novel engineered cell models advance STK11 functional studies?

Novel engineered cell models represent a powerful approach for advancing STK11 functional studies by enabling precise manipulation and observation of variant effects in a controlled cellular context. The research demonstrates this through the successful generation of a clonal NCI-H441 cell line where both STK11 alleles harbored the V4 variant (c.465-2A>C) using CRISPR-Cas9 base editing technology . This engineered model allowed for direct comparison between wild-type and variant STK11 expression and function in an otherwise identical genetic background. Using this system, researchers made the unexpected discovery that despite equivalent mRNA production between wild-type and V4 variant cell lines, no STK11 protein was detected in the variant line, revealing that the predicted 14 amino acid deletion likely rendered the protein unstable . Such insights would be difficult to obtain from patient samples alone. Advanced engineered models can further incorporate reporter systems for real-time visualization of STK11-dependent signaling, allow for temporal control of variant expression, or recreate complex mutational landscapes observed in tumors (such as concurrent KRAS and STK11 mutations). These approaches provide mechanistic insights into how specific variants affect STK11 function, potentially helping resolve discrepancies between computational predictions and clinical observations.

How can CRISPR-Cas9 base editing be applied to model STK11 variants?

CRISPR-Cas9 base editing represents a cutting-edge approach for modeling STK11 variants with high precision and efficiency. The research successfully employed this technique to generate a clonal cell line where both STK11 alleles harbored the V4 variant (c.465-2A>C) . Unlike traditional CRISPR-Cas9 techniques that create double-strand breaks and rely on error-prone repair mechanisms, base editing allows for direct conversion of one nucleotide to another without DNA cleavage, reducing the risk of unwanted insertions or deletions. For STK11 variant modeling, researchers can design specific guide RNAs targeting the desired mutation site along with an appropriate base editor (cytosine or adenine base editor) depending on the desired nucleotide change. After transfection and selection, single-cell cloning and screening identify successfully edited clones. The research demonstrated this approach's power by confirming the edit through DNA sequencing (shown by a red arrow in Figure 3A) and validating the functional consequences through RT-PCR, which revealed the expected aberrant splicing patterns matching those observed in tumor samples with the same variant . This technique can be applied to model various STK11 variants, including missense mutations, splice-site alterations, and regulatory region changes, enabling systematic functional characterization of variants of uncertain significance identified in patient tumors.

What are reliable assays for measuring STK11 kinase activity?

Reliable assays for measuring STK11 kinase activity are essential for functionally characterizing STK11 variants. The research utilized complementary methods focusing on STK11 kinase activity and downstream signaling effects . For direct assessment of kinase activity, in vitro kinase assays using recombinant STK11 protein (often in complex with its regulatory partners STRAD and MO25) and appropriate substrates such as AMPK can measure phosphorylation efficiency through radioactive labeling or phospho-specific antibodies. Additionally, cellular assays measuring the phosphorylation status of direct STK11 substrates like AMPK (at Thr172) provide insights into kinase function in a more physiological context. The research also assessed a well-characterized STK11-dependent signaling pathway, namely p53-mediated transcriptional activation, as an indirect measure of STK11 function . This approach recognizes that kinase activity alone may not capture all aspects of STK11 functionality, as some mutations may affect protein-protein interactions or subcellular localization while preserving catalytic activity. Combining multiple assays provides a more comprehensive assessment of variant impact, as demonstrated by the research's approach of using complementary methods to categorize variants. Western blotting for STK11 protein expression should accompany kinase assays to distinguish between variants that affect protein stability versus those that specifically impair catalytic function.

How can RT-PCR be optimized to detect aberrant splicing in STK11 variant studies?

Optimizing RT-PCR for detecting aberrant splicing in STK11 variant studies requires strategic primer design and careful experimental validation. The research demonstrates several key principles in their approach to characterizing splice-site variants. First, they designed multiple primer pairs spanning different exon junctions to capture various potential splicing outcomes. For instance, when analyzing the V4 variant, they detected two distinct RT-PCR products: an "upper band" resulting from cryptic splice-site utilization and a "lower band" representing exon 4 skipping . Second, they employed both standard RT-PCR for qualitative assessment of splicing patterns and quantitative RT-PCR with primers targeting specific exon junctions (such as STK11 exons 2/3 and 6/7) to measure relative abundance of different transcript variants . Third, they sequenced the aberrant RT-PCR products to precisely characterize the molecular nature of the splicing alterations, confirming that the bands matched perfectly with the amplicons identified from corresponding tumors harboring the same variant . For optimal detection, researchers should design primers in exons flanking the variant of interest, ensuring they are at sufficient distance to distinguish normal from aberrant products on gel electrophoresis. Controls should include wild-type samples and, ideally, positive controls with known splicing alterations. Additionally, nested PCR approaches may enhance sensitivity for detecting low-abundance aberrant transcripts in heterozygous samples.

What approaches can detect STK11 protein expression and stability differences between variants?

Detecting STK11 protein expression and stability differences between variants requires a combination of biochemical and cellular approaches. Western blotting with antibodies targeting different regions of STK11 provides the foundation for these analyses, as demonstrated in the research where they used an antibody targeting the STK11 N-terminus to reveal complete absence of STK11 protein in the V4 variant expressing cell line despite equivalent mRNA levels to wild-type . This unexpected finding highlighted that the 14 amino acid deletion resulting from cryptic splice-site utilization likely rendered the protein unstable. For comprehensive analysis, researchers should consider several specialized techniques: pulse-chase experiments using metabolic labeling to directly measure protein half-life; proteasome inhibition studies to determine if variant proteins undergo accelerated degradation; co-immunoprecipitation assays to assess interactions with stabilizing partners like STRAD and MO25; and subcellular fractionation to examine potential alterations in protein localization that might affect function or stability. Additionally, expression of epitope-tagged wild-type and variant STK11 under controlled conditions (such as inducible expression systems) allows for normalization of transcription rates to focus specifically on post-transcriptional differences. For variants affecting protein folding, techniques like limited proteolysis can reveal structural alterations that might compromise stability, while thermal shift assays can quantify differences in protein stability between wild-type and variant STK11.

How should discordant results between in silico predictions and experimental data be resolved?

Resolving discordant results between in silico predictions and experimental data for STK11 variants requires a systematic, evidence-based approach prioritizing experimental validation. The research clearly demonstrates this issue, finding that among 28 STK11 missense variants, only 6 showed uniform agreement across all predictive platforms, while certain variants like S31F and P275L were predicted to behave exactly opposite to experimental findings . When facing such discrepancies, researchers should first verify experimental methodology to ensure reliable results, including appropriate controls and replication. Next, they should explore whether the variant affects protein aspects not captured by most prediction algorithms, such as post-translational modifications, protein-protein interactions, or subcellular localization. Integration of multiple prediction tools, rather than reliance on any single algorithm, provides a more balanced computational assessment. The research demonstrates this by evaluating 22 different in silico algorithms . For critical variants, orthogonal experimental approaches should be employed to confirm findings—for example, combining kinase activity assays with cellular phenotype assessments and structural studies. Finally, when available, clinical data should inform interpretation; the research noted that their functional assessments for four variants agreed with ClinVar categorizations based on germline studies . In scientific communications, researchers should transparently report both predicted and experimental outcomes, highlighting discrepancies to improve future algorithm development.

What criteria should be used to classify a novel STK11 variant as pathogenic?

Classifying novel STK11 variants as pathogenic requires integration of multiple evidence types in a systematic framework. Based on the research approach, several critical criteria emerge. First, functional studies demonstrating significant impact on protein activity are essential—the researchers utilized complementary methods examining both STK11 kinase activity and downstream signaling through p53-mediated transcriptional activation to categorize variants . Second, protein expression and stability must be assessed, as some variants (like the V4 splice variant) render the protein unstable despite normal mRNA production . Third, aberrant splicing should be evaluated for variants near exon-intron boundaries through RT-PCR and sequencing, as demonstrated for the four splice-site variants in the study . Fourth, conservation analysis across species and within the protein kinase family provides evolutionary context for the variant position. Fifth, when available, clinical association data strengthens classification—the research noted agreement between their functional assessments and ClinVar categorizations for variants previously reported in germline studies . Sixth, structural analysis predicting how the variant might disrupt protein folding, catalytic activity, or interactions with regulatory partners adds mechanistic insight. Finally, co-segregation with disease in family studies (for germline variants) or recurrence in multiple independent tumors (for somatic variants) supports pathogenicity. When these criteria collectively point toward functional impairment, a variant can be classified as likely pathogenic or pathogenic.

How can STK11 research findings be translated into clinical applications?

Translating STK11 research findings into clinical applications requires bridging the gap between molecular characterization and patient care through several strategic approaches. First, developing clinical assays for STK11 functional status assessment, rather than merely detecting mutations, would provide more accurate prognostic and predictive information. The research emphasizes this need by demonstrating that different variants have variable effects on protein function, which cannot be reliably predicted by computational methods alone . Second, creating a comprehensive, curated database of functionally characterized STK11 variants would support clinical interpretation of genomic testing results. The research contributes to this goal by functionally characterizing 28 missense and 4 splice-site variants . Third, stratifying clinical trials by STK11 functional status, particularly in KRAS-driven cancers, could identify patient subgroups more likely to benefit from specific therapies. The research highlights the clinical relevance of this approach, noting that "patients with KRAS-driven NSCLC lacking functional STK11 respond poorly to anti-PD-1 mono-therapy" . Fourth, developing STK11 pathway-targeted therapies or synthetic lethal approaches could address the current lack of effective options for STK11-deficient tumors. Fifth, implementing STK11 testing in routine clinical care for relevant cancer types, particularly lung adenocarcinoma, would improve treatment selection. Finally, creating decision support tools for clinicians that integrate STK11 functional data with other biomarkers would enhance personalized treatment planning, advancing the goal of comprehensive personalized genomic medicine that the research identifies as currently limited by challenges in variant interpretation .

How do STK11 variants affect interactions with upstream regulators and downstream effectors?

STK11 variants can disrupt critical protein interactions throughout its signaling network, affecting both upstream regulators and downstream effectors. While the specific research results focus primarily on functional outcomes rather than detailed interaction analyses, broader STK11 biology provides context for understanding these effects. Upstream, STK11 forms a heterotrimeric complex with STRAD (STE20-related kinase adapter protein) and MO25 (mouse protein 25), which activates STK11 kinase activity and influences its subcellular localization. Variants in STK11's protein-binding domains may impair formation of this complex, preventing proper activation even if the catalytic domain remains intact. Downstream, STK11 phosphorylates and activates AMPK and at least 12 other AMPK-related kinases, creating a complex signaling network. The research assessed STK11's role in p53-mediated transcriptional activation as one downstream pathway , but variants could differently affect various effector pathways depending on their location within STK11. Kinase domain mutations typically disrupt all downstream signaling, while mutations in specific substrate-recognition regions might selectively impair certain interactions while preserving others. Additionally, some STK11 variants may create neomorphic functions—new protein interactions not present in the wild-type protein—though such effects are challenging to predict and require specialized interactome studies. Understanding these interaction disruptions is critical for developing targeted therapeutic approaches and understanding the diverse phenotypic consequences of different STK11 variants.

What are the emerging technologies that may enhance future STK11 variant classification?

Emerging technologies promise to revolutionize STK11 variant classification by enabling higher-throughput, more precise functional characterization. First, multiplex functional assays using CRISPR-based variant libraries can simultaneously assess hundreds to thousands of STK11 variants in parallel, dramatically accelerating the pace of functional classification beyond the 32 variants characterized in the current research . Second, advanced structural biology techniques including cryo-electron microscopy and AlphaFold-based protein structure prediction can provide atomic-level insights into how specific variants disrupt STK11 structure, potentially improving predictive accuracy. Third, patient-derived organoids and xenografts offer more physiologically relevant systems than cell lines for evaluating variant effects on tumor biology and treatment response. Fourth, single-cell genomic and transcriptomic analyses can reveal how STK11 variants affect cellular heterogeneity within tumors and influence the tumor microenvironment. Fifth, liquid biopsy technologies enable longitudinal monitoring of STK11 variant allele frequencies during treatment, potentially correlating functional variant classes with treatment outcomes. Sixth, machine learning approaches integrating multiple data types (structural, functional, clinical) could develop more accurate prediction algorithms than the current in silico tools, which showed significant limitations in the research with only 6 of 28 variants demonstrating uniform agreement across prediction platforms . Finally, high-throughput splicing assays like massively parallel reporter assays can systematically characterize how variants affect mRNA processing, addressing the research finding that "predicting splicing outcomes for splice-site variants based solely on primary sequence data remains an unconquered challenge" .

Product Science Overview

Gene and Protein Structure

The STK11 gene is located on chromosome 19 in humans . The protein encoded by this gene is a serine/threonine kinase, which means it phosphorylates serine and threonine residues on target proteins. This phosphorylation is essential for the activation or deactivation of these target proteins, thereby regulating their function.

Function and Mechanism

STK11/LKB1 is a primary upstream kinase of the AMP-activated protein kinase (AMPK) family . It phosphorylates the T-loop of AMPK family proteins, promoting their activity. This phosphorylation is crucial for maintaining cellular energy homeostasis, especially under conditions of low energy . Some of the key processes regulated by STK11 include:

  • Cell Metabolism: STK11 regulates glucose homeostasis in the liver and activates autophagy during nutrient deprivation .
  • Cell Polarity: It maintains cell polarity, which is vital for the proper organization and function of cells .
  • Tumor Suppression: STK11 acts as a tumor suppressor by inhibiting signaling pathways that promote cell growth and proliferation when energy levels are low .
Clinical Significance

Mutations in the STK11 gene are associated with several diseases, including:

  • Peutz-Jeghers Syndrome: An autosomal dominant disorder characterized by the development of benign polyps in the gastrointestinal tract and an increased risk of various cancers .
  • Cancers: Mutations in STK11 are linked to skin, pancreatic, and testicular cancers .
Research and Therapeutic Potential

Given its role in regulating critical cellular processes, STK11 is a significant focus of research. Understanding its function and regulation can provide insights into developing therapeutic strategies for diseases associated with its dysfunction, particularly cancer.

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.