STK16 Human

Serine/Threonine Kinase 16 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

STK16 regulates multiple cellular processes:

  • Golgi Structure Maintenance: Binds actin directly to stabilize Golgi architecture. Knockdown causes Golgi fragmentation and disrupts secretory pathways .

  • Cell Cycle Control: Modulates mitotic entry, prometaphase progression, and cytokinesis. Inhibition delays mitosis and increases binucleated cells .

  • TGF-β Signaling: Acts as TGF-β-stimulated factor 1 (TSF-1), influencing stromal-epithelial interactions .

  • Constitutive Secretion: Phosphorylates substrates like WDR1 to regulate vesicle trafficking .

Enzymatic Activity & Regulation

STK16 exhibits constitutive kinase activity due to its unique activation loop architecture . Key regulatory mechanisms include:

  • Autophosphorylation: Tyr198 is essential for kinase activity, Golgi localization, and function. Mutations (Y198A/Y198E) impair these properties .

  • Fatty Acylation: Myristoylation/palmitoylation at N-terminal residues (Gly2, Cys6/8) anchor it to membranes .

  • Degradation: KCTD17 mediates ubiquitin-proteasome degradation, with protein levels fluctuating in circadian rhythms .

Key Research Findings

Recent studies reveal novel insights:

Study FocusMethodologyKey OutcomeSource
Actin Dynamics & Golgi IntegrityRNAi knockdown + kinase inhibitionSTK16 stabilizes actin polymers; depletion causes Golgi fragmentation
Degradation MechanismCo-immunoprecipitation assaysKCTD17 binds STK16, promoting its degradation in dark-phase hypothalamus
Substrate IdentificationPhospho-proteomics screenIdentified 28 potential substrates (e.g., WDR1) regulating secretion
Structural AnalysisMutagenesis (T185A/S197A/Y198A variants)Tyr198 autophosphorylation is critical for catalytic activity and localization

Clinical & Pharmacological Implications

While STK16 is underexplored therapeutically, its roles suggest potential applications:

  • Cancer Research: Overexpression in epithelial tissues links it to mammary gland morphogenesis and TGF-β signaling .

  • Neuroendocrine Regulation: Circadian degradation patterns in the hypothalamus imply roles in metabolic or sleep disorders .

  • Drug Development: Small-molecule inhibitors targeting its kinase domain could modulate secretory pathways .

Recombinant STK16 Production

Commercial STK16 Human Recombinant (e.g., Prospec Bio PKA-032) is expressed in E. coli with the following properties:

  • Purity: >90% by SDS-PAGE

  • Formulation: 1 mg/mL in 20 mM Tris-HCl (pH 8.0), 20% glycerol, 0.1M NaCl, 1mM DTT

  • Applications: In vitro kinase assays, substrate phosphorylation studies

Future Directions

Current gaps include:

  • Identification of physiological substrates beyond WDR1 and 4EBP1 .

  • Mechanistic links between STK16-mediated actin regulation and mitotic defects .

  • Tissue-specific knockout models to validate roles in development and disease .

Product Specs

Introduction
Serine/threonine-protein kinase 16 (STK16) is a membrane-associated protein kinase that adds phosphate groups to serine and threonine residues. It plays a role in the movement of secretory vesicles and intracellular signaling. Additionally, STK16 might be involved in regulating how stromal and epithelial cells interact during the formation of ducts in the mammary gland. While STK16 can add a phosphate group to itself at a Tyrosine residue, it's unclear if it functions as a tyrosine-protein kinase for other proteins. STK16 may also be part of the TGF-beta signaling pathway.
Description
Recombinant human STK16, produced in E. coli, is a single polypeptide chain without any sugar modifications. It consists of 329 amino acids (specifically, amino acids 1 to 305) and has a molecular weight of 37.2 kDa. A 24-amino acid His-tag is attached to the N-terminus of STK16. The protein is purified using specialized chromatographic methods.
Physical Appearance
A clear and sterile solution.
Formulation
The STK16 protein is provided in a solution at a concentration of 1mg/ml. The solution also contains 20mM Tris-HCl buffer (pH 8.0), 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), keep the vial at 4°C. For longer periods, store the protein at -20°C. Adding a carrier protein (like HSA or BSA) to a final concentration of 0.1% is recommended for long-term storage. Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Serine/threonine-protein kinase 16, Myristoylated and palmitoylated serine/threonine-protein kinase, MPSK, Protein kinase PKL12, TGF-beta-stimulated factor 1, TSF-1, Tyrosine-protein kinase STK16, hPSK, STK16, MPSK1, PKL12, TSF1, KRCT.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMGHALC VCSRGTVIID NKRYLFIQKL GEGGFSYVDL VEGLHDGHFY ALKRILCHEQ QDREEAQREA DMHRLFNHPN ILRLVAYCLR ERGAKHEAWL LLPFFKRGTL WNEIERLKDK GNFLTEDQIL WLLLGICRGL EAIHAKGYAH RDLKPTNILL GDEGQPVLMD LGSMNQACIH VEGSRQALTL QDWAAQRCTI SYRAPELFSV QSHCVIDERT DVWSLGCVLY AMMFGEGPYD MVFQKGDSVA LAVQNQLSIP QSPRHSSALR QLLNSMMTVD PHQRPHIPLL LSQLEALQPP APGQHTTQI.

Q&A

What is STK16 and where is it located in the human genome?

STK16 is a serine/threonine protein kinase with a complete kinase catalytic domain, characterized by a very short N-terminal domain and a brief C-terminus. The human STK16 gene maps to chromosome 2q35 . Unlike many other kinases, STK16 has a compact structure that contributes to its specific functions in cellular processes including TGF-β signaling, protein trafficking, and cell cycle regulation .

To investigate STK16's genomic characteristics, researchers typically employ PCR-based genotyping, DNA sequencing, and chromosome mapping. For gene expression analysis, quantitative real-time PCR (qRT-PCR) is recommended to measure STK16 mRNA levels across different experimental conditions, though it's worth noting that STK16's effects in cancer appear to be primarily post-translational rather than transcriptional .

What is the subcellular localization of STK16 and how does this relate to its function?

STK16 demonstrates dual localization within the cell, primarily found at the cell membrane and the Golgi apparatus throughout the cell cycle . This distinct localization pattern is functionally significant and regulated by its autophosphorylation status, particularly at tyrosine 198 . When this autophosphorylation site is mutated, both the Golgi and membrane localization of STK16 are abolished .

The protein's strategic positioning at the Golgi enables its participation in:

  • Trans-Golgi Network (TGN) protein secretion and sorting

  • Golgi apparatus assembly regulation

  • Cell cycle regulation

  • Phosphorylation of target proteins such as c-MYC

For investigating STK16 localization, immunofluorescence microscopy with specific antibodies against STK16 combined with Golgi markers provides the most reliable visualization. Western blotting of subcellular fractions can biochemically confirm STK16's presence in specific cellular compartments.

What signaling pathways involve STK16 in human cells?

STK16 participates in several crucial cellular signaling pathways that impact various physiological and pathological processes. Based on current research, STK16 is involved in:

  • c-MYC signaling pathway: STK16 directly phosphorylates c-MYC at serine 452, preventing its degradation via the ubiquitin-proteasome pathway and thereby enhancing c-MYC-dependent gene expression . This represents a key mechanism in colorectal cancer progression.

  • TGF-β signaling pathway: STK16 contributes to the activation of this pathway, which regulates cell growth, differentiation, and immune responses .

  • Vesicular trafficking pathways: Through its Golgi localization, STK16 participates in protein secretion and sorting in the Trans-Golgi Network .

  • Cell cycle regulation: STK16 influences cell cycle progression, with its dysregulation potentially contributing to abnormal cell proliferation .

  • AKT signaling: Some studies suggest STK16 modulates AKT pathway activation in certain cancer types .

GSEA analysis of TCGA databases indicates that STK16 positively activates the MYC signaling pathway in both colon and rectal cancers, confirming its oncogenic role in these malignancies .

What is the significance of STK16 autophosphorylation for its function?

STK16 undergoes autophosphorylation at several key residues that critically influence both its kinase activity and subcellular localization. The three main autophosphorylation sites are:

  • Threonine 185 (Thr185)

  • Serine 197 (Ser197)

  • Tyrosine 198 (Tyr198)

Among these, Tyr198 holds particular significance as research has demonstrated that mutation of this single residue significantly reduces STK16's kinase activity, abolishes both its Golgi and membrane localization, and affects cell cycle progression . This highlights Tyr198 autophosphorylation as a critical molecular switch that determines both the function and localization of STK16.

The enzyme-deficient mutant STK16 T198A has been shown to be catalytically inactive and unable to upregulate c-MYC and its downstream targets GLUT1 and CDK4, confirming that STK16-mediated oncogenic signaling depends on its enzymatic activity . This provides a strong rationale for targeting STK16's kinase activity as a therapeutic approach in cancer.

How does STK16 expression differ between normal and cancer tissues?

STK16 expression shows significant variation between normal and cancer tissues, particularly in colorectal cancer. Analysis of The Cancer Genome Atlas (TCGA) database reveals that STK16 is significantly overexpressed in colorectal cancer compared to normal colorectal tissues . This finding has been validated through multiple experimental approaches:

These findings collectively establish STK16 as a potential biomarker for colorectal cancer progression and prognosis, with higher expression correlating with more advanced disease and worse outcomes.

What experimental approaches are most effective for studying STK16 phosphorylation targets?

Investigating STK16 phosphorylation targets requires a multi-faceted approach combining both targeted and unbiased strategies. The following experimental approaches have proven effective in STK16 research:

  • In vitro kinase assays: Recombinant STK16 protein is incubated with potential substrate proteins in the presence of ATP to directly assess phosphorylation. This approach was instrumental in identifying c-MYC as a substrate, particularly at serine 452 .

  • Phospho-specific antibodies: For known substrates like c-MYC, developing phospho-specific antibodies against the target residue (e.g., phospho-S452-c-MYC) enables detection of endogenous phosphorylation events in cell lysates or tissue samples. This approach allows monitoring of STK16 activity in various experimental conditions.

  • Phosphoproteomic mass spectrometry: For unbiased discovery of novel substrates, phosphoproteomic approaches comparing cells with and without active STK16 can reveal differential phosphorylation patterns. Quantitative techniques like SILAC or TMT labeling are particularly valuable.

  • Substrate validation through mutagenesis: Creating phospho-deficient mutants of potential substrates (e.g., c-MYC S452A) helps confirm the functional significance of STK16-mediated phosphorylation. This approach was used to demonstrate that STK16's effects on colorectal cancer proliferation depend on c-MYC S452 phosphorylation .

  • Genetic and pharmacological perturbation: Using STK16 knockdown/knockout (via CRISPR) or specific inhibitors (e.g., STK16-IN-1) followed by western blotting for known or suspected phosphorylation events provides physiologically relevant validation .

A comprehensive approach typically combines unbiased discovery methods with targeted validation strategies to establish bona fide STK16 substrates and their functional consequences.

How can researchers effectively design CRISPR/Cas9 knockout experiments for STK16?

Designing effective CRISPR/Cas9 knockout experiments for STK16 requires careful consideration of guide RNA design, validation strategies, and phenotypic assessments. Based on published research and available tools, the following methodology is recommended:

  • Guide RNA design:

    • Target early constitutive exons within the STK16 gene to ensure complete functional knockout

    • Commercial tools like the STK16 CRISPR/Cas9 KO Plasmid system provide a pool of 3 plasmids with target-specific 20 nt guide RNAs designed for maximum knockout efficiency

    • Consider targeting the 5' constitutive exons of STK16 mapping to chromosome 2q35

  • Delivery method:

    • Plasmid-based systems provide a cost-effective approach for most cell lines

    • For colorectal cancer research, RKO and Lovo cell lines have been successfully used for STK16 knockout studies

    • Each vial of commercial CRISPR/Cas9 KO Plasmid contains 20 μg of lyophilized plasmid DNA suitable for up to 20 transfections

  • Validation of knockout:

    • Western blotting using specific antibodies (e.g., STK16 (B-10): sc-374356) at appropriate dilutions (1:200 starting dilution)

    • Genomic PCR and sequencing across the target site to confirm indel formation

    • Functional assays to confirm loss of STK16 activity, such as measuring c-MYC phosphorylation at S452

  • Phenotypic analysis:

    • Proliferation assays (CCK8, BrdU incorporation) to assess impact on cell growth

    • Colony formation assays to evaluate clonogenic potential

    • Migration and invasion assays to assess metastatic capabilities

    • Analysis of c-MYC, GLUT1, and CDK4 expression levels as downstream readouts

Research has demonstrated that STK16 knockout significantly hinders colorectal cancer cell proliferation, colony formation, migration, and invasion capabilities, confirming its oncogenic role .

What are the best in vivo models for studying STK16's role in cancer progression?

Selecting appropriate in vivo models is critical for understanding STK16's role in cancer progression. Based on current research approaches, the following models offer complementary advantages:

  • Xenograft mouse models:

    • System: Immunodeficient nude female mice (6-8 weeks old) injected with human colorectal cancer cells

    • Cell lines: RKO and Lovo cells with STK16 knockout or overexpression have been successfully used

    • Injection protocol: 3-4 × 10^6 pretreated cells in 100 μl PBS injected into the backs of mice

    • Measurements: Tumor volume measured at specified intervals; mice sacrificed according to humane endpoint guidelines

    • This model has successfully demonstrated that STK16 knockout significantly inhibits colorectal cancer growth in vivo

  • Pharmacological inhibition in xenograft models:

    • STK16 inhibitor: STK16-IN-1 administered at 10 mg/kg via intraperitoneal injection once daily

    • Research has shown that pharmacological inhibition of STK16 significantly curtails colorectal cancer proliferation and c-MYC expression in vivo

    • This approach provides validation of STK16 as a druggable target with therapeutic potential

  • Tissue analysis protocols:

    • After sacrifice, tumors should be isolated, weighed, and fixed with 4% paraformaldehyde

    • Immunohistochemistry for STK16, c-MYC, and downstream targets provides mechanistic insights

    • Western blotting of tumor lysates confirms target engagement and pathway modulation

These models have been instrumental in establishing the oncogenic role of STK16 in colorectal cancer and validating its potential as a therapeutic target. The observation that both genetic knockout and pharmacological inhibition of STK16 significantly reduce tumor growth in vivo provides compelling evidence for the development of STK16 inhibitors as cancer therapeutics.

How can researchers distinguish between direct and indirect effects of STK16 inhibition?

Distinguishing between direct and indirect effects of STK16 inhibition is crucial for understanding its true mechanism of action and therapeutic potential in cancer. Researchers can employ several complementary strategies:

  • Biochemical validation of direct interactions:

    • In vitro kinase assays with purified recombinant STK16 and potential substrates (e.g., c-MYC)

    • Direct binding studies using techniques such as surface plasmon resonance or isothermal titration calorimetry

    • Co-immunoprecipitation assays to verify physical interactions

  • Genetic manipulation approaches:

    • Compare phenotypes obtained by STK16 knockdown/knockout with those observed using STK16 inhibitors

    • Create phosphorylation-deficient mutants of proposed substrates (e.g., c-MYC S452A) to determine if preventing phosphorylation recapitulates STK16 inhibition effects

    • This approach established that STK16-mediated colorectal cancer proliferation depends on c-MYC S452 phosphorylation

  • Temporal analysis:

    • Conduct time-course experiments after STK16 inhibition to identify primary (rapid) versus secondary (delayed) effects

    • Use cycloheximide chase assays to separate effects on protein stability from effects on protein synthesis

    • Research has shown that STK16 has no effect on c-MYC mRNA levels, pointing to post-translational regulation

  • Pathway validation:

    • Western blotting for c-MYC and its downstream targets (GLUT1, CDK4) after STK16 manipulation

    • Compare wild-type STK16 with enzyme-deficient mutant (STK16 T198A) effects on pathway activation

    • Research demonstrated that STK16 WT, but not the enzyme-deficient mutant, upregulates c-MYC, GLUT1, and CDK4 expression

Through these approaches, researchers established that STK16 directly phosphorylates c-MYC at S452, hindering its degradation via the ubiquitin-proteasome pathway. This mechanistic insight provides a clear example of how direct and indirect effects of STK16 can be distinguished in experimental systems.

What mechanisms explain STK16's role in regulating c-MYC stability?

STK16 regulates c-MYC protein stability through a specific phosphorylation-dependent mechanism that interferes with ubiquitin-mediated proteasomal degradation. Understanding this process requires investigation of several interconnected molecular events:

  • Direct phosphorylation mechanism:

    • STK16 directly phosphorylates c-MYC at serine 452 (S452)

    • This phosphorylation event is dependent on STK16's kinase activity, as the enzyme-deficient mutant STK16 T198A cannot induce this modification

    • In vitro kinase assays with purified components can confirm this direct phosphorylation event

  • Impact on c-MYC stability:

    • S452 phosphorylation hinders c-MYC degradation via the ubiquitin-proteasome pathway

    • This leads to accumulation of c-MYC protein without affecting its mRNA expression levels

    • Cycloheximide chase assays can measure the half-life of c-MYC protein with or without STK16 activity

  • Ubiquitination analysis:

    • Ubiquitination assays comparing wild-type c-MYC versus S452A mutant in the presence or absence of STK16

    • Immunoprecipitation of c-MYC followed by ubiquitin western blotting reveals the impact of STK16 on c-MYC ubiquitination patterns

  • Proteasome involvement:

    • Treatment with proteasome inhibitors (e.g., MG132) in combination with STK16 manipulation can clarify the role of proteasomal degradation

    • Analysis of c-MYC levels after STK16 knockdown with or without proteasome inhibition helps establish the degradation mechanism

  • Pathway confirmation:

    • Gene Set Enrichment Analysis (GSEA) of TCGA and GEO databases confirms that STK16 positively activates the MYC signaling pathway in both colon and rectal cancers

    • Analysis of known c-MYC target genes (GLUT1, CDK4) provides functional validation of enhanced c-MYC activity

This mechanism represents a novel regulatory pathway for c-MYC protein stability that could be therapeutically targeted in cancers where MYC signaling drives tumor progression. The discovery that STK16 inhibition, either genetically or pharmacologically, reduces c-MYC levels and cancer cell growth provides a strong rationale for developing STK16 inhibitors as cancer therapeutics.

What are the challenges in developing STK16 inhibitors as cancer therapeutics?

Developing effective STK16 inhibitors for cancer therapy presents several challenges that researchers must address through systematic approaches:

  • Kinase selectivity considerations:

    • STK16 belongs to the serine/threonine kinase family with structural similarities to other kinases

    • Designing inhibitors with high selectivity for STK16 over other kinases requires detailed structural understanding

    • STK16-IN-1 has shown promising results in preclinical models, but comprehensive selectivity profiling is essential

    • Kinome-wide screening of candidate compounds helps identify off-target effects

  • Delivery to subcellular locations:

    • STK16 localizes to the Golgi apparatus and cell membrane

    • Inhibitors must penetrate these compartments to effectively block STK16 activity

    • Physicochemical properties of inhibitors should be optimized for membrane permeability

    • Subcellular fractionation followed by activity assays can confirm target engagement in relevant compartments

  • In vivo efficacy and pharmacokinetics:

    • STK16-IN-1 (10 mg/kg) administered intraperitoneally once daily showed efficacy in xenograft models

    • Optimizing drug formulation, dosing regimen, and administration route remains challenging

    • Pharmacokinetic studies to ensure adequate tumor exposure are essential

    • Tumor penetration assessment using techniques like MALDI imaging can guide formulation optimization

  • Biomarker development:

    • Identifying patients most likely to respond to STK16 inhibition requires reliable biomarkers

    • c-MYC S452 phosphorylation status could serve as a pharmacodynamic marker

    • STK16 expression levels might predict sensitivity to inhibitors

    • Patients with higher STK16 expression have shown poorer clinical outcomes, suggesting a target population

  • Resistance mechanisms:

    • Anticipating and overcoming resistance to STK16 inhibition

    • Combination strategies with other targeted therapies or standard treatments

    • Understanding alternative pathways that might compensate for STK16 inhibition

The promising preclinical findings that STK16 inhibition significantly curtails colorectal cancer proliferation and c-MYC expression in vivo provide strong rationale for continued development of STK16 inhibitors . The clear molecular mechanism linking STK16 to c-MYC stability offers opportunities for rational therapeutic design and patient selection strategies.

How does STK16's autophosphorylation status influence its cancer-promoting effects?

STK16's autophosphorylation status serves as a critical regulatory mechanism that influences its oncogenic potential through multiple interconnected effects on its activity, localization, and substrate interactions:

  • Impact on kinase activity:

    • Autophosphorylation, particularly at Tyr198, is essential for STK16's kinase activity

    • The Y198F mutation significantly reduces STK16's catalytic function

    • Reduced kinase activity directly impairs STK16's ability to phosphorylate oncogenic substrates like c-MYC

    • The enzyme-deficient mutant STK16 T198A cannot upregulate c-MYC, GLUT1, and CDK4 expression, confirming the importance of catalytic activity

  • Regulation of subcellular localization:

    • Tyr198 autophosphorylation is required for proper localization of STK16 to the Golgi apparatus and cell membrane

    • Y198 mutation abolishes both Golgi and membrane localization of STK16

    • This mislocalization potentially prevents STK16 from accessing its relevant substrates in these compartments

    • Proper subcellular targeting is likely essential for STK16 to participate in oncogenic signaling pathways

  • Cell cycle effects:

    • STK16's autophosphorylation status affects cell cycle progression

    • This may contribute to its influence on cancer cell proliferation

    • The Y198 mutation has been shown to impact cell cycle regulation

  • Downstream signaling consequences:

    • In colorectal cancer, STK16-dependent phosphorylation of c-MYC at S452 prevents its degradation

    • This stabilization of c-MYC depends on STK16's catalytic activity, which is governed by its autophosphorylation status

    • The resulting increased c-MYC levels drive expression of pro-growth genes like GLUT1 and CDK4

These findings highlight autophosphorylation as a potential vulnerability that could be targeted therapeutically. Strategies that interfere with STK16 autophosphorylation, particularly at Tyr198, might provide an alternative approach to direct kinase inhibition for blocking STK16's oncogenic functions. Understanding the structural basis of this autophosphorylation mechanism could inform the design of novel therapeutic approaches targeting STK16 in cancer.

What are the most promising research directions for STK16 in cancer therapy?

Based on current findings, several promising research directions for STK16 in cancer therapy warrant further investigation:

  • Development of next-generation STK16 inhibitors:

    • Structure-based design of selective, potent STK16 inhibitors with improved pharmacokinetic properties

    • STK16-IN-1 has demonstrated efficacy in preclinical models at 10 mg/kg daily dosing

    • Optimization of existing scaffolds and exploration of novel chemical classes

    • Development of degraders (PROTACs) targeting STK16 as an alternative to catalytic inhibition

  • Combination therapy strategies:

    • Identifying synergistic combinations of STK16 inhibitors with established therapies

    • Exploring combinations with direct c-MYC pathway inhibitors

    • Testing STK16 inhibition in combination with standard-of-care agents for colorectal cancer

    • Investigating potential for overcoming resistance to existing therapies

  • Biomarker development for patient selection:

    • STK16 expression levels correlate with poorer clinical outcomes in colorectal cancer

    • Higher T stage and clinical stage exhibit elevated STK16 expression levels

    • Developing immunohistochemistry or other assays to identify patients most likely to benefit

    • Exploring c-MYC S452 phosphorylation as a predictive biomarker for response

  • Expanding therapeutic indications:

    • Beyond colorectal cancer, STK16 has shown oncogenic potential in fibrosarcoma and lung cancer

    • Systematic screening across cancer types to identify additional indications

    • Understanding tissue-specific mechanisms that might influence response to STK16 inhibition

  • Investigation of STK16's role in cancer metabolism:

    • STK16 upregulates GLUT1 expression through c-MYC stabilization

    • GLUT1 is a key glucose transporter that supports cancer cell metabolic demands

    • Exploring how STK16 inhibition might disrupt cancer metabolism provides another mechanistic angle

  • Immunological consequences of STK16 inhibition:

    • Examining how STK16 inhibition affects the tumor microenvironment and anti-tumor immunity

    • Potential for combining STK16 inhibitors with immunotherapy

The discovery that STK16 phosphorylates c-MYC at serine 452, hindering its degradation via the ubiquitin-proteasome pathway, provides a strong mechanistic rationale for targeting STK16 in c-MYC-driven cancers . Both genetic knockout and pharmacological inhibition of STK16 have shown promising results in curtailing cancer growth and c-MYC expression in vivo, highlighting STK16 as a potential therapeutic target with a clear path to clinical translation .

Product Science Overview

Molecular Characteristics

STK16 is a membrane-associated protein primarily localized to the Golgi apparatus . It has a molecular weight of approximately 38.7 kDa and is expressed in various tissues, including the fetal liver . The protein is involved in several cellular processes, including signal transduction pathways and transcriptional regulation .

Functional Roles

STK16 plays a crucial role in the TGF-β signaling pathway, protein secretion and sorting at the trans-Golgi network (TGN), and regulation of the cell cycle and Golgi assembly . It acts on both serine and threonine residues, which are essential for its kinase activity .

Post-Translational Modifications

STK16 undergoes post-translational modifications such as fatty acylation (myristoylation and palmitoylation) and phosphorylation . These modifications are vital for its proper localization and function within the cell.

Research and Applications

Recombinant human STK16 is widely used in research to study its role in various cellular processes and its potential implications in diseases. It is available in purified forms, often tagged with His-tag for easy detection and purification . Researchers utilize STK16 in kinase assays to understand its activity and interactions with other proteins .

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.