Kctd10, also known as Potassium Channel Tetramerization Domain Containing 10, is a protein that functions as a substrate-specific adapter of a BCR (BTB-CUL3-RBX1) E3 ubiquitin-protein ligase complex . This complex, specifically BCR(BACURD3), mediates the ubiquitination of target proteins, leading to their degradation by the proteasome . Kctd10 is involved in various cellular processes, including cardiac development and regulation of Rac1 activity in certain cancers .
BTB/POZ Domain: Kctd10 contains a BTB/POZ domain, which is present in a family of proteins that mediate protein degradation in the proteasome . These domains are known for their role in protein-protein interactions and are crucial for the function of KCTD proteins .
E3 Ubiquitin Ligase Complex: Kctd10 functions as an adapter for the Cullin-3 (CUL3)-Rbx1 E3 ubiquitin ligase complex . This complex is responsible for ubiquitinating target proteins, marking them for degradation by the proteasome .
Functional Diversity: The KCTD protein family, including Kctd10, exhibits diverse functions, ranging from regulating GABA receptors to influencing cell proliferation .
Kctd10 plays a critical role in heart morphogenesis by directly binding to Tbx5, a transcription factor, and repressing its transcriptional activity . This interaction is essential for regulating the expression of has2, a gene involved in the formation of the atrioventricular canal (AVC) . In zebrafish embryos, Kctd10 restricts the expression of has2 to the AVC, ensuring proper cardiac jelly formation and valve development . Mutations in kctd10 lead to ectopic expression of has2, resulting in heart malformations .
| Experiment | Details | Results |
|---|---|---|
| Reporter Assay | G5E1b-luciferase and Gal4-Tbx5(266–518) used to measure Tbx5 transcriptional activity. | Kctd10 significantly suppressed Tbx5-induced Luciferase gene expression in a dose-dependent manner. |
| Tbx5 Target Gene Regulation | Luciferase reporter assays with constructs of Tbx5 target genes. | Kctd10 regulates the expression of Tbx5 target genes, indicating its role in modulating Tbx5 function. |
| Phenotype Analysis in Mutants | Examination of kctd10 mutant zebrafish embryos. | Ectopic expression of has2 throughout the heart tube, leading to excessive cardiac jelly and heart malformations. |
| Model of Kctd10 Function | Proposed mechanism for Kctd10's role in heart morphogenesis. | Kctd10 moderates Tbx5 activity to maintain proper has2 expression at the AV boundary, ensuring correct cardiac jelly formation and valve development. |
In human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells, Kctd10, as part of the Cullin-3 complex, is essential for Rac1 activation through RhoB degradation . The CUL3/KCTD10 complex ubiquitinates RhoB, leading to its degradation and subsequent activation of Rac1 . Rac1, a Rho GTPase, regulates F-actin organization and signal transduction, controlling cell proliferation and plasma membrane dynamics . High expression of Rac1 mRNA is correlated with poor prognosis in HER2-positive breast cancers .
| Experiment | Details | Results |
|---|---|---|
| siRNA Knockdown of CUL3 or KCTD10 | SKBR-3 cells treated with siRNA oligos for CUL3 or KCTD10. | Increased RhoB expression in cells depleted of CUL3 or KCTD10, suggesting that CUL3 and KCTD10 regulate the degradation of RhoB in SKBR-3 cells. Expression of RhoA, RhoC, Rac1/2/3, or Cdc42, was not affected by knockdown of CUL3 or KCTD10. |
| Expression of siRNA-Resistant CUL3 or KCTD10 | Expression of FLAG-tagged siRNA-resistant CUL3 or siRNA-resistant non-tagged KCTD10 in knockdown cells. | Reduced RhoB expression in CUL3 or KCTD10 knockdown cells, excluding off-target effects of siRNA. |
| Treatment with Cullin Inhibitors | Treatment of SKBR-3 cells with MLN-4924 (cullin inhibitor) and BafA1 (vacuolar-type H+-ATPase inhibitor). | Increased RhoB expression, suggesting RhoB is degraded through the lysosomal pathway after ubiquitination by CUL3/KCTD10. |
| F-actin Organization | Examination of F-actin organization in CUL3- or KCTD10-depleted cells. | RhoB knockdown restored EGF-induced membrane ruffle formation in CUL3- or KCTD10-depleted cells. Enforced expression of RhoB inhibited dorsal membrane ruffle formation. Aberrant F-actin organization caused by CUL3 or KCTD10 knockdown is, in part, due to the accumulation of RhoB. |
Congenital Heart Diseases: Given its role in cardiac development, Kctd10 may be a candidate for genetic screening and identification of congenital heart diseases .
Cancer Therapeutics: The involvement of Kctd10 in Rac1 activation in HER2-positive breast cancer suggests that targeting the CUL3/KCTD10/RhoB pathway could offer new treatment options for HER2- and Rac1-positive breast cancers .
Further Research: Further studies are needed to fully elucidate the mechanisms by which Kctd10 interacts with other proteins and regulates various cellular processes.
Note: We will ship the format currently in stock. If you require a specific format, please specify this in your order notes.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Recombinant Rat BTB/POZ domain-containing adapter for CUL3-mediated RhoA degradation protein 3 (Kctd10) is a substrate-specific adapter of a BCR (BTB-CUL3-RBX1) E3 ubiquitin-protein ligase complex. This complex mediates the ubiquitination of target proteins, leading to their proteasomal degradation.
KCTD10 is a member of the potassium channel tetramerization domain-containing protein family that functions primarily as a substrate receptor in Cullin-3 (CUL3)-based E3 ubiquitin ligase complexes . The protein contains a BTB/POZ domain that facilitates its interaction with Cullin-3 to form a functional E3 ligase complex capable of targeting specific proteins for ubiquitination and subsequent degradation . In endothelial cells, KCTD10 has been demonstrated to target RhoB for degradation, which is essential for maintaining endothelial barrier function . More recently, KCTD10 has been identified as a critical regulator of brain development through its role in mediating the degradation of KCTD13, another member of the KCTD family associated with neuropsychiatric disorders . Unlike some other KCTD family members such as KCTD13 and TNFAIP1 that primarily regulate RhoA, KCTD10 appears to have specificity for RhoB degradation in endothelial cells and possibly other cell types . This selective targeting of substrates by KCTD10 makes it a crucial component in multiple cellular signaling pathways that regulate cytoskeletal dynamics, cell morphology, and tissue development.
Recombinant Rat KCTD10 protein is available in several forms for research applications, including untagged versions and variants with different affinity tags to facilitate purification and detection . Common configurations include His-tagged KCTD10 and more complex fusion proteins such as His(Fc)-Avi-tagged KCTD10, which offers multiple options for protein detection, purification, and immobilization . The recombinant protein can be expressed in mammalian expression systems, with HEK293 cells being commonly used to ensure proper folding and post-translational modifications . The purity of commercially available recombinant KCTD10 typically exceeds 80-85% as determined by SDS-PAGE analysis, making it suitable for most research applications . Available preparations generally include both liquid formulations in PBS buffer and lyophilized powder forms that can be reconstituted as needed . Stability testing indicates that properly stored recombinant KCTD10 remains stable for at least 6 months, though repeated freeze-thaw cycles should be avoided to maintain protein integrity . For long-term storage, temperatures between -20°C and -80°C are recommended, while short-term storage can be accomplished at 4°C . These diverse formulations provide researchers with flexibility to select the most appropriate variant for specific experimental applications ranging from functional assays to structural studies.
KCTD10 demonstrates distinct expression patterns throughout development, with particularly notable expression in neural tissues during brain development . Recent studies have shown that KCTD10 is highly expressed in neuronal progenitors and layer V neurons throughout the developmental process of the brain . This specific expression pattern suggests a crucial role for KCTD10 in cortical neurogenesis and the proper formation of brain architecture . In addition to neural tissues, KCTD10 expression has been documented in endothelial cells, where it plays a critical role in regulating endothelial barrier function through its effects on RhoB degradation and subsequent control of cytoskeletal dynamics . The tissue-specific expression patterns of KCTD10 align with its diverse functional roles across different cell types and developmental stages. Notably, the expression pattern of KCTD10 appears to be distinct from that of other KCTD family members such as KCTD13 and TNFAIP1, which may explain their differential roles in regulating RhoGTPases and other cellular processes . Understanding these expression patterns provides important context for interpreting KCTD10 function in different experimental models and for designing targeted therapeutic approaches that modulate KCTD10 activity in specific tissues.
When investigating KCTD10-substrate interactions, researchers should employ multiple complementary approaches to establish both physical interaction and functional relationships . Co-immunoprecipitation (Co-IP) experiments have successfully demonstrated the interaction between KCTD10 and its substrates, as evidenced by studies showing that RhoB interacts with both Cullin-3 and KCTD10 in vivo . These experiments can be performed by transfecting cells with tagged versions of KCTD10 (e.g., HA-tagged) and the potential substrate protein (e.g., mCherry-tagged RhoB), followed by immunoprecipitation with antibodies against either protein tag and immunoblotting to detect the interacting partner . To validate the specificity of interactions, researchers should consider using different mutants of the substrate protein, as demonstrated in studies using wild-type RhoB, RhoB T19N, and RhoB G14V to assess interaction with the Cullin-3/KCTD10 complex . Ubiquitination assays are essential for confirming that KCTD10 targets the substrate for degradation, which can be performed by immunoprecipitating the substrate protein and immunoblotting for ubiquitin . These assays can be further validated by examining how depletion of KCTD10 affects substrate ubiquitination, as observed in studies showing reduced RhoB ubiquitination following KCTD10 knockdown . Functional validation should include rescue experiments where siRNA-resistant KCTD10 constructs are used to restore normal substrate levels and cellular phenotypes in KCTD10-depleted cells .
To effectively manipulate KCTD10 expression in experimental systems, researchers have successfully employed both RNA interference and overexpression approaches . For knockdown experiments, siRNA targeting specific regions of KCTD10 mRNA has proven effective in decreasing protein expression in various cell types, including endothelial cells and breast cancer cell lines . When designing such experiments, it is crucial to validate knockdown efficiency through western blot analysis and include appropriate controls to rule out off-target effects . A robust validation approach involves rescue experiments using siRNA-resistant KCTD10 constructs, which can restore normal KCTD10 function despite the presence of the siRNA . For overexpression studies, recombinant KCTD10 with various tags (His, HA, Fc, or Avi tags) can be employed depending on the specific experimental requirements for detection or purification . Cell-type specific considerations are important, as the effects of KCTD10 manipulation appear to vary between different cell lines; for instance, KCTD10 knockdown increases RhoB expression in HER2-positive breast cancer cells and endothelial cells but not in luminal or basal breast cancer cell lines . When studying the effects of KCTD10 on specific substrates, double knockdown experiments (e.g., simultaneously targeting KCTD10 and RhoB) can help establish causality in observed phenotypes . For in vivo studies, brain-specific knockout models have been developed to study KCTD10's role in neuronal development while avoiding potential embryonic lethality associated with complete knockout .
Identifying novel substrates of the KCTD10-Cullin-3 E3 ligase complex requires a comprehensive strategy combining proteomic, biochemical, and genetic approaches . Quantitative proteomics comparing protein abundance in wild-type versus KCTD10-depleted cells can reveal proteins that accumulate when KCTD10 is absent, as exemplified by studies identifying increased RhoB levels following KCTD10 knockdown . This approach should include treatments with protein degradation inhibitors such as MG-132 (proteasome), BafA1 (lysosomal), or MLN-4924 (neddylation inhibitor) to distinguish between different degradation pathways . Immunoprecipitation of KCTD10 followed by mass spectrometry analysis can identify proteins that physically interact with KCTD10, as demonstrated by the identification of KCTD13 as a KCTD10-interacting protein . Candidate validation requires demonstrating direct interaction through co-immunoprecipitation experiments and confirming that KCTD10 mediates ubiquitination of the candidate substrate . Functional validation should demonstrate that the phenotypic effects of KCTD10 depletion can be rescued by concurrent knockdown of the substrate, as shown in studies where RhoB knockdown restored normal actin cytoskeletal organization in KCTD10-depleted cells . Comparison across different cell types is important, as KCTD10 appears to have cell-type specific functions; for example, while KCTD10 regulates RhoB in endothelial and HER2-positive breast cancer cells, it does not appear to have this function in luminal or basal breast cancer cells .
KCTD10 plays a critical role in maintaining endothelial barrier integrity through its function as a substrate receptor in the Cullin-3 E3 ubiquitin ligase complex that targets RhoB for degradation . In endothelial cells, KCTD10 forms a complex with Cullin-3 that specifically recognizes RhoB and facilitates its ubiquitination, leading to proteasomal or lysosomal degradation . This degradation mechanism is crucial for controlling RhoB protein levels, as evidenced by the sixfold increase in RhoB expression observed following KCTD10 knockdown in endothelial cells . The regulatory effect appears to be post-transcriptional, as KCTD10 depletion actually decreased RhoB mRNA levels while dramatically increasing protein expression . Functionally, the accumulation of RhoB following KCTD10 knockdown induces strong actin polymerization and cell contraction, leading to disruption of the endothelial barrier . These cytoskeletal effects appear to be mediated through RhoB's inhibitory effect on Rac1 activation, a key regulator of cytoskeletal dynamics and cell spreading . The essential role of this pathway is demonstrated by rescue experiments showing that reintroduction of siRNA-resistant KCTD10 in KCTD10-depleted cells restores RhoB to basal levels and reverses cell contraction and F-actin accumulation . Furthermore, co-depletion of RhoB in KCTD10-knockdown cells completely abolishes the barrier-disruptive effects, confirming that RhoB is the critical downstream effector through which KCTD10 regulates endothelial barrier function .
KCTD10's function appears to be context-dependent, with distinct roles in cancer cells compared to normal cells that could have important implications for understanding cancer biology and developing targeted therapies . In HER2-positive breast cancer cells (SKBR-3 and MDA-MB-453), KCTD10 functions as part of the Cullin-3 E3 ligase complex to regulate RhoB degradation, similar to its role in endothelial cells . This is evidenced by increased RhoB expression following KCTD10 knockdown in these cell lines, an effect that could be reversed by expressing siRNA-resistant KCTD10 . Interestingly, this regulatory mechanism appears to be specific to HER2-positive breast cancer cells, as KCTD10 knockdown did not affect RhoB expression in other breast cancer subtypes, including luminal (MCF-7) and basal (MDA-MB-231) breast cancer cell lines . The functional consequence of this pathway in HER2-positive cancer cells involves the regulation of the actin cytoskeleton and cell morphology through modulation of Rac1 activation . Specifically, KCTD10 knockdown in SKBR-3 cells resulted in aberrant F-actin organization and impaired membrane ruffle formation in response to EGF stimulation . These effects were mediated by RhoB accumulation, as they could be reversed by simultaneous knockdown of RhoB . The cancer-specific function of KCTD10 suggests it may be involved in the aggressive phenotype of HER2-positive breast cancers, potentially through effects on cell migration, invasion, or response to HER2-targeted therapies . This differential activity across cancer subtypes highlights the importance of considering tumor-specific molecular contexts when studying KCTD10 function and developing therapeutic strategies targeting this pathway.
Production of high-quality recombinant KCTD10 protein for research applications presents several challenges that can be addressed through careful optimization of expression systems and purification protocols . One major consideration is the choice of expression system, with mammalian cells such as HEK293 generally preferred for KCTD10 expression to ensure proper folding and post-translational modifications . When using mammalian expression systems, optimization of transfection conditions, including DNA:transfection reagent ratios and cell density at transfection, is critical for maximizing protein yield . For purification, the selection of appropriate affinity tags can significantly impact both yield and purity; His-tagged and His(Fc)-Avi-tagged versions of KCTD10 are commercially available and facilitate purification through nickel affinity chromatography . To improve protein solubility and prevent aggregation, buffer optimization is essential, with PBS buffer being commonly used for storage of purified KCTD10 . Quality control testing should include verification of protein purity by SDS-PAGE (typically aiming for >80-85% purity) and endotoxin testing using the LAL method to ensure levels remain below 1.0 EU per μg of protein . For experiments requiring higher purity, additional purification steps such as size exclusion chromatography may be necessary to achieve >95% purity . To maintain protein stability, proper storage conditions are crucial, with recommendations for aliquoting the protein to avoid repeated freeze-thaw cycles and storing at -20°C to -80°C for long-term preservation . For functional studies, verification of the recombinant protein's activity through binding assays with known interaction partners (e.g., Cullin-3) or substrate ubiquitination assays may be necessary to confirm that the purified protein retains its native activity .
Distinguishing the specific functions of KCTD10 from other KCTD family members requires experimental approaches that account for potential functional overlap while identifying unique activities and substrates . Comparative knockdown experiments have proven valuable in this regard, as demonstrated by studies showing that depletion of KCTD10, but not KCTD13 or TNFAIP1, induced strong actin polymerization and contraction in endothelial cells, similar to Cullin-3 knockdown . These experiments revealed that while KCTD13 and TNFAIP1 primarily regulate RhoA degradation, KCTD10 specifically targets RhoB for degradation, illustrating the substrate specificity that differentiates KCTD family members . Rescue experiments can further delineate specific functions by expressing one family member in cells depleted of another; if the phenotype is not rescued, this suggests non-redundant functions . Domain swap experiments, where specific domains of different KCTD proteins are exchanged, can help identify the regions responsible for substrate specificity or distinct functions . Co-expression studies can reveal potential interactions or competitive relationships between different KCTD family members, as seen in the discovery that KCTD10 can target KCTD13 for degradation . Tissue-specific expression analysis can also highlight differential roles, with KCTD10 showing high expression in neuronal progenitors and layer V neurons during brain development, suggesting specialized functions in these contexts . When designing these experiments, researchers should consider potential compensatory mechanisms between family members and the possibility of context-dependent functions that vary across cell types and developmental stages .
Studying KCTD10 in disease models requires tailored methodological approaches that address the specific pathological context while enabling detailed mechanistic investigations . For neuropsychiatric disorder models, brain-specific knockout of KCTD10 in mice has proven valuable, revealing motor deficits and abnormal neural development that mirror aspects of neurodevelopmental disorders . These models allow for detailed analysis of cortical development through techniques such as immunohistochemistry to assess neuronal progenitor proliferation, differentiation, and migration, as well as quantification of layer-specific neuronal populations . In vascular disease models, endothelial-specific manipulation of KCTD10 expression can be achieved through cell-type specific promoters or by using isolated endothelial cells, allowing researchers to investigate KCTD10's role in endothelial barrier function and vascular permeability . For cancer models, comparison across different cancer subtypes is crucial, as demonstrated by the finding that KCTD10 regulates RhoB in HER2-positive breast cancer cells but not in luminal or basal subtypes . Patient-derived xenografts or cell lines can be used to study KCTD10's role in tumor progression, metastasis, and response to therapy . Across all disease models, molecular analyses should include assessment of both protein and mRNA levels of KCTD10 and its substrates to distinguish between transcriptional and post-transcriptional regulatory mechanisms . Functional readouts should be selected based on the relevant pathological process, such as measuring endothelial barrier function in vascular models, neuronal migration and differentiation in neuropsychiatric models, or cell migration and invasion in cancer models . Integration of multiple experimental approaches, including in vitro cell culture, ex vivo tissue analysis, and in vivo animal models, provides the most comprehensive understanding of KCTD10's role in disease pathogenesis .
Therapeutic targeting of the KCTD10 pathway represents an emerging area with potential applications across multiple diseases, including neuropsychiatric disorders, vascular pathologies, and cancer . For neuropsychiatric disorders associated with KCTD10 dysfunction, therapeutic strategies could focus on normalizing the levels of KCTD13, which accumulates when KCTD10 function is compromised . This might involve small molecule enhancers of KCTD10 activity or alternative approaches to promote KCTD13 degradation in cases of KCTD10 deficiency . In the context of vascular disorders, targeting the KCTD10-RhoB-Rac1 axis could provide avenues for modulating endothelial barrier function . Since KCTD10 knockdown increases RhoB expression and subsequently impairs Rac1 activation, compounds that either enhance KCTD10 activity or inhibit RhoB function might help maintain endothelial barrier integrity in conditions characterized by vascular leakage . For HER2-positive breast cancers, the specific role of KCTD10 in regulating RhoB degradation suggests potential for developing targeted therapies that exploit this pathway . This could involve combination approaches with existing HER2-targeted therapies, particularly if KCTD10 function influences response to these treatments . From a technical perspective, development of selective small molecule modulators of the Cullin-3/KCTD10 E3 ligase complex would represent a significant advance, though achieving specificity among different Cullin-RING ligase complexes remains challenging . Alternative approaches might include targeted protein degradation technologies such as PROTACs (Proteolysis Targeting Chimeras) that could be designed to selectively degrade disease-relevant substrates of KCTD10 or potentially KCTD10 itself in contexts where its inhibition would be beneficial .
The integration of KCTD10 with broader signaling networks represents an important frontier for understanding its full biological significance in development and disease . In neuronal development, KCTD10's regulation of KCTD13 likely interfaces with signaling pathways governing neuronal progenitor proliferation, differentiation, and migration . Given the association of both KCTD13 and Cullin-3 with neuropsychiatric disorders, exploring how KCTD10 intersects with risk pathways for conditions such as autism could reveal important disease mechanisms . In endothelial cells, KCTD10's regulation of RhoB impacts Rac1 activation, suggesting potential cross-talk with growth factor signaling pathways that utilize these GTPases as downstream effectors . This is particularly evident in HER2-positive breast cancer cells, where KCTD10 knockdown affected EGF-induced membrane ruffle formation, indicating integration with the EGF receptor signaling pathway . The observation that KCTD10 regulates RhoB degradation in HER2-positive breast cancer cells but not in other breast cancer subtypes suggests tumor-specific signaling contexts that determine KCTD10 function . Beyond RhoB and KCTD13, identification of additional KCTD10 substrates through comprehensive proteomic approaches could reveal unexpected connections to other signaling networks . The potential for KCTD10 to regulate multiple substrates in different cellular contexts suggests it may function as a signaling node that integrates various pathways depending on cell type, developmental stage, or disease state . Investigating how KCTD10 expression and activity are themselves regulated could uncover upstream signaling pathways that modulate this E3 ligase complex in response to various cellular stimuli or stressors .
Advancing KCTD10 research would benefit significantly from several technological innovations that could overcome current limitations in studying E3 ligase complexes and their substrates . Development of specific antibodies with high affinity for different KCTD family members would facilitate more precise detection of endogenous proteins and their interactions, addressing a common challenge in distinguishing between closely related family members . CRISPR-Cas9 gene editing technologies enable generation of cell lines and animal models with defined modifications to KCTD10, from complete knockouts to specific domain mutations or endogenous tags, allowing for more physiologically relevant studies than traditional overexpression approaches . Advanced proteomics methods, including proximity-based labeling techniques like BioID or APEX, could identify proteins that transiently interact with KCTD10 in living cells, potentially uncovering novel substrates or regulatory partners . Real-time monitoring of ubiquitination dynamics using fluorescent biosensors would provide insights into the spatial and temporal regulation of KCTD10-mediated substrate degradation within cells . High-throughput screening platforms for identifying small molecule modulators of KCTD10 activity could accelerate therapeutic development while providing chemical tools for mechanistic studies . Structural biology approaches, including cryo-electron microscopy of the complete Cullin-3/KCTD10 complex with substrates, would provide crucial insights into the molecular basis of substrate recognition and the conformational changes associated with the ubiquitination process . Single-cell transcriptomics and proteomics could reveal cell-type specific functions of KCTD10 and identify populations particularly sensitive to KCTD10 dysregulation in complex tissues like the developing brain . Integration of these technological advances would enable a more comprehensive understanding of KCTD10's diverse roles and facilitate translation of basic research findings into therapeutic applications for conditions involving KCTD10 dysfunction .