Recombinant Mouse Ubiquitin carboxyl-terminal hydrolase CYLD (Cyld), partial

Shipped with Ice Packs
In Stock

Description

Introduction

Recombinant Mouse Ubiquitin carboxyl-terminal hydrolase CYLD (Cylindromatosis) also known as CYLD lysine 63 deubiquitinase, is a deubiquitinating (DUB) enzyme that removes ubiquitin from specific proteins, thereby regulating their activity . CYLD is an evolutionarily ancient gene that is present as far back on the evolutionary scale as in sponges . In humans, the CYLD gene is located on chromosome 16q12.1 and encodes a protein of 956 amino acids . CYLD is involved in various cellular processes, including NF-κB signaling, cell cycle progression, apoptosis, immune response, autophagy, and the regulation of ciliogenesis .

Structure and Function

The CYLD protein contains three CAP-Gly domains (cytoskeletal-associated protein-glycine-conserved), which control critical functions . The C-terminal region of CYLD contains a catalytic domain with sequence homology to USP family members . The recombinant mouse CYLD protein (partial) typically includes a specific amino acid range (e.g., 579-952) and has a molecular weight of approximately 50.6 kDa . This recombinant protein often includes N-terminal and C-terminal tags (e.g., 10xHis and Myc tags) to facilitate detection and purification .

CYLD functions as a tumor suppressor by regulating cell growth and division . It negatively regulates NF-κB signaling by deubiquitinating key components in the pathway . CYLD also influences other cellular processes such as cell cycle progression, apoptosis, and immune response .

Mechanism of Action

CYLD is a deubiquitinating enzyme with specificity for removing K63-linked polyubiquitin chains from target proteins . By removing these ubiquitin chains, CYLD controls different signaling pathways like NF-κB, MAPK, and Wnt signaling . In the NF-κB pathway, K63-linked ubiquitin chains are attached to upstream signaling factors, facilitating protein-protein interactions in the assembly of signaling complexes. CYLD deconjugates these K63-linked ubiquitin chains, thereby negatively regulating NF-κB activation . CYLD can regulate the signaling of several proteins, affecting autophagy in a cell- and condition-dependent manner .

Role in Ciliogenesis

CYLD plays a critical role in the assembly of both primary and motile cilia in multiple organs . CYLD-mediated deconjugation of polyubiquitin chains from centrosomal protein of 70 kDa (Cep70) is required for Cep70 to interact with γ-tubulin and localize at the centrosome . CYLD-mediated inhibition of histone deacetylase 6 (HDAC6) promotes tubulin acetylation, which constitutes another mechanism for the ciliary function of CYLD .

Disease Associations

Mutations in the CYLD gene have been associated with several diseases, including:

  • CYLD Cutaneous Syndrome: Inactivating mutations in the CYLD gene occur in essentially all cases of CYLD cutaneous syndrome, a hereditary disorder in which individuals develop multiple skin tumors . This syndrome includes multiple familial trichoepithelioma-type, Brooke–Spiegler syndrome-type, and familial cylindromatosis-type .

  • Cancers: CYLD mutations are also associated with T-Cell Acute Lymphoblastic Leukemia, multiple myeloma, hepatocellular carcinoma, neuroblastoma, pancreatic cancer, uterine cancer, stomach cancer, colon cancer, lung cancer, and human papillomavirus-associated cancers .

  • Inflammatory Disorders: Dysregulation of CYLD has been linked to inflammatory disorders .

Research Findings

  • Tumor Suppressor Function: CYLD is a tumor suppressor, and its DUB activity is critical for this function . Mutations in the CYLD gene that result in truncated proteins lacking a functional DUB domain are commonly found in familial cylindromatosis and multiple familial trichoepithelioma patients .

  • NF-κB Regulation: CYLD negatively regulates the NF-κB signaling pathway by deubiquitinating key components . This regulation is important for controlling inflammation and cell survival .

  • Cell Cycle Regulation: CYLD regulates entry into mitosis, and this cell-cycle function appears to be independent of its role in NF-κB pathway regulation .

  • Interactions with Adaptor Proteins: The association of CYLD with some of its targets occurs indirectly through adaptors such as p62 and NEMO . These adaptors recruit CYLD to TRAF6 and other IKK regulators .

  • A20 and CYLD: Studies suggest that A20 and CYLD do not share significant overlapping functions in B cell homeostasis .

Therapeutic Potential

Understanding the molecular functions of CYLD provides insights into its potential therapeutic applications in cancers and inflammatory disorders . Small-molecule inhibitors of HDAC6 could partially rescue the ciliary defects in CYLD knockout mice .

Tables and Data

FeatureDescription
Gene NameCYLD (Cylindromatosis)
Protein FunctionDeubiquitinating enzyme that removes ubiquitin from specific proteins, regulating their activities
DomainContains three CAP-Gly domains and a catalytic domain with sequence homology to USP family members
Molecular Weight (Recombinant)Approximately 50.6 kDa (partial)
Cellular ProcessesNF-κB signaling, cell cycle progression, apoptosis, immune response, autophagy, and regulation of ciliogenesis
Disease AssociationsCYLD cutaneous syndrome, T-Cell Acute Lymphoblastic Leukemia, multiple myeloma, hepatocellular carcinoma, neuroblastoma, pancreatic cancer, uterine cancer, stomach cancer, colon cancer, lung cancer, and human papillomavirus-associated cancers
RegulationRegulates NF-κB, Wnt, notch, TGF-β, and JNK cell signaling pathways

Product Specs

Form
Lyophilized powder

Note: We will prioritize shipping the format currently in stock. However, if you have specific format requirements, please specify them during order placement. We will fulfill your request to the best of our ability.

Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery timelines.

Note: All proteins are shipped with standard blue ice packs. For dry ice shipment, please contact us in advance; additional fees will apply.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid form has a 6-month shelf life at -20°C/-80°C, while lyophilized form has a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.

Tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.

Synonyms
Cyld; Cyld1; Kiaa0849Ubiquitin carboxyl-terminal hydrolase CYLD; EC 3.4.19.12; Deubiquitinating enzyme CYLD; Ubiquitin thioesterase CYLD; Ubiquitin-specific-processing protease CYLD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
CYLD is a deubiquitinase that specifically cleaves Lys-63- and linear Met-1-linked polyubiquitin chains. It plays a crucial role in NF-κB activation and TNF-α-induced necroptosis. CYLD negatively regulates NF-κB activation by deubiquitinating upstream signaling factors, influencing cell survival, proliferation, and differentiation. It also negatively regulates Wnt signaling, inhibits HDAC6 (thereby promoting α-tubulin acetylation and microtubule stabilization), and regulates microtubule dynamics, affecting cell proliferation, polarization, migration, and angiogenesis. CYLD is essential for normal cell cycle progression and cytokinesis, inhibits NF-κB nuclear translocation, and regulates inflammation and innate immunity. While dispensable for the maturation of intrathymic natural killer cells, it is crucial for the survival of immature natural killer cells. It negatively regulates TNFRSF11A signaling and osteoclastogenesis and is involved in ciliogenesis regulation. Its ability to remove linear (Met-1-linked) polyubiquitin chains regulates innate immunity and TNF-α-induced necroptosis by restricting linear polyubiquitin formation on target proteins, particularly RIPK2 in response to NOD2 stimulation and RIPK1 in TNF-α-induced necroptosis, thereby modulating RIPK1 kinase activity. CYLD also removes Lys-63-linked polyubiquitin chains from MAP3K7, inhibiting phosphorylation and downstream activation of the JNK-p38 kinase cascades.
Gene References Into Functions
  1. Loss of CYLD catalytic activity impairs DNA damage-induced p53 stabilization and activation, sensitizing mice to carcinogen-induced tumorigenesis. PMID: 27561390
  2. CYLD regulates NF-κB inflammatory signaling induced by elevated glucose, suggesting a potential therapeutic target for diabetic nephropathy. PMID: 29259980
  3. CYLD's role in development, tissue homeostasis, and tumorigenesis. PMID: 28750888
  4. CYLD and HDAC6 interplay in ciliary homeostasis. PMID: 27028867
  5. Thrombin-mediated MALT1 activation disrupts endothelial barrier integrity via CYLD cleavage. PMID: 27681433
  6. SPATA2 acts as an adaptor between HOIP and CYLD, enabling CYLD recruitment to signaling complexes. PMID: 27545878
  7. TLR4 activates CASPASE-8 to cleave CYLD, disabling necroptosis in macrophages. PMID: 27264187
  8. CYLD contributes to adventitial fibroblast transdifferentiation by deubiquitinating Nox4. PMID: 28751569
  9. CYLD polarizes T cell cytokine response, favoring allergic airway disease. PMID: 27372382
  10. Intestinal epithelial CYLD suppresses colitis-associated carcinogenesis. PMID: 27042826
  11. CYLD negatively regulates MyD88-mediated signaling by deubiquitinating MyD88. PMID: 26719415
  12. CYLD interrupts ERK- and p38-/AP-1 and c-Myc pathways, enhancing oxidative stress. PMID: 25935309
  13. Inefficient negative selection in the thymus of CYLD(ex7/8) mice results from a defect in mTEC maturation. PMID: 25601276
  14. CYLD regulates apical docking of basal bodies in ciliated epithelial cells. PMID: 25134987
  15. In utero death of NEMO and CYLD double mutant mice is mediated by TNFR1 signaling. PMID: 26224629
  16. CYLD regulates apoptotic cell death in hepatocytes by controlling NF-κB dependent anti-apoptotic signaling. PMID: 25493017
  17. Dendritic cell-specific sCYLD regulates listeriosis pathogenesis. PMID: 25675948
  18. CYLD's ciliary function involves deconjugation of polyubiquitin chains from Cep70. PMID: 25342559
  19. CYLD negatively regulates NTHi-induced IL-8 expression via MKP-1-dependent inhibition of ERK. PMID: 25389768
  20. CYLD regulates HSC function, restricting cycling and promoting dormancy. PMID: 25824820
  21. RBP-J-mediated Notch signaling upregulates NF-κB activation through CYLD in hepatic fibrosis. PMID: 25145286
  22. CYLD is required for Toll-like receptor-induced necroptosis. PMID: 24706750
  23. CYLD ameliorates hepatocellular damage and liver fibrogenesis by regulating HGF levels. PMID: 24811579
  24. CYLD regulates RIP1 ubiquitination in TNFα-induced necroptosis. PMID: 24098568
  25. CYLD downregulation induces tumor cell proliferation in hepatocellular carcinoma. PMID: 24104553
  26. Therapeutic inhibition of CYLD augments NF-κB/IL-6/STAT3 pathway and fibrin production. PMID: 23825949
  27. CYLD inactivation promotes metastasis of tumor epidermal cells. PMID: 23426135
  28. PKCθ/β and CYLD antagonistically regulate NFκB and NFAT transactivation pathways in T lymphocytes. PMID: 23335970
  29. CYLD maintains T cell homeostasis and Treg function. PMID: 23066153
  30. CYLD regulates resolution of lung injury and prevents fibrosis by deubiquitinating Akt. PMID: 22491319
  31. CYLD regulates hepatocyte homeostasis by preventing uncontrolled TAK1 and JNK activation. PMID: 22698400
  32. Itch-Cyld-mediated regulatory mechanism in innate inflammatory cells. PMID: 22057290
  33. CYLD promotes inflammatory responses in VSMCs via MAPK activation. PMID: 22406061
  34. CYLD regulates antiviral innate immunity and IFN receptor signaling. PMID: 21946435
  35. CYLD regulates TGF-β signaling in T cells and Treg development through Smad7 deubiquitination. PMID: 21931165
  36. CYLD regulates thymocyte selection in an IKK2 and NF-κB dependent manner. PMID: 21728169
  37. SRF acts as a positive regulator of CYLD expression. PMID: 21573132
  38. Hes1 expression and CYLD repression are essential downstream of Notch1 in T-cell leukemia. PMID: 21389783
  39. CYLD regulates monocyte-macrophage activation in response to inflammatory stimuli. PMID: 21283724
  40. CYLD establishes the activation threshold for thymocyte selection. PMID: 20644164
  41. CYLD regulates homeostasis of immature NKT cells by regulating IL-7R signaling and ICOS expression. PMID: 20224552
  42. CYLD is induced by NF-κB. PMID: 15226292
  43. CYLD is a positive regulator of proximal TCR signaling in thymocytes. PMID: 16501569
  44. CYLD negatively regulates different NF-κB pathways; TRAF2 inactivation controls survival and inflammation, while Bcl-3 inhibition controls proliferation and tumor growth. PMID: 16713561
  45. CYLD limits inflammation and tumorigenesis by regulating ubiquitination. PMID: 17053834
  46. CYLD regulates basal NF-κB activity and B cell phenotype and activation. PMID: 17392286
  47. CYLD prevents spontaneous Tak1 axis activation in T cells. PMID: 17548520
  48. CYLD is a negative regulator of host antiviral response. PMID: 17608805
  49. CYLD is detrimental for host survival in pneumococcal pneumonia. PMID: 17723219
  50. CYLD positively and negatively regulates B cell signal transduction and homeostasis depending on splice variants. PMID: 17923499
Database Links
Protein Families
Peptidase C19 family
Subcellular Location
Cytoplasm. Cytoplasm, perinuclear region. Cytoplasm, cytoskeleton. Cell membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Cytoplasm, cytoskeleton, spindle. Cytoplasm, cytoskeleton, cilium basal body.

Q&A

What is the structural composition of recombinant mouse CYLD and how does it compare to native CYLD?

Recombinant mouse CYLD (partial form) typically contains the amino acid range 579-952, representing the catalytic domain of the protein. The partial recombinant protein has a theoretical molecular weight of approximately 50.6 kDa when expressed in E. coli systems. For research applications, the recombinant protein is commonly engineered with an N-terminal 10xHis tag and C-terminal Myc tag to facilitate detection and purification .

The crystal structure analysis reveals that CYLD, like other ubiquitin carboxyl-terminal hydrolases (UCHs), contains a catalytic triad consisting of cysteine, histidine, and aspartate residues in the active site. This catalytic domain adopts a conformation that allows specific recognition and processing of ubiquitin chains. The partial recombinant form maintains the essential catalytic activity while eliminating domains not required for the deubiquitinating function.

How does mouse CYLD function as a deubiquitinating enzyme in molecular signaling pathways?

Mouse CYLD functions as a specialized deubiquitinating enzyme that selectively removes ubiquitin chains from target proteins, thereby regulating protein stability and modulating multiple cellular signaling pathways. The primary mechanism of action involves:

  • Recognition of specific ubiquitin chain linkages (particularly K63-linked chains)

  • Hydrolysis of the isopeptide bond between ubiquitin molecules or between ubiquitin and substrate proteins

  • Consequent alteration of protein stability or signaling capacity

One of the most well-characterized functions of CYLD is negative regulation of NF-κB signaling through deubiquitination of key pathway components such as TRAF2, TRAF6, and NEMO. This deubiquitination prevents sustained pathway activation, thereby controlling inflammatory responses. Additionally, CYLD influences other cellular processes including cell cycle progression, apoptosis, and immune responses through its deubiquitinating activity on various substrates .

What are the optimal expression systems for producing functional recombinant mouse CYLD, and what yields can be expected?

While multiple expression systems can be used to produce recombinant mouse CYLD, E. coli remains the most commonly employed platform due to its simplicity and cost-effectiveness . For researchers seeking higher yields or post-translational modifications, alternatives include:

Expression SystemAdvantagesDisadvantagesExpected YieldPurification Method
E. coliCost-effective, rapid growth, simple genetic manipulationLimited post-translational modifications5-20 mg/LNi-NTA chromatography
Pichia pastorisProper protein folding, higher yields for secreted proteins, post-translational modificationsLonger cultivation time50-210 mg/LNi-NTA chromatography
Mammalian cellsNative-like post-translational modificationsHigher cost, lower yields1-5 mg/LAffinity chromatography

Based on similar UCH family proteins, when expression is optimized in P. pastoris with the appropriate induction conditions (pH 6.0 in BMMY/methanol medium), yields of up to 210 mg/L can be achieved for UCH family proteins . For E. coli expression systems, optimizing growth temperature (typically 16-25°C post-induction), IPTG concentration (0.1-0.5 mM), and culture density at induction (OD600 of 0.6-0.8) can significantly improve the yield of soluble, active CYLD.

What purification strategy provides the highest purity and activity for recombinant mouse CYLD?

A multi-step purification strategy is recommended to obtain highly pure and active recombinant mouse CYLD:

  • Initial Capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin is the primary purification step, exploiting the N-terminal His tag. Optimization of imidazole concentration in washing buffers (20-50 mM) is critical to remove non-specific binding proteins while retaining CYLD .

  • Intermediate Purification: Size exclusion chromatography (SEC) effectively separates aggregates and contaminants of different molecular weights.

  • Polishing Step: Ion exchange chromatography (IEX) can be employed as a final polishing step to achieve >95% purity.

  • Activity Preservation: Throughout purification, maintain reducing conditions (typically 1-5 mM DTT or 0.5-2 mM TCEP) to preserve the catalytic thiol group. Additionally, including 10% glycerol in storage buffers and flash-freezing aliquots helps maintain enzymatic activity during long-term storage.

The final purified protein should be assessed for both purity (by SDS-PAGE and Western blot) and enzymatic activity using a deubiquitination assay with appropriate substrates. Typical yields after complete purification range from 5-15 mg of active protein per liter of bacterial culture.

How can the deubiquitinating activity of recombinant mouse CYLD be accurately measured in vitro?

Multiple methodological approaches can be employed to assess the deubiquitinating activity of recombinant mouse CYLD:

  • Fluorogenic Ubiquitin Substrate Assay: Using ubiquitin-AMC (7-amino-4-methylcoumarin) as a substrate, where cleavage of the ubiquitin-AMC bond by active CYLD releases fluorescent AMC that can be quantified. This assay provides real-time kinetic data but only measures general DUB activity.

  • Chain-Specific Deubiquitination Assay: Using purified K63-linked polyubiquitin chains (CYLD's preferred substrate) followed by SDS-PAGE analysis to visualize the pattern of ubiquitin chain disassembly over time. This method more accurately reflects CYLD's natural substrate specificity.

  • Mass Spectrometry-Based Assay: Employing mass spectrometry to analyze the products of deubiquitination reactions provides precise information about the cleavage sites and specificity. Similar approaches have been used successfully with UCH enzymes to confirm their precise cleavage of ubiquitin fusion proteins .

For activity confirmation, ubiquitin fusion proteins can serve as substrates, with cleavage products analyzed by techniques such as Tricine-SDS-PAGE and ESI-MS. This approach has been validated for UCH family enzymes, demonstrating their ability to precisely recognize and cleave at the carboxyl terminus of ubiquitin .

What are the optimal buffer conditions and reaction parameters for studying CYLD enzymatic activity?

The optimization of reaction conditions is crucial for accurate assessment of CYLD enzymatic activity:

ParameterOptimal RangeNotes
pH7.5-8.0Activity decreases significantly below pH 7.0
Temperature30-37°CHigher temperatures may increase activity but reduce stability
NaCl50-150 mMHigher ionic strength may inhibit activity
Reducing agent1-5 mM DTT or 0.5-2 mM TCEPEssential to maintain active site cysteine in reduced state
BSA0.1-0.5 mg/mLReduces non-specific binding and protein loss
Incubation time30-60 minutesLonger incubations may show substrate depletion

When planning deubiquitination assays, it's important to note that CYLD shows preference for K63-linked polyubiquitin chains, with limited activity against K48-linked chains. Additionally, including deubiquitinase inhibitors such as N-ethylmaleimide (NEM) in control reactions can confirm the specificity of observed activity.

For kinetic analysis, determining Km and kcat values requires testing a range of substrate concentrations (typically 0.1-10× Km) and measuring initial reaction velocities under steady-state conditions. These parameters provide valuable insights into CYLD's catalytic efficiency and substrate preference.

How does CYLD dysregulation contribute to inflammatory disorders and cancer pathogenesis?

CYLD dysregulation has been implicated in various pathological conditions through several mechanistic pathways:

  • Inflammatory Disorders: CYLD negatively regulates NF-κB signaling, which is a master regulator of inflammation. Loss of CYLD function leads to enhanced and prolonged NF-κB activation, resulting in excessive inflammatory responses and contributing to conditions such as inflammatory bowel disease and autoimmune disorders . The mechanism involves CYLD's deubiquitination of TRAF proteins and IKK complex components, which normally activate NF-κB.

  • Cancer Development: CYLD functions as a tumor suppressor in multiple tissue types. Its downregulation or inactivation promotes:

    • Cell proliferation through enhanced cyclin D1 expression

    • Resistance to apoptosis via increased anti-apoptotic protein expression

    • Metastatic potential through altered cell adhesion and migration

    • Enhanced angiogenesis via VEGF upregulation

These effects stem from CYLD's role in regulating not only NF-κB but also other signaling pathways including Wnt/β-catenin, JNK, and p38 MAPK pathways.

  • Fibrotic Conditions: Similar to other deubiquitinating enzymes like UCHL5, CYLD may influence TGF-β signaling components, potentially affecting fibrotic processes in conditions such as idiopathic pulmonary fibrosis . UCHL5, another member of the UCH family, has been shown to de-ubiquitinate Smad2 and Smad3, stabilizing these proteins and promoting TGF-β1-induced expression of profibrotic proteins.

Understanding these pathological mechanisms provides the foundation for developing targeted therapeutic approaches aimed at restoring normal CYLD function or compensating for its loss.

What experimental models are most appropriate for studying CYLD function in specific disease contexts?

Various experimental models have been developed to study CYLD's role in disease pathogenesis:

Model TypeApplicationsAdvantagesLimitations
Cell lines with CYLD knockdown/knockoutMolecular signaling studies, drug screeningEasily manipulated, controlled conditionsLack of tissue context, compensatory mechanisms
Primary cells from CYLD knockout miceEx vivo functional studiesPhysiologically relevant, genetic homogeneityLimited lifespan, adaptation to culture
CYLD knockout miceIn vivo disease models, systemic effectsFull physiological context, multiple tissue analysisPotential developmental adaptations, strain variability
Conditional/inducible CYLD knockoutTissue-specific and temporal studiesAvoids developmental effects, tissue specificityTechnical complexity, incomplete recombination
Patient-derived samplesTranslational relevance, biomarker studiesDirect disease relevance, genetic diversityVariability, limited availability, ethical considerations

For inflammatory disorders, myeloid-specific conditional CYLD knockout models are particularly valuable, as they allow assessment of CYLD's role in immune cell function without confounding effects on other tissues. For cancer studies, xenograft models using CYLD-manipulated cell lines provide insights into tumor growth and metastasis.

When designing experiments, it's essential to consider the specific ubiquitin chain types (K63 vs. K48) relevant to the pathway being studied, as CYLD's selectivity for K63-linked chains influences which cellular processes will be affected by its manipulation.

What techniques can be used to identify novel substrates and interaction partners of CYLD in different cellular contexts?

Identifying the complete interactome of CYLD requires sophisticated methodological approaches:

  • Proximity-Based Labeling: BioID or APEX2 fusion proteins with CYLD allow biotinylation of proximal proteins, which can be purified and identified by mass spectrometry. This technique captures both stable and transient interactions in living cells.

  • Ubiquitinome Analysis: Quantitative proteomics comparing ubiquitination patterns in CYLD-expressing versus CYLD-deficient cells identifies substrates based on differential ubiquitination. This typically involves:

    • Tandem ubiquitin binding entity (TUBE) pulldown to enrich ubiquitinated proteins

    • Stable isotope labeling with amino acids in cell culture (SILAC) for quantitative comparison

    • Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis

  • Genetic Screens: CRISPR-based screens can identify synthetic lethal or synthetic viable interactions with CYLD, revealing functional relationships not detectable by direct protein interaction studies.

  • Structural Biology Approaches: Hydrogen-deuterium exchange mass spectrometry (HDX-MS) or crosslinking mass spectrometry (XL-MS) can map interaction interfaces and conformational changes upon substrate binding. UCH family enzymes are known to undergo significant conformational changes upon substrate binding, transitioning from inactive to active states .

When implementing these approaches, researchers should consider tissue-specific or context-dependent interactions, as CYLD may have different substrate preferences in different cell types or under various cellular stresses.

How can structural analysis of CYLD inform the development of specific inhibitors or activators for therapeutic applications?

Structural biology approaches provide critical insights for rational drug design targeting CYLD:

  • Active Site Targeting: X-ray crystallography and cryo-electron microscopy structures of CYLD's catalytic domain reveal the spatial arrangement of the catalytic triad (cysteine, histidine, and aspartate) that is essential for deubiquitinating activity. Similar to other UCH enzymes, CYLD likely has an unstructured loop that restricts access to the active site, presenting opportunities for selective inhibitor design .

  • Allosteric Modulation: Nuclear magnetic resonance (NMR) spectroscopy can identify allosteric sites where small molecule binding induces conformational changes affecting enzymatic activity. UCH family enzymes are known to exist in active and inactive conformations, with significant structural rearrangements occurring upon ubiquitin binding .

  • Structure-Activity Relationship (SAR) Studies: Virtual screening combined with enzymatic assays can identify lead compounds that selectively inhibit or activate CYLD. Molecular dynamics simulations can then predict how structural modifications might improve specificity or pharmacokinetic properties.

  • Protein-Protein Interaction (PPI) Targeting: Structural characterization of CYLD-substrate complexes identifies interfaces that could be targeted to selectively disrupt specific CYLD interactions while preserving others, potentially reducing side effects.

The development of selective CYLD-targeting compounds requires careful consideration of specificity, as the UCH family shares structural similarities in the catalytic domain. Focusing on unique structural features of CYLD and targeting non-catalytic domains involved in substrate selection could increase specificity.

What are the common pitfalls in working with recombinant CYLD and how can they be addressed?

Researchers frequently encounter several challenges when working with recombinant CYLD:

  • Low Solubility and Aggregation:

    • Problem: CYLD can form insoluble aggregates during expression and purification.

    • Solution: Express at lower temperatures (16-20°C), include solubility enhancers like sorbitol (0.5-1 M) or arginine (50-100 mM) in buffers, and consider fusion tags like SUMO or MBP that enhance solubility.

  • Loss of Enzymatic Activity:

    • Problem: The catalytic cysteine residue is susceptible to oxidation, resulting in activity loss.

    • Solution: Maintain reducing conditions throughout purification and storage (1-5 mM DTT or 0.5-2 mM TCEP), minimize freeze-thaw cycles, and consider adding protease inhibitors to prevent autolysis.

  • Non-specific DUB Activity in Assays:

    • Problem: Cell lysates contain multiple DUBs that can confound CYLD-specific activity measurements.

    • Solution: Use selective inhibitors of other DUB classes in assays, employ immunoprecipitation to isolate CYLD complexes, and include appropriate negative controls (catalytically inactive CYLD mutants).

  • Substrate Specificity Challenges:

    • Problem: Distinguishing CYLD activity from other DUBs with overlapping specificity.

    • Solution: Use defined chain linkage-specific ubiquitin substrates (K63-linked for CYLD), perform comparative assays with other purified DUBs, and validate results using CYLD knockout controls.

  • Variable Expression Levels:

    • Problem: Inconsistent yields between batches.

    • Solution: Standardize growth conditions, optimize codon usage for expression system, and consider inducible expression systems with tightly controlled promoters.

How can experimental design be optimized when using CYLD to study ubiquitin-dependent signaling pathways?

Robust experimental design for studying CYLD function requires careful consideration of several factors:

  • Appropriate Controls:

    • Catalytically inactive CYLD mutant (C601A) to distinguish enzymatic versus scaffolding functions

    • Domain deletion mutants to map interaction regions

    • Chain linkage specificity controls (K48 vs. K63 polyubiquitin) to confirm substrate selectivity

  • Temporal Considerations:

    • Ubiquitin-mediated signaling is dynamic, requiring time-course analyses

    • Synchronize cells when studying cell cycle-dependent processes

    • Use inducible systems (Tet-On/Off) for temporal control of CYLD expression

  • Cellular Context:

    • Different cell types may express different CYLD substrates or regulatory proteins

    • Primary cells versus cell lines may show different CYLD-dependent phenotypes

    • Consider tissue-specific conditional knockout models for in vivo studies

  • Readout Selection:

    • Direct measures of ubiquitination status using linkage-specific antibodies

    • Downstream signaling events (NF-κB activation, MAP kinase phosphorylation)

    • Functional outcomes (cell proliferation, apoptosis, inflammatory cytokine production)

    • Combine multiple readouts for comprehensive pathway analysis

  • Data Analysis Approaches:

    • Quantitative analysis of ubiquitination levels (Western blot densitometry or mass spectrometry)

    • Kinetic modeling of deubiquitination reactions

    • Network analysis to understand system-wide effects of CYLD manipulation

By carefully considering these experimental design elements, researchers can generate more reliable and interpretable data on CYLD's role in ubiquitin-dependent signaling 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 2025 TheBiotek. All Rights Reserved.