Recombinant Human Keratin, type I cytoskeletal 26 (KRT26)

Shipped with Ice Packs
In Stock

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 accommodate your request to the best of our ability.

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

Note: All proteins are shipped with standard blue ice packs. If dry ice shipping is required, 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%, which can serve as a guideline.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C. Lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.

The tag type will be determined during the production process. If you require a specific tag type, please inform us, and we will prioritize its development.

Synonyms
KRT26; KRT25B; Keratin; type I cytoskeletal 26; Cytokeratin-26; CK-26; Keratin-25B; K25B; Keratin-26; K26; Type I inner root sheath-specific keratin-K25irs2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-468
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
Target Protein Sequence
MSFRLSGGSR RICSRTGSGR LSGGGTGFVA GNVCVGSGAR SSFSCTLEGI SSGGSFCNSG GGLGSGACAG FLGNEHSLLS GNEKVTMQNL NDRLASYLDH VHALEEANAD LEQKIKGWYE KCEPGSSREH DHDYSRYFSV IEDLKRQIIS ATICNASIVL QNDNARLTAD DFRLKYENEL ALHHSVEADT SGLRRVLDEL TLCTTDLEIQ CETLSEELTY LKKSHEEEME VLQYTAGGNV NVEMNATPGV DLTVLLNNMR AEYEDLAEQN RKDAEAWFNE RSATLQQQIS DHEGAATAAR NELTELKRNL QTLEIELQSL MAVKHSYECS LAETEGNYCN QLQQIQDQIG VMEEQLQQIR TETEGQKLEY EQLLDVKIFL EKEIDIYCNL LDGEERKSKS TCYKSKGYRP VNSGNQAKDS TEETIVKTVV EELDQIGNLL SLRVHSVEEK SSKISNITVE QRVPSKAP
Uniprot No.

Q&A

What is the molecular structure and basic properties of Recombinant Human KRT26?

Recombinant Human Keratin, type I cytoskeletal 26 (KRT26) is a protein belonging to the keratin superfamily. It has the following properties:

  • Molecular Weight: Approximately 51.7 kDa

  • Genomic Location: Chromosome 17q21.2, existing in a cluster with other keratin genes

  • Expression: Primarily specific for the inner root sheath of the hair follicle

  • Alternative Names: Cytokeratin 26, CK26, K25, K26, KRT25B, K25IRS2

  • NCBI Accession: NP_853517

When produced as a recombinant protein, it typically contains a purification tag (commonly His-tag) and is expressed in expression systems such as Escherichia coli .

How is the purity and identity of recombinant KRT26 typically verified in laboratory settings?

Verification of recombinant KRT26 involves multiple analytical techniques:

  • SDS-PAGE and Coomassie Blue Staining: For purity assessment (typically >80% purity is considered acceptable for research applications)

  • Western Blotting: For identity confirmation using specific antibodies against KRT26 or the attached tag

  • Mass Spectrometry: For protein identity verification and detection of post-translational modifications

  • End-Sequencing: Of the ORF clone used for expression to confirm sequence integrity

  • Analytical SEC (HPLC): For higher resolution purity assessment and evaluation of oligomeric state

For recombinant KRT26 with His-tag, additional validation may include anti-tag ELISA to verify tag accessibility and functionality .

What are the primary research applications for recombinant KRT26 protein?

Recombinant KRT26 protein has several key applications in research:

  • Native Antigen Source: Serves as an optimal antigen for antibody production against KRT26

  • Positive Control: Acts as a standard in ELISA, Western blot, and other antibody-based assays

  • Functional Studies: Investigating protein-protein interactions involving KRT26

  • Structural Analysis: Understanding the structural properties of type I keratins

  • Neurodevelopmental Research: Exploring potential roles in autism spectrum disorders based on identified genetic variants

  • Hair Follicle Biology: Studying inner root sheath development and hair morphogenesis

  • Animal Science: Investigating associations with hair/fiber quality in livestock

How can researchers design experiments to study the association between KRT26 polymorphisms and phenotypic traits?

To effectively study KRT26 polymorphisms and their phenotypic associations:

Experimental Design Framework:

  • Population Selection:

    • Choose appropriate populations with measurable phenotypic variation

    • Consider adequate sample sizes for statistical power (typically >100 individuals)

    • Include diverse genetic backgrounds when applicable

  • Genotyping Approach:

    • PCR-Seq polymorphism detection

    • Sequence alignment for SNP identification

    • Focus on functional regions (exons, promoters, regulatory elements)

  • Phenotypic Measurements:

    • Standardize collection methods

    • Include multiple relevant traits (e.g., hair parameters)

    • Use quantitative and qualitative assessments

  • Statistical Analysis:

    • Association testing using SPSS or similar software

    • Haplotype analysis for combined genetic effects

    • Correction for multiple testing and population stratification

Example Study Design: In a study examining KRT26 in cashmere goats, researchers identified two significant SNPs (A559T and A6839G) through sequence alignment and PCR-Seq polymorphism analysis. The AA genotype at the KRT26 A559T locus was identified as the dominant genotype, and this specific genotype was associated with finer cashmere fibers, providing a marker for selective breeding programs .

What are the optimal storage and handling conditions for maintaining KRT26 protein stability and activity?

Storage and Handling Protocol:

ParameterRecommended ConditionNotes
Storage Temperature-80°CCritical for long-term stability
Buffer Composition25 mM Tris, pH 8.0, 150 mM NaCl, 10% glycerol, 1% SarkosylMaintains protein solubility
AliquotingSingle-use aliquots after initial thawMinimizes freeze-thaw damage
Freeze-Thaw CyclesMaximum 2-3 cyclesMore cycles significantly reduce activity
Working ConditionsKeep on ice during experimentsPrevents degradation
ConcentrationTypically supplied at 50 μg/mLMay require adjustment for specific applications
Stability PeriodAt least 3 months from receiptUnder proper storage conditions

When thawing, allow protein to thaw completely on ice before use. Avoid repeated freeze-thaw cycles as they can lead to protein degradation, aggregation, and loss of function .

How can researchers properly design quasi-experimental studies to investigate the role of KRT26 in hair follicle development?

When full experimental control isn't possible (especially in human studies), quasi-experimental designs offer viable alternatives:

  • Time-Series Design:

    • Monitor KRT26 expression during different stages of hair follicle development

    • Take measurements at regular intervals using ex vivo hair follicle models

    • Analyze temporal changes in expression patterns

  • Non-Equivalent Control Group Design:

    • Compare tissues with different levels of KRT26 expression

    • Match samples based on relevant characteristics (age, sex, hair type)

    • Control for confounding variables through statistical methods

  • Regression Discontinuity Design:

    • Focus on threshold effects in KRT26 expression levels

    • Assign samples to groups based on quantitative expression cutoffs

    • Analyze differences in hair development outcomes

  • Methodological Controls:

    • Include positive and negative tissue controls

    • Use siRNA knockdown or CRISPR/Cas9 for KRT26 manipulation

    • Employ multiple detection methods (IHC, qPCR, Western blot)

These quasi-experimental approaches help maximize internal validity while acknowledging the practical constraints of studying human keratin biology .

What evidence supports the role of KRT26 variants in autism spectrum disorders (ASD)?

Current Evidence Base:

  • Genetic Association Studies:

    • Rare inherited loss-of-function and damaging missense variants identified in ASD probands from:

      • Simons Simplex Collection (Krumm et al., 2015)

      • Cohort of Chinese ASD probands (Li J et al., 2017)

    • Transmission and De Novo Association (TADA) analysis identified KRT26 as an ASD candidate gene with PTADA of 0.005182

  • Specific Variants Identified:

    Allele ChangeResidue ChangeVariant TypeInheritanceStudy
    c.43C>Tp.Arg15TerStop-gainedDe novoZhou X et al. (2022)
    c.1060G>Tp.Glu354TerStop-gainedFamilialLi J et al. (2017)
    c.277C>Tp.Arg93CysMissenseFamilialLi J et al. (2017)
    G>Tp.Ser37TerStop-gainedPaternalKrumm N et al. (2015)
  • Functional Implications:

    • Multiple truncating mutations suggest potential haploinsufficiency mechanism

    • Variants occur across different domains of the protein

    • Both de novo and inherited variants identified, suggesting complex genetic architecture

How should researchers design experiments to investigate the functional impact of KRT26 variants on neuronal development?

Experimental Framework for Functional Studies:

  • In Vitro Neuronal Models:

    • Neural progenitor cells derived from patient iPSCs carrying KRT26 variants

    • CRISPR/Cas9 engineered cell lines with specific KRT26 variants

    • Isogenic control lines to minimize background genetic effects

  • Experimental Readouts:

    • Neuronal differentiation markers and morphology

    • Synapse formation and electrophysiological properties

    • Protein localization and interaction network analyses

    • Transcriptomic and proteomic profiling

  • Rescue Experiments:

    • Re-expression of wild-type KRT26 in variant-containing cells

    • Downstream pathway modulation to identify intervention points

    • Small molecule screening for phenotypic rescue

  • Animal Models:

    • KRT26 knockout or variant knock-in models

    • Behavioral assessments relevant to ASD symptomatology

    • Histological examination of brain development and architecture

  • Controls and Validation:

    • Include multiple variants to establish genotype-phenotype correlations

    • Test effects in multiple cell types and developmental stages

    • Correlate findings with patient clinical data when available

What techniques provide the most comprehensive tissue expression profiling of KRT26?

Multi-level Expression Analysis Methodology:

  • Transcriptional Analysis:

    • RNA-Seq for quantitative expression across tissues

    • Single-cell RNA-Seq for cellular resolution

    • qRT-PCR for targeted expression quantification

    • In situ hybridization for spatial localization

  • Protein-Level Detection:

    • Immunohistochemistry (IHC) for tissue localization

    • Fluorescence microscopy for subcellular localization

    • Western blotting for protein size verification

    • Mass spectrometry for proteoform identification

  • High-Resolution Anatomical Mapping:

    • Laser capture microdissection combined with expression analysis

    • Spatial transcriptomics for regional expression patterns

    • Multiplexed immunofluorescence for co-expression studies

Current data indicate that KRT26 expression shows tissue specificity with primary expression in the hair follicle inner root sheath. The Human Protein Atlas data demonstrates restricted expression patterns across brain regions and other tissues, with notable absence in many common tissue types .

How can researchers design experiments to resolve contradictory findings on KRT26 function across different model systems?

Experimental Strategy for Resolving Contradictions:

  • Systematic Comparison Approach:

    • Conduct parallel experiments across multiple model systems

    • Standardize experimental conditions and analysis methods

    • Use identical reagents and detection methods when possible

    • Include positive and negative controls relevant to each system

  • Sequential Hypothesis Testing:

    • Develop clear hypotheses explaining contradictions

    • Design experiments to specifically test each hypothesis

    • Use statistical methods appropriate for reconciling divergent data

    • Consider dose-response and temporal dynamics

  • Technical Validation:

    • Verify antibody specificity across species and platforms

    • Confirm KRT26 knockdown/overexpression efficiency

    • Use multiple detection methods for each finding

    • Assess for potential interfering factors (post-translational modifications, isoforms)

  • Biological Context Consideration:

    • Map differences in signaling pathway contexts

    • Evaluate species-specific differences in keratin biology

    • Examine developmental timing and microenvironmental factors

    • Consider compensatory mechanisms that may vary between models

How do structural features of KRT26 compare to other type I keratins, and what implications does this have for functional studies?

Structural Comparison Analysis:

  • Domain Organization:

    • Like other type I keratins, KRT26 contains:

      • N-terminal head domain (variable region)

      • Central rod domain (conserved α-helical region)

      • C-terminal tail domain (variable region)

  • Distinguishing Features:

    • Specific amino acid sequences in the head and tail domains confer functional specificity

    • The rod domain contains heptad repeats that facilitate coiled-coil formation with type II keratins

    • Unique regions likely mediate specific protein-protein interactions

  • Implications for Functional Studies:

    • Mutation studies should target both conserved and unique regions

    • Partner identification studies should consider heterodimeric interactions

    • Localization studies should examine both cellular and tissue distribution

    • Post-translational modification sites may differ from other keratins

Researchers should design experiments that account for these structural features, potentially using domain-swapping approaches to identify regions responsible for KRT26-specific functions .

What advanced methodologies can researchers employ to study KRT26 interactions with other hair follicle proteins?

Advanced Interaction Analysis Methods:

  • Proximity-Based Interaction Mapping:

    • BioID or APEX2 proximity labeling with KRT26 as bait

    • In situ proximity ligation assay (PLA) for endogenous interaction detection

    • FRET/BRET analysis for real-time interaction dynamics

  • Co-Immunoprecipitation Variations:

    • Tandem affinity purification for high-confidence interactions

    • Quantitative SILAC-based co-IP for dynamic interaction mapping

    • Crosslinking-assisted co-IP for transient interaction capture

  • Structural Biology Approaches:

    • Cryo-EM for filament structure determination

    • Hydrogen-deuterium exchange mass spectrometry for interaction interfaces

    • Integrative structural modeling combining multiple data types

  • Functional Validation Techniques:

    • CRISPR interference/activation to modulate interactor levels

    • Competitive peptide inhibition to disrupt specific interactions

    • Domain mapping through truncation and point mutation analysis

When studying KRT26 interactions with other hair follicle proteins like trichohyalin (TCHH), these approaches can reveal both structural and functional relationships that contribute to hair shaft formation and properties .

What emerging technologies might advance our understanding of KRT26 function in both normal development and disease states?

Emerging Technologies with Potential Impact:

  • Single-Cell Multi-omics:

    • Integrated single-cell RNA-seq, ATAC-seq, and proteomics to map KRT26 regulation

    • Spatial transcriptomics with subcellular resolution

    • In situ sequencing for tissue context preservation

  • Advanced Genome Engineering:

    • Base editing for precise introduction of KRT26 variants

    • Prime editing for complex genetic modifications

    • Inducible/conditional CRISPR systems for temporal control

    • Tissue-specific in vivo editing approaches

  • Organoid and Advanced Culture Systems:

    • Hair follicle organoids for developmental studies

    • Skin-on-chip microfluidic platforms

    • Bioprinted skin equivalents with controlled KRT26 expression

    • Patient-derived organoids for personalized disease modeling

  • Computational Approaches:

    • AI-driven protein structure prediction (AlphaFold2) for KRT26

    • Network-based analysis of KRT26 in keratin interaction landscapes

    • Systems biology modeling of filament assembly dynamics

    • Virtual screening for compounds affecting KRT26 function

How can researchers design transgenic animal models to better understand KRT26 function in vivo?

Transgenic Model Design Strategy:

  • Model Selection Considerations:

    • Mouse models offer genetic tractability and established protocols

    • Larger animals (e.g., sheep, goats) provide better hair follicle parallels

    • Consider evolutionary conservation of KRT26 across species

  • Genetic Modification Approaches:

    • Conventional knockout for complete loss-of-function

    • Conditional knockout using Cre/loxP for tissue-specific deletion

    • Knock-in of reporter genes (GFP, LacZ) for expression tracking

    • Human variant knock-in for disease modeling

  • Experimental Design Guidelines:

    • Include appropriate controls (littermates, isogenic lines)

    • Perform comprehensive phenotyping across tissues and time points

    • Analyze at both macroscopic and molecular levels

    • Consider compensatory mechanisms and genetic background effects

  • Tissue-Specific Considerations:

    • Use hair follicle-specific promoters (e.g., K14, K5) for targeted expression

    • Consider inducible systems for developmental timing studies

    • Examine both morphological and functional hair properties

    • Evaluate potential neurological phenotypes based on ASD associations

What are the key methodological pitfalls researchers should avoid when working with KRT26?

Critical Considerations and Pitfalls:

  • Protein Handling Issues:

    • Inappropriate storage conditions leading to degradation

    • Excessive freeze-thaw cycles reducing protein activity

    • Buffer incompatibility causing precipitation or aggregation

    • Insufficient protein concentration for detection methods

  • Experimental Design Flaws:

    • Inadequate controls (positive, negative, isotype)

    • Lack of validation across multiple techniques

    • Insufficient replication for statistical significance

    • Failure to account for cross-reactivity with related keratins

  • Interpretation Challenges:

    • Over-interpretation of correlative findings as causal

    • Neglecting species-specific differences in keratin biology

    • Failing to consider developmental and context-dependent effects

    • Disregarding technical limitations of detection methods

  • Translational Considerations:

    • Extrapolating from in vitro to in vivo systems without validation

    • Assuming direct relevance of animal models to human biology

    • Overlooking potential compensatory mechanisms

    • Not accounting for genetic background effects

What interdisciplinary approaches would be most productive for advancing KRT26 research?

Interdisciplinary Research Framework:

  • Integrative Research Teams:

    • Dermatologists and hair biologists for phenotypic expertise

    • Structural biologists for keratin assembly insights

    • Neuroscientists for ASD-related investigations

    • Geneticists for variant interpretation

    • Bioengineers for advanced model development

  • Methodological Integration:

    • Combine genetics, genomics, and proteomics approaches

    • Link structural studies with functional analysis

    • Connect molecular findings with clinical observations

    • Integrate computational modeling with experimental validation

  • Collaborative Research Design:

    • Standardize protocols across research groups

    • Develop shared resources (antibodies, cell lines, animal models)

    • Establish data sharing platforms for KRT26 research

    • Create interdisciplinary training opportunities

  • Translational Approaches:

    • Connect basic KRT26 biology to clinical manifestations

    • Develop biomarkers based on KRT26 variants or expression

    • Explore therapeutic targeting of KRT26-associated pathways

    • Create patient-derived models for personalized medicine applications

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.