KRT16 (Keratin 16) is a type I intermediate filament gene that encodes the K16 protein. It is constitutively expressed in ectoderm-derived appendages and palmar/plantar epidermis, and is robustly induced when the epidermis experiences chemical, mechanical, or environmental stress . Spatial analysis reveals that KRT16 is strongly expressed in the suprabasal layers of the epidermis in lesional skin, contrasting with the pattern of keratins K14 and K5, which are predominantly expressed in the basal layer .
Methodological approach: To investigate KRT16 expression patterns, researchers can employ immunofluorescence staining with K16-specific antibodies, RNA in situ hybridization, single-cell RNA sequencing, and spatial transcriptomics. These techniques allow for precise mapping of KRT16 expression within the tissue architecture and can reveal relationships with other markers.
Missense mutations in the KRT16 locus can cause pachyonychia congenita (PC, OMIM:167200) or focal non-epidermolytic palmoplantar keratoderma (FNEPPK, OMIM:613000), which both involve painful calluses on palmar and plantar skin . Specific mutations identified in human patients include K16-R127C and K16-R127G .
Methodological approach: To study these mutations, researchers typically use:
DNA extraction and sequencing to identify mutations
Patient-derived keratinocyte cultures
CRISPR-Cas9 gene editing to introduce specific mutations in cell models
Transgenic mouse models expressing mutant KRT16
Comparative transcriptomic analysis between wild-type and mutant tissues
KRT16 is extensively used as a biomarker for psoriasis, hidradenitis suppurativa, atopic dermatitis, and other inflammatory disorders . In these conditions, KRT16 expression is significantly upregulated in the suprabasal layers of the epidermis.
Methodological approach: To study KRT16 changes in inflammatory conditions, researchers can:
Perform immunohistochemistry on biopsy samples
Compare KRT16 mRNA levels using RT-qPCR in affected versus unaffected skin
Conduct single-cell RNA sequencing to identify cell populations with altered KRT16 expression
Use spatial transcriptomics to map KRT16 expression patterns in relation to inflammatory infiltrates
Correlate KRT16 expression levels with clinical severity scores
Krt16-null mice develop footpad lesions that mimic PC-associated palmoplantar keratoderma (PPK) . These models show similar transcriptional changes to human PC cases, with significant overlap in differentially expressed genes.
Methodological approach: When working with Krt16-null mouse models, researchers should:
Perform time-course analyses to track disease progression
Use histopathological assessment to characterize tissue architecture changes
Conduct transcriptomic profiling at different disease stages
Compare findings with human patient samples for translational relevance
Consider developing conditional knockout models for temporal control of gene deletion
Methodological approach: Multiple complementary techniques should be used:
Protein level detection:
Immunohistochemistry or immunofluorescence on tissue sections
Western blotting for semi-quantitative analysis
Flow cytometry for cell-level quantification
mRNA level detection:
RT-qPCR for targeted quantification
RNA-seq for genome-wide expression analysis
In situ hybridization for spatial localization
Single-cell approaches:
Single-cell RNA-seq for cellular heterogeneity
CyTOF for protein expression at single-cell resolution
Spatial transcriptomics for preserving tissue context
Research shows that K16 negatively regulates type-I interferon (IFN) signaling and innate immune responses . In mouse skin, loss of Krt16 leads to exacerbation of imiquimod-induced psoriasiform disease and heightened recruitment of neutrophils in phorbol ester-induced acute sterile inflammation models .
Methodological approach: To investigate this regulatory function, researchers should:
Use co-immunoprecipitation followed by mass spectrometry to identify K16 interaction partners
Employ proximity ligation assays to detect protein-protein interactions in situ
Compare inflammatory responses in wild-type versus Krt16-null cells/tissues
Analyze activation of immune signaling pathways using phospho-specific antibodies
Perform ChIP-seq to identify transcription factors regulated by K16-dependent pathways
K16 interacts with effectors of the RIG-I-like receptor (RLR) pathway, including 14-3-3ɛ, and inhibits the 14-3-3ɛ:RIG-I interaction upstream of IFN activation both in vivo and ex vivo . In KRT16 null human keratinocytes, loss of K16 amplifies IFN signaling including phospho-IRF7 and ISG15 after treatment with synthetic dsRNA poly(I:C) .
Methodological approach: To elucidate this mechanism:
Use CRISPR-Cas9 to generate KRT16 knockout in human keratinocytes
Treat cells with poly(I:C) to stimulate IFN pathways
Analyze phosphorylation status of key signaling molecules (IRF3, IRF7, STAT1)
Perform domain mapping to identify regions of K16 critical for interaction with 14-3-3ɛ
Reconstitute knockout cells with wild-type or mutant K16 to assess rescue effects
Research indicates that altered keratinocyte differentiation is an early driver of keratin mutation-based palmoplantar keratoderma . In Krt16-null mouse models, decreased Krt9 expression precedes the onset of lesions in paw skin, suggesting differentiation defects are an early event in pathogenesis .
Methodological approach: To study differentiation effects:
Perform time-course analysis of differentiation markers in Krt16-null versus wild-type models
Use 3D organotypic cultures to recapitulate differentiation in vitro
Apply single-cell RNA-seq to identify altered cell state transitions
Conduct calcium-shift assays to induce differentiation in cultured keratinocytes
Analyze expression of differentiation-associated genes using RT-qPCR and immunostaining
The relationship between different keratin genes during stress responses reveals complex regulatory mechanisms. In Krt16-null paw skin, Krt6a mRNA is significantly increased at 2, 4, and especially in 8-week-old mice relative to wild-type footpad skin . This induction increases over time and reaches very high levels in established PPK-like lesions .
Methodological approach: To investigate keratin gene relationships:
Perform comprehensive keratin profiling using RNA-seq in different conditions
Use ChIP-seq to identify shared regulatory elements between keratin genes
Apply CRISPR interference or activation to modulate expression of individual keratins
Analyze compensatory responses in knockout models
Study promoter activities using reporter assays
Spatial transcriptomics reveals that KRT16 is strongly expressed in the suprabasal layers in the epidermis of lesional skin, while IFN responsive genes like IFITM1, IFITM3, and CD74 are spatially restricted to the basal layer . This spatial separation appears critical for maintaining immune homeostasis.
Methodological approach: For spatial analysis:
Apply spatial transcriptomics platforms (e.g., 10x Visium) to skin biopsies
Use computational analysis to identify spatially co-regulated gene modules
Validate key findings with multiplexed RNA in situ hybridization
Integrate spatial data with single-cell RNA-seq for cell type resolution
Correlate spatial patterns with histopathological features
Transcriptomic comparisons show significant overlap between Krt16-null mouse footpad lesions and human PC cases. Of the 391 upregulated genes in mouse data, 22 are shared with KRT16 human cases (p = 5.0e-19) and 32 with KRT6 human cases (p = 1.4e-25) . Similarly, of 564 downregulated genes, 30 are shared with KRT16 human cases (p = 1.1e-16) and 9 with KRT6 human cases (p = 1.2e-6) .
Methodological approach: For comparative transcriptomics:
Use standardized RNA isolation and sequencing protocols
Apply robust bioinformatic pipelines for cross-species comparisons
Validate key differentially expressed genes by RT-qPCR
Perform pathway enrichment analysis to identify shared biological processes
Use network analysis to identify conserved regulatory hubs
Comparison | Upregulated Genes Shared | Downregulated Genes Shared | Statistical Significance |
---|---|---|---|
Krt16-null vs. KRT16 human cases | 22 | 30 | p = 5.0e-19 (up), p = 1.1e-16 (down) |
Krt16-null vs. KRT6 human cases | 32 | 9 | p = 1.4e-25 (up), p = 1.2e-6 (down) |
The N/TERT human keratinocyte cell line can be adapted to generate a post-confluent, uniformly bilayered epithelium in submerged culture under standard KSFM growth medium, without requiring 3D growth at the liquid-air interface . This system allows for the study of KRT16 function in a physiologically relevant context.
Methodological approach: For 3D model development:
Optimize culture conditions (media, substrate, cell density)
Validate model using immunostaining for differentiation markers
Apply CRISPR-Cas9 to generate KRT16 knockout or mutant lines
Test model responses to inflammatory stimuli (e.g., poly(I:C))
Compare findings with primary human skin samples for validation
Cytokeratin 16 is encoded by the KRT16 gene and is typically expressed in the basal layer of stratified epithelial tissues, such as the skin, hair follicles, and nails. The protein consists of 473 amino acids and has a molecular weight of approximately 50 kDa . It forms heterodimers with type II keratins, such as KRT6A and KRT6B, to create a filamentous network within the cytoplasm of epithelial cells.
The primary function of Cytokeratin 16 is to provide structural support to epithelial cells. It helps maintain the integrity of the skin and other epithelial tissues by forming a dense network of intermediate filaments. Additionally, Cytokeratin 16 plays a role in the regulation of cellular processes such as proliferation, differentiation, and apoptosis.
Mutations in the KRT16 gene are associated with several skin disorders, including:
Recombinant Cytokeratin 16 is produced using various expression systems, such as Escherichia coli and wheat germ . The recombinant protein is often tagged with His or GST tags to facilitate purification and detection. It is used in research to study the protein’s structure, function, and interactions with other cellular components.
Recombinant Cytokeratin 16 is utilized in several applications, including: