DYRK1A/HIPK2 complex: Acts as an essential adaptor for kinase-substrate interactions, enabling phosphorylation of adenovirus E1A protein .
IRS1-FOXO1 axis: Maintains AKT activation (phosphorylation at S473/T308) to suppress FOXO1-mediated transcription of G2 arrest genes (GADD45A, CCNG2) .
Developmental roles:
| Function | Mechanism | Outcome |
|---|---|---|
| Proliferation maintenance | Stabilizes IRS1-AKT signaling | Prevents G2/M arrest |
| Oncogenic transformation | Mediates E1A hyperphosphorylation | Enhances viral oncogenesis |
Nasopharyngeal carcinoma (NPC):
Hepatocellular carcinoma: DCAF7 depletion decreases HepG2 proliferation by 40% (Day 5 post-transfection) .
Linked to craniofacial abnormalities due to impaired palatoquadrate formation .
Source: E. coli-expressed His-tagged protein (1 mg/ml in Tris-HCl buffer)
Applications: Kinase interaction studies, antibody production
DDB1- and CUL4-associated factor 7, WD repeat-containing protein 68, WD repeat-containing protein An11 homolog, DCAF7, HAN11, WDR68, AN11, SWAN-1.
MGSSHHHHHH SSGLVPRGSH MVYAMNWSVR PDKRFRLALG SFVEEYNNKV QLVGLDEESS EFICRNTFDH PYPTTKLMWI PDTKGVYPDL LATSGDYLRV WRVGETETRL ECLLNNNKNS DFCAPLTSFD WNEVDPYLLG TSSIDTTCTI WGLETGQVLG RVNLVSGHVK TQLIAHDKEV YDIAFSRAGG GRDMFASVGA DGSVRMFDLR HLEHSTIIYE DPQHHPLLRL CWNKQDPNYL ATMAMDGMEV VILDVRVPCT PVARLNNHRA CVNGIAWAPH SSCHICTAAD DHQALIWD.
DCAF7 is characterized as an evolutionarily conserved protein with a single WD40 repeat domain and has no catalytic activity of its own . WD40 domains typically contain several WD40 repeats, which are sequences of 44–60 amino acids in length that often harbor a defining tryptophan-aspartate (WD) dipeptide . These repeats form a circularized β-propeller structure that provides multiple binding surfaces for diverse protein–protein interactions, which is consistent with DCAF7's role as a scaffolding protein . This structural arrangement is common among numerous scaffold proteins and facilitates DCAF7's function in mediating protein interactions rather than acting as an enzyme itself.
DCAF7 has been identified as a binding partner for several key proteins:
Kinase interactions: DCAF7 binds to DYRK1A, DYRK1B, and HIPK2 but notably not to HIPK1 or class 2 DYRKs (DYRK2, DYRK3, DYRK4) .
Insulin signaling components: DCAF7 interacts with IRS1 (Insulin Receptor Substrate 1) and the p85 subunit of PI3K, functioning as a scaffold in insulin signaling .
Viral proteins: DCAF7 mediates the interaction between adenovirus E1A protein and DYRK1A/HIPK2 .
These interactions are highly specific and evolutionarily conserved. The binding interfaces between DCAF7 and its partners have been mapped to specific regions in these proteins, demonstrating the specialized nature of these interactions .
DCAF7's function appears to be highly conserved throughout evolution. Research has demonstrated that:
Mouse DYRK1A, human DYRK1B, and Xenopus DYRK1B can all bind to zebrafish DCAF7 .
The DCAF7 binding motif identified in DYRK1A is functionally conserved in DYRK1 orthologs from Xenopus, Danio rerio, and even the slime mold Dictyostelium discoideum .
This high degree of evolutionary conservation suggests that DCAF7 plays a fundamental role in cellular processes that have been maintained throughout evolution, emphasizing its biological significance . Studies in various model systems including Drosophila (where DCAF7 is known as Wap) show that loss or mutation of DCAF7 is associated with growth retardation, confirming its essential role across diverse species .
DCAF7 acts as a scaffold for IRS1-PI3K that is required for insulin signaling to AKT and FOXO1 inhibition . Experimental evidence supports this role:
Scaffold function: DCAF7 interacts with IRS1 and the p85 subunit of PI3K, suggesting it brings these proteins together to facilitate signaling .
Effect on AKT phosphorylation: Knockdown of DCAF7 significantly diminishes insulin-stimulated AKT S473 and T308 phosphorylation, while not affecting ERK phosphorylation, demonstrating specificity for the PI3K-AKT pathway .
IRS1 phosphorylation: Following DCAF7 knockdown, phosphorylation of S612 in IRS1 increases. Since S612 is a known inhibitory site, this may explain the observed decrease in insulin signaling .
The role of DCAF7 as an IRS1 scaffolding protein is further supported by the finding that knockdown of DCAF7 decreases the interaction of IRS1 with DYRK1A, demonstrating its importance in facilitating protein complexes necessary for proper signaling .
DCAF7 serves as an adaptor protein for DYRK1A and mediates interactions between DYRK1A and its substrates:
Complex formation: DCAF7 binds to a conserved 12-amino acid motif in the N-terminal domain of DYRK1A, forming a stable complex at physiological expression levels .
Substrate recruitment: DCAF7 functions as a substrate recruiting subunit for DYRK1A . This is evidenced by experiments showing that DCAF7 is required for the hyperphosphorylation of the adenovirus E1A protein in DYRK1A overexpressing cells .
Binding interface: The interaction does not depend on DYRK1A's catalytic activity, as kinase-deficient mutants maintain DCAF7 binding .
This relationship appears to be essential for proper DYRK1A function, as DCAF7 helps bring together DYRK1A and its substrates, facilitating efficient phosphorylation .
DCAF7 plays a critical role in regulating cell proliferation through its effects on cell cycle progression:
Cell number effects: Knockdown of DCAF7 significantly reduces cell number over time, with measurable differences observed by days 4-5 post-transfection .
Cell cycle arrest: Flow cytometry analysis reveals that DCAF7 knockdown induces cell cycle arrest specifically at the G2 phase, similar to the effect of Dinaciclib, a cyclin-dependent kinase inhibitor .
Molecular mechanism: DCAF7 knockdown leads to:
This regulatory mechanism appears to be conserved, as studies in Drosophila show that knockdown of the DCAF7 ortholog (wap) reduces wing area and cell number through a dFOXO-dependent mechanism, mirroring observations in flies lacking chico (the IRS ortholog) .
Several experimental approaches have proven effective for investigating DCAF7's interactions:
Co-immunoprecipitation (co-IP): This has been successfully used to demonstrate interactions between DCAF7 and its partners. Both overexpressed tagged proteins and endogenous proteins can be immunoprecipitated to verify interactions . For example, immunoprecipitation of endogenous DYRK1A from HeLa cells confirmed interaction with DCAF7 at physiological expression levels .
GST pulldown assays: Recombinant GST-fused proteins (like E1A-X2) can be used as bait to pull down DCAF7 and its binding partners from cell lysates . This approach helps identify direct versus indirect interactions.
In vitro translation: Producing proteins like GFP-DCAF7 by in vitro translation for use as prey in GST pulldown experiments can further verify direct binding to partners like E1A and DYRK1A .
Deletion mapping: Creating truncated constructs of DCAF7's binding partners has been effective in mapping specific binding domains. For example, this approach identified that amino acids 1-135 of HIPK2 contain the DCAF7 binding site, while a shorter construct (amino acids 1-114) does not bind DCAF7 .
Site-directed mutagenesis: Point mutations within potential binding interfaces can confirm critical residues. The substitution of Thr125 by proline abolished DCAF7 binding to HIPK2, demonstrating the importance of this residue .
These methodologies, especially when used in combination, provide robust evidence for DCAF7's role as an adaptor protein in various complexes.
Researchers have employed several techniques to investigate DCAF7's impact on cell proliferation:
siRNA knockdown: Efficient reduction of DCAF7 protein abundance can be achieved using siRNA transfection, with effects observable up to 5 days post-transfection .
Cell counting assays: Direct measurement of cell number over time following DCAF7 knockdown demonstrates its effect on proliferation .
Flow cytometry:
Western blotting: Analysis of phosphorylation status of key signaling proteins (AKT, ERK, IRS1) after insulin stimulation in DCAF7-depleted cells reveals pathway-specific effects .
Immunofluorescence: Subcellular localization of factors like FOXO1 can be visualized following DCAF7 knockdown to determine nuclear versus cytoplasmic distribution .
RT-qPCR: Expression analysis of FOXO1-target genes involved in cell cycle regulation (GADD45A, CCNG2, CDKN1B, RBL2) provides mechanistic insight into how DCAF7 affects proliferation .
These methodological approaches provide complementary data to establish DCAF7's role in regulating cell proliferation through specific signaling pathways.
Although DCAF7's name suggests a function in E3 ubiquitin ligase complexes, the research presents a nuanced picture:
Potential role: DCAF7 is described as a substrate-specific adaptor of the DDB1-Cul4 E3 ubiquitin ligase complex, potentially promoting proteasomal degradation of targeted proteins .
Proteasome-independent functions: DCAF7 also regulates interactor levels independent of the proteasome . For instance, knockdown of DCAF7 did not affect IRS1 protein abundance, suggesting a functional role for the DCAF7-IRS1 interaction rather than mediating protein stability .
Research limitations: While DCAF7's name (DDB1 and CUL4 associated factor 7) refers to its identification in DDB1 complexes and its deduced function as a substrate receptor in CLR4 E3 ubiquitin ligase complexes, this specific role has not been fully experimentally verified .
Understanding this aspect of DCAF7 function remains an area where additional research is needed to clarify its precise role in protein degradation pathways versus its adaptor/scaffold functions.
DCAF7 shows remarkable specificity in its kinase interactions:
Class 1 vs. Class 2 DYRKs: DCAF7 binds to class 1 DYRKs (DYRK1A, DYRK1B) but not to class 2 DYRKs (DYRK2, DYRK3, DYRK4) . This specificity correlates with sequence conservation in the N-terminal region of class 1 DYRKs, which diverges in class 2 DYRKs.
HIPK1 vs. HIPK2: Despite HIPK1 and HIPK2 being closely related with the same domain architecture, DCAF7 specifically binds to HIPK2 but not HIPK1 . This selectivity was traced to critical residues in the binding interface - specifically, the presence of a proline in HIPK1 at a position corresponding to Thr125 in HIPK2 disrupts binding .
Binding motif conservation: The DCAF7 binding motif consists of 12 amino acids in DYRK1A (amino acids 93-104) with a similar sequence essential for DCAF7 binding to HIPK2 .
Kinase activity independence: The interaction of DCAF7 with both DYRK1A and HIPK2 does not depend on the catalytic activity of these kinases, as binding is maintained with kinase-deficient mutants .
This binding specificity highlights DCAF7's evolved role as a selective adaptor for specific kinases, suggesting specialized functions in distinct signaling pathways.
The literature presents some nuanced findings regarding DCAF7's effects on cell viability versus proliferation:
Temporal progression: DCAF7 knockdown initially causes cell cycle arrest (observable early) followed by compromised cell viability at later timepoints (days 4-5 post-transfection) . This suggests a sequential rather than contradictory relationship between these observations.
Methodological considerations:
Experimental design recommendations:
Conduct time-course experiments to distinguish early versus late effects
Include appropriate positive controls (e.g., Dinaciclib for cell cycle arrest at G2, Staurosporine for cell death)
Use multiple cell lines to ensure findings are not cell-type specific
Employ rescue experiments with wild-type DCAF7 to confirm specificity of knockdown effects
Mechanistic relationship: The data suggests that prolonged G2 arrest following DCAF7 knockdown eventually leads to cell death, consistent with known cellular responses to sustained cell cycle checkpoint activation .
Understanding this relationship requires careful experimental design with appropriate time points and complementary methodologies to distinguish primary from secondary effects.
Despite significant progress in understanding DCAF7, several important questions remain:
E3 ligase function: Though DCAF7 is named as a CUL4 associated factor, its precise role in ubiquitin ligase complexes requires further verification and characterization .
Substrate specificity: While DCAF7 functions as a substrate recruiting subunit for DYRK1A and HIPK2, the complete set of substrates it helps recruit remains to be fully characterized .
Tissue-specific functions: Most studies have used cell lines or model organisms; the tissue-specific roles of DCAF7 in human physiology and disease remain largely unexplored.
Disease relevance: The potential contribution of DCAF7 dysfunction to human diseases, particularly those involving insulin signaling, cell cycle regulation, or DYRK1A function (e.g., neurodevelopmental disorders) warrants investigation.
Therapeutic targeting: Given DCAF7's role as an adaptor protein in multiple signaling pathways, its potential as a therapeutic target for modulating these pathways remains an open question.
Addressing these questions will require integrative approaches combining structural biology, proteomics, genetic models, and clinical studies.
Future research on DCAF7 could benefit from several cutting-edge approaches:
CRISPR-based technologies:
CRISPR-Cas9 knockout and knockin models to study DCAF7 function in various cell types
CRISPRi/CRISPRa for tunable control of DCAF7 expression
Base editors for introducing specific mutations in binding interfaces
Proximity labeling proteomics:
BioID or APEX2 fusion proteins to identify the complete DCAF7 interactome under various cellular conditions
TurboID for temporal resolution of dynamic interactions
Structural biology:
Cryo-EM structures of DCAF7 complexes with various binding partners
Hydrogen-deuterium exchange mass spectrometry to map binding interfaces
Single-cell approaches:
Single-cell RNA-seq to understand cell-type-specific responses to DCAF7 modulation
Live-cell imaging with fluorescent reporters to visualize DCAF7-dependent signaling dynamics
Animal models:
Conditional tissue-specific knockout models to assess DCAF7 function in specific physiological contexts
Humanized mouse models expressing human DCAF7 variants
These methodological advances would provide deeper insights into DCAF7's structure, interactions, and functions in normal physiology and disease states.
DCAF7 is a scaffold protein that is essential for the formation of protein complexes involved in kinase signaling. It has been shown to interact with DYRK1A, a kinase located on chromosome 21, which is significant in the context of Down syndrome . The protein is also involved in craniofacial development, acting upstream of the EDN1 pathway and is required for the formation of the upper jaw equivalent .
Recent studies have highlighted the importance of DCAF7 in hematopoiesis. Mutations in DCAF7 have been detected in a subset of patients with Myeloid Leukemia in Down syndrome. The absence of DCAF7 in hematopoietic cell lines leads to cell cycle arrest and decreased proliferation . This protein is crucial for the differentiation and proliferation of hematopoietic stem cells (HSCs) and progenitor cells. In conditional knockout mouse models, the deletion of DCAF7 resulted in a decrease in white blood cells, hemoglobin, and platelet counts, indicating its essential role in hematopoiesis .
DCAF7 is associated with several diseases, including Dystonia 35, Childhood-Onset, and Noonan Syndrome 3 . Its role in kinase signaling and protein folding pathways makes it a potential target for therapeutic interventions. The protein’s involvement in craniofacial development and hematopoiesis further underscores its clinical significance.
Recombinant human DCAF7 is produced using advanced biotechnological methods. The gene encoding DCAF7 is cloned into an expression vector, which is then introduced into a suitable host cell line, such as E. coli or CHO cells. The recombinant protein is expressed, purified, and characterized to ensure its functionality and structural integrity. This recombinant protein is used in various research applications, including studies on protein-protein interactions, kinase signaling pathways, and hematopoiesis.