PDZK1 (PDZ Domain Containing 1), also known as NHERF3 or CAP70, is a scaffolding protein encoded by the PDZK1 gene on human chromosome 1q21. It contains four PDZ domains that mediate interactions with C-terminal motifs of membrane transporters and receptors, enabling cellular localization and functional regulation . Recombinant PDZK1 Human (e.g., produced in E. coli) is widely used to study its structural, functional, and therapeutic roles .
Four PDZ domains: Facilitate binding to C-terminal motifs of partner proteins (e.g., SR-BI, OATP1A2) .
Extended L-shaped conformation: Full-length PDZK1 adopts an asymmetric arrangement critical for scaffolding, contrasting earlier "beads-on-string" predictions .
OATP1A2 modulation: Enhances plasma membrane localization and stability of the organic anion transporter, increasing substrate uptake (e.g., estrone-3-sulfate) by 1.6–1.8-fold .
SR-BI interaction: Maintains hepatic HDL receptor stability; PDZK1 knockout mice show 95% reduced SR-BI levels and abnormal HDL metabolism .
AKT/mTOR activation: Binds AKT1 to promote glioma progression .
EGFR regulation: Suppresses EGFR expression and phosphorylation, sensitizing triple-negative breast cancer (TNBC) cells to erlotinib .
Cholesterol homeostasis: Regulates SR-BI-mediated HDL uptake in hepatocytes .
Visual function: Zebrafish pdzk1 knockouts exhibit retinal dysfunction and altered optomotor responses .
Transcriptional control: HNF1α binds the PDZK1 promoter (-25 to -39 bp), enhancing mRNA expression in renal cells .
Post-translational regulation: PDZ domain interactions stabilize partner proteins (e.g., OATP1A2) and modulate clathrin-dependent internalization .
Recombinant PDZK1: Used in binding assays, structural studies, and functional screens (e.g., ProSpec Bio, Cat# PRO-1447) .
Animal models: Pdzk1-KO mice exhibit hypercholesterolemia and HDL abnormalities ; zebrafish mutants show visual defects .
PDZK1 is an adaptor protein containing four PDZ protein interaction domains that was first identified in human carcinomas. It has a molecular weight of approximately 70 kDa and contains four PDZ domains followed by a C-terminal region . Each PDZ domain consists of about 80-90 amino acids that fold into a compact structure characterized by six β-strands and two α-helices . The human genome encodes over 250 PDZ domains incorporated into more than 100 proteins, with PDZK1 being one of the significant members of this protein family .
PDZ domains typically interact with the five most carboxy-terminal amino acids of their binding partners, though some recognize up to seven carboxy-terminal residues . PDZK1's structure allows it to act as a scaffolding protein that can promote clustering of groups of proteins, often cell surface proteins like receptors and ion channels.
PDZK1 typically interacts with membrane proteins through its PDZ domains, which recognize and bind to specific PDZ-binding motifs in the C-terminal regions of target proteins . In the case of SR-BI (Scavenger Receptor Class B Type 1), the PDZ1 domain of PDZK1 is responsible for binding to its C-terminal PDZ-binding motif .
For OATP1A2 (Organic Anion Transporting Polypeptide 1A2), PDZK1 interacts with its C-terminal PDZ-binding domain (residues 667-670, sequence KTKL) . This interaction can be confirmed through co-immunoprecipitation experiments, which show that PDZK1 directly binds to OATP1A2 in cell models . The interaction is specific and functionally significant, as deletion of the PDZ-binding domain abolishes the regulatory effects of PDZK1 on target proteins.
For studying PDZK1 function, several experimental systems have proven effective:
Cell Culture Models: HEK-293 cells transfected with PDZK1 and its target proteins (e.g., OATP1A2, SR-BI) provide a reliable system for studying protein-protein interactions and functional effects . This approach allows for controlled expression and manipulation of proteins of interest.
Animal Models: PDZK1 knockout mice have been instrumental in understanding the physiological roles of PDZK1, particularly in lipoprotein metabolism and cardiovascular function . These models reveal that PDZK1 is essential for maintaining hepatic SR-BI levels and controlling HDL metabolism.
Transgenic Models: Mice expressing specific domains of PDZK1 (e.g., PDZ1, PDZ1.2, PDZ1.2.3, or PDZ1.2.3.4) have helped determine the roles of individual PDZ domains in PDZK1 function . These models showed that all four PDZ domains, but not the C-terminal region, are necessary for normal hepatic regulation of SR-BI.
Co-immunoprecipitation Assays: These are crucial for confirming direct protein-protein interactions between PDZK1 and its target proteins . Using tagged constructs (e.g., N-myc tagged PDZK1 and N-flag tagged target proteins) improves specificity of detection.
To validate PDZK1 interactions with target proteins, researchers should employ multiple complementary techniques:
Co-immunoprecipitation (Co-IP): This technique directly demonstrates physical interaction between PDZK1 and its target proteins. For example, HEK-293 cells co-expressing N-flag tagged OATP1A2 and N-myc tagged PDZK1 can be lysed and subjected to immunoprecipitation with anti-flag antibody, followed by immunoblotting with anti-myc antibody to detect PDZK1 .
Domain Deletion Studies: Creating mutants lacking the PDZ-binding domain of target proteins (e.g., OATP1A2-del mutant lacking residues 667-670) helps confirm the specificity of interaction and identify critical binding domains .
Functional Assays: Measuring changes in target protein function (e.g., substrate uptake by OATP1A2) following PDZK1 co-expression provides functional validation of the interaction. For example, estrone-3-sulfate uptake increases approximately 1.6-fold when PDZK1 is co-expressed with OATP1A2 .
Cellular Localization Studies: Immunofluorescence or cell surface biotinylation assays can determine whether PDZK1 affects the subcellular localization of target proteins, which further validates functional interaction .
To measure PDZK1-mediated effects on protein stability, researchers can employ several methodological approaches:
Protein Synthesis Inhibition: Treating cells with protein synthesis inhibitors like puromycin allows researchers to track the degradation rate of target proteins in the presence or absence of PDZK1 . This reveals whether PDZK1 affects protein stability over time.
Pulse-Chase Experiments: These experiments involve metabolic labeling of proteins (pulse) followed by a chase period to monitor protein turnover rates. This approach can quantitatively assess how PDZK1 affects the half-life of target proteins.
Western Blotting Time Course: After inhibiting protein synthesis, collecting cell lysates at various time points and performing Western blotting for the target protein can reveal degradation kinetics. The ~95 kDa isoform of OATP1A2, for example, shows a significantly decreased degradation rate when co-expressed with PDZK1 .
Proteasome and Lysosome Inhibitors: Using specific inhibitors of degradation pathways can help determine which pathway is affected by PDZK1. This approach helps elucidate the mechanism by which PDZK1 enhances protein stability.
The four PDZ domains of PDZK1 play distinct but coordinated roles in its regulatory function:
Recent evidence suggests that PDZ3 may be involved in PDZK1 dimerization, while the C-terminus may associate with PDZ1 in intramolecular interactions .
PDZK1 regulates membrane protein trafficking through several mechanisms:
Reduced Internalization: PDZK1 decreases the internalization of membrane proteins like OATP1A2 in a clathrin-dependent manner . This regulation occurs post-translationally and affects the dynamic equilibrium of proteins at the cell surface.
Caveolin-Independent Mechanism: While PDZK1 regulates protein internalization through clathrin-dependent pathways, this process appears to be independent of caveolin .
Enhanced Protein Stability: PDZK1 enhances the stability of target proteins, reducing their degradation rate . This effect contributes to increased steady-state levels of membrane proteins at the cell surface.
Subcellular Localization Control: In PDZK1 knockout mice expressing only the PDZ1 domain, SR-BI is mislocalized in the cytoplasm instead of being properly associated with the plasma membrane . This indicates that PDZK1, through its multiple PDZ domains, ensures proper subcellular targeting of its partner proteins.
The combined effect of these mechanisms results in increased expression and function of target membrane proteins, as evidenced by enhanced substrate uptake when PDZK1 is co-expressed with transporters like OATP1A2 .
PDZK1 plays important roles in cardiovascular disease pathophysiology through multiple mechanisms:
HDL Metabolism Regulation: PDZK1 is essential for maintaining hepatic SR-BI levels, which control HDL metabolism . PDZK1 knockout mice exhibit hypercholesterolemia due to large HDL particles, resembling the phenotype of SR-BI knockout mice .
Atheroprotection: PDZK1 protects against the development of atherosclerosis in murine models . This protection is likely mediated through its regulation of SR-BI, which facilitates reverse cholesterol transport.
Endothelial Cell Regulation: PDZK1 mediates SR-BI-dependent regulation of endothelial cell biology by HDL . This suggests that PDZK1 influences vascular function beyond its effects on lipoprotein metabolism.
Tissue-Specific Effects: While PDZK1 is crucial for SR-BI expression in the liver, it doesn't appear to significantly affect SR-BI expression in steroidogenic tissues . This tissue-specific regulation may have important implications for cardiovascular disease progression.
The exploration of PDZK1 structure and function may provide additional insights into HDL metabolism and could form the basis for new therapeutic approaches for cardiovascular disease .
For optimal study of PDZK1-protein interactions, researchers should consider the following experimental conditions:
Expression Systems: HEK-293 cells provide a reliable expression system for studying PDZK1 interactions . These cells have low endogenous expression of many membrane transporters and are easily transfectable.
Protein Tagging Strategies: N-terminal tagging with epitope tags like Flag (for target proteins) and myc (for PDZK1) enables specific detection without interfering with C-terminal PDZ-binding interactions . Verify that the tags do not affect protein function through appropriate control experiments.
Co-expression Ratios: Optimize the ratio of PDZK1 to target protein expression to avoid artifacts from extreme overexpression. A 1:1 ratio of expression plasmids is often a good starting point, but titration experiments may be necessary.
Cell Lysis Conditions: Use mild detergents (e.g., 1% Triton X-100) for cell lysis to preserve protein-protein interactions. Include protease inhibitors to prevent degradation during sample processing.
Control Experiments: Always include appropriate controls, such as:
Target proteins with deleted PDZ-binding domains
Unrelated PDZ proteins that should not interact with your target
Empty vector controls to establish baseline expression and function
Distinguishing between direct and indirect effects of PDZK1 on target proteins requires a multi-faceted approach:
In Vitro Binding Assays: Purified recombinant proteins can be used in pull-down assays to demonstrate direct physical interaction without cellular cofactors.
Domain Mapping: Systematic mutation or deletion of binding domains in both PDZK1 and target proteins helps establish which domains are essential for interaction and functional effects .
Subcellular Localization Studies: Colocalization studies using confocal microscopy can reveal whether PDZK1 and target proteins occupy the same subcellular compartments, supporting direct interaction.
Kinetic Analysis: For transporters like OATP1A2, measuring changes in kinetic parameters (Vmax and Km) can help distinguish between effects on protein abundance versus intrinsic activity . PDZK1 co-expression with OATP1A2 increases Vmax from 55.5±3.2 to 138.9±4.1 pmol*(μg*4 min)⁻¹ without significantly affecting Km, suggesting an effect on expression rather than transport mechanism .
Scaffold Protein Analysis: Investigate whether other proteins are involved in the PDZK1-target protein complex using techniques like mass spectrometry after co-immunoprecipitation.
Developing PDZK1-targeted therapeutic strategies requires careful consideration of several factors:
Several emerging technologies hold promise for advancing PDZK1 research:
CRISPR/Cas9 Genome Editing: This technology enables precise modification of endogenous PDZK1 or its target genes, allowing study of physiological expression levels and natural regulation.
Proximity Labeling Techniques: Methods like BioID or APEX2 could identify the complete interactome of PDZK1 in different cellular contexts by labeling proteins that come into close proximity with PDZK1 in living cells.
Single-Cell Analysis: Single-cell RNA sequencing and proteomics could reveal cell-type-specific expression patterns and functions of PDZK1 that are masked in bulk tissue analyses.
Structural Biology Approaches: Cryo-electron microscopy and X-ray crystallography could elucidate the three-dimensional structure of PDZK1 in complex with its binding partners, providing insights into the molecular mechanisms of interaction.
In Silico Modeling: Recent advances in predicting PDZ domain binding specificity based on primary sequence could facilitate identification of novel PDZK1 binding partners .
Despite significant progress, several questions about PDZK1 in disease states remain unresolved:
Clinical Relevance: Whether naturally occurring variations in human PDZK1 structure and function influence SR-BI and have clinical consequences (e.g., risk for atherosclerotic or other cardiovascular diseases) is still unknown .
Relative Importance of Different Cell Types: The relative importance of hepatic versus endothelial PDZK1 in atheroprotection has not been established .
Molecular Mechanisms: The precise molecular and cellular mechanisms by which PDZK1 mediates tissue-specific regulation of SR-BI are not fully understood .
Therapeutic Potential: Whether modulation of PDZK1 function could provide therapeutic benefits in cardiovascular or other diseases remains to be determined.
Interplay with Other Regulators: How PDZK1 function is integrated with other regulatory pathways that control membrane protein expression and function in health and disease states requires further investigation.
Computational approaches offer several advantages for advancing PDZK1 research:
Binding Prediction Models: Recently described models designed to predict the target binding sequence of a PDZ domain based on that domain's primary sequence can facilitate exploration of the roles of PDZ2-4 in binding other cellular components .
Molecular Dynamics Simulations: These can model the conformational changes that occur when PDZK1 interacts with binding partners, providing insights into the mechanisms of regulation.
Systems Biology Approaches: Integration of proteomics, transcriptomics, and functional data can reveal how PDZK1 functions within larger cellular networks and how these networks are perturbed in disease states.
Drug Design Platforms: In silico screening and structure-based drug design could identify small molecules that modulate specific PDZK1-protein interactions, potentially leading to novel therapeutic approaches.
Machine Learning Algorithms: These could identify patterns in experimental data that might not be apparent through conventional analysis, potentially revealing new aspects of PDZK1 biology.
By combining these computational approaches with experimental validation, researchers can develop a more comprehensive understanding of PDZK1 function and its therapeutic potential.
PDZ domains typically consist of 80-90 amino acids forming 5-6 β-strands and 2-3 α-helices . They are involved in anchoring receptor proteins in the membrane to cytoskeletal components, thus organizing signaling complexes at cellular membranes . This structural arrangement allows PDZ domains to bind to the C-terminal sequences of their target proteins through a mechanism known as beta sheet augmentation .
PDZ domains are abundant in various species, including bacteria, yeast, plants, viruses, and animals . They regulate multiple biological processes such as transport, ion channel signaling, and other signal transduction systems . In humans, PDZ domain-containing proteins are involved in critical functions like cell proliferation, cell polarity, and the formation of signal transduction complexes .
PDZ Domain Containing 1 (PDZK1) is a specific protein that contains PDZ domains. It is known for its role in the regulation of various transporters and channels. PDZK1 interacts with several membrane proteins, including ion channels and transporters, thereby influencing their localization and function .
Human recombinant PDZK1 is a laboratory-produced version of the naturally occurring PDZK1 protein. Recombinant proteins are typically produced using techniques such as gene cloning and expression in host cells like bacteria or yeast. These recombinant proteins are used in research to study protein-protein interactions, signaling pathways, and potential therapeutic targets.
The study of PDZ domains and their interactions has been greatly enhanced by proteomic methods such as large-scale protein arrays and peptide libraries . These methods have provided insights into the binding properties and regulatory mechanisms of PDZ-mediated interactions, which are essential for understanding many cellular and biological processes .