The GRK5 Antibody (Catalog # AF4539) is a polyclonal antibody designed to detect human, mouse, and rat G protein-coupled receptor kinase 5 (GRK5). It is derived from E. coli-expressed recombinant human GRK5 (Pro463-Ser590) and is widely used in molecular biology research to study GRK5’s role in cellular signaling, cancer, inflammation, and neurodegenerative diseases. This antibody is validated for Western blot, immunoprecipitation, and immunocytochemistry (ICC), making it a versatile tool for protein expression analysis.
GRK5 is implicated in oncogenesis, with studies using the AF4539 antibody demonstrating its overexpression in non-small-cell lung cancer (NSCLC) and glioblastoma multiforme (GBM) . Western blot analysis with this antibody revealed GRK5 knockdown-induced G2/M cell cycle arrest and apoptosis in NSCLC cells, highlighting its role in tumor progression .
In rheumatoid arthritis (RA) models, GRK5 inhibition via this antibody correlated with reduced synovitis and cartilage degradation. Immunohistochemistry confirmed GRK5 localization in synovial lining cells, with knockout mice showing lower IL-6 production and NF-κB activity .
ICC studies using AF4539 demonstrated GRK5’s role in neuronal morphogenesis. Overexpression of GRK5 promoted F-actin bundling and membrane targeting, critical for dendritic spine formation .
GRK5 (G protein-coupled receptor kinase 5) is a serine/threonine kinase that phosphorylates preferentially the activated forms of various G protein-coupled receptors (GPCRs). It plays a crucial role in regulating GPCR signaling through agonist-mediated desensitization, which maintains cellular responsiveness to external signals . This desensitization occurs through phosphorylation of activated receptors, primarily mediated by GRKs including GRK5 . GRK5 specifically phosphorylates receptors in their active state, ensuring signaling pathways can be finely tuned, preventing overstimulation and allowing cells to adapt to prolonged exposure to agonists . Beyond its canonical role in GPCR regulation, GRK5 also phosphorylates non-GPCR substrates such as α-synuclein, tubulin, and p53, expanding its functional significance .
Research on GRK5 is particularly important because dysregulation of this kinase has been implicated in various pathophysiological conditions, including heart disease, neurological disorders, and Alzheimer's disease .
GRK5 can be detected in most tissues, with the highest levels found in heart, lung, and placenta . In comparison to cardiovascular tissues, GRK5 content in the brain is less abundant, though its importance in neurological function is significant . For cellular models, validated cell lines for GRK5 detection include:
When selecting tissues or cell lines for GRK5 studies, researchers should consider that GRK5 expression patterns may vary based on physiological or pathological states, and expression levels might change in response to various stimuli or treatments .
Optimal dilutions for GRK5 antibodies vary by application and specific antibody. Based on the provided data from multiple vendors, the following ranges are recommended starting points:
For Western blotting, start with a moderate dilution (e.g., 1:1000) and adjust based on signal intensity and background. When performing immunohistochemistry on paraffin-embedded sections, antigen retrieval is critical – several antibodies recommend TE buffer pH 9.0 or citrate buffer pH 6.0 . For immunofluorescence, higher concentrations (e.g., 25 μg/ml) may be necessary for optimal visualization . Always perform a dilution series in preliminary experiments to determine the optimal concentration for your specific experimental conditions and samples.
Proper validation of GRK5 antibodies requires several controls:
Positive controls: Use tissues or cell lines known to express GRK5, such as:
Negative controls:
Specificity controls:
The most definitive validation comes from showing antibody reactivity in wild-type samples that is absent in GRK5 knockout samples. This approach has been successfully demonstrated for certain GRK5 antibodies, where a specific band for GRK5 was detected at approximately 65 kDa in parental HeLa cells but was not detectable in a GRK5 knockout HeLa cell line .
Distinguishing between cytosolic and membrane-bound GRK5 is crucial because the protein's localization affects its function. GRK5 shuttles between membrane and cytosolic compartments, with its membrane association being important for GPCR phosphorylation . To differentiate between these pools:
Cell fractionation approach:
Perform subcellular fractionation to separate membrane and cytosolic fractions
Use Western blotting with GRK5 antibody on each fraction
Include compartment-specific markers (e.g., Na+/K+ ATPase for membrane, GAPDH for cytosol)
Immunofluorescence approach:
Use confocal microscopy with GRK5 antibody
Co-stain with membrane markers (e.g., WGA, CD44) and nuclear markers (DAPI)
Analyze co-localization coefficients
Biochemical approach:
When interpreting results, consider that stimuli like amyloid-β can cause rapid (within minutes) GRK5 membrane dissociation, leading to membrane GRK5 deficiency while increasing cytosolic GRK5 . This translocation may affect downstream signaling and has implications for conditions like Alzheimer's disease .
When investigating GRK5's role in neurodegenerative conditions like Alzheimer's disease (AD) and mild cognitive impairment (MCI), several critical methodological considerations emerge:
Expression level analysis:
Cellular localization:
Model selection:
Antibody selection:
Experimental design:
Include age-matched controls as GRK5 expression may change with aging
Consider both acute and chronic experimental paradigms to capture dynamic changes in GRK5 localization and function
The connection between GRK5 deficiency and MCI/AD suggests that GRK5 may serve as a prophylactic therapeutic target, making accurate detection and quantification particularly important .
When encountering difficulties detecting GRK5 in Western blot experiments, consider these methodological approaches:
Sample preparation optimization:
Ensure complete lysis with appropriate buffers (RIPA or NP-40 based buffers are commonly used)
Include protease and phosphatase inhibitors to prevent degradation
Avoid repeated freeze-thaw cycles of samples
Protein amount and loading:
Antibody selection and protocol adjustment:
Tissue-specific considerations:
Positive controls:
Specific detection systems:
For low abundance samples, use high-sensitivity detection reagents like enhanced chemiluminescence (ECL) Plus or SuperSignal West Femto
Consider using fluorescent secondary antibodies with digital imaging systems for better quantification
If signal remains problematic after these optimizations, verify target expression using mRNA analysis (RT-PCR) to confirm GRK5 expression in your samples before further antibody troubleshooting.
Selecting between monoclonal and polyclonal GRK5 antibodies depends on your specific research needs:
Monoclonal GRK5 Antibodies:
Advantages:
High specificity for a single epitope
Reduced batch-to-batch variation
Lower background in most applications
Example: GRK5 Antibody (D-9) is a mouse monoclonal IgG2a kappa light chain antibody
Limitations:
May have reduced sensitivity due to recognition of a single epitope
More susceptible to epitope masking by fixation or denaturation
Less robust to antigen conformational changes
May not work across multiple species unless the epitope is highly conserved
Polyclonal GRK5 Antibodies:
Advantages:
Higher sensitivity due to recognition of multiple epitopes
More tolerant to protein denaturation or fixation
Better for detecting proteins with low expression levels
Often work well across multiple species (e.g., human, mouse, rat)
Limitations:
Potential for higher background
Batch-to-batch variation may require revalidation
May exhibit some non-specific binding
Application-Specific Recommendations:
| Application | Recommended Antibody Type | Rationale |
|---|---|---|
| Western Blot | Either type suitable | Polyclonals may be more sensitive; monoclonals may give cleaner background |
| IHC/IF for localization studies | Monoclonal | Higher specificity for subcellular localization |
| Co-IP studies | Monoclonal | Reduced background binding to other proteins |
| Cross-species detection | Polyclonal | Greater likelihood of recognizing conserved epitopes |
| Detecting GRK5 in brain tissue | Polyclonal | Higher sensitivity for low-abundance targets |
When possible, validate important findings with both antibody types to ensure robust results.
Beyond its canonical role in GPCR desensitization, GRK5 has several non-GPCR substrates and functions that can be investigated using specialized antibody-based approaches:
Nuclear localization and transcriptional regulation:
Use subcellular fractionation followed by Western blotting to quantify nuclear versus cytoplasmic GRK5
Employ co-immunoprecipitation with GRK5 antibodies to identify nuclear binding partners
Combine with ChIP assays to study GRK5's direct interaction with DNA
α-synuclein phosphorylation:
p53 regulation:
HDAC5 phosphorylation and MEF2 signaling:
LRP6 and Wnt signaling:
For each of these applications, appropriate controls are essential, including GRK5 knockout or knockdown systems , kinase-dead GRK5 mutants, and specificity validation through competing peptides or alternative antibodies targeting different epitopes.
GRK5 plays significant roles in cardiovascular pathophysiology, and several antibody-based methodological approaches can advance this research:
Cardiac hypertrophy models:
Monitor GRK5 nuclear accumulation in response to hypertrophic stimuli using immunofluorescence
Quantify changes in GRK5 expression levels via Western blot in normal versus hypertrophic heart tissue
Use cardiac-specific GRK5 genetic models (overexpression or knockout) to assess functional outcomes
β-adrenergic receptor (βAR) desensitization:
Measure GRK5-mediated βAR phosphorylation using phospho-specific receptor antibodies
Track GRK5 membrane recruitment in response to βAR agonists via fractionation and immunoblotting
Assess receptor internalization via immunofluorescence co-localization studies
GRK5 polymorphism studies:
Blood pressure regulation:
Cardiomyocyte-specific analysis:
Perform immunohistochemistry with GRK5 antibodies on cardiac sections
Co-stain with cardiomyocyte markers to assess cell-type specific expression
Use isolated primary cardiomyocytes for more detailed subcellular localization studies
When conducting cardiovascular GRK5 research, it's important to consider the dynamic nature of GRK5 localization and its response to various stimuli. Changes in subcellular distribution may be as important as changes in total expression levels, requiring careful experimental design and interpretation.
Investigating GRK5-mediated receptor phosphorylation requires specialized methodological approaches:
Phospho-specific antibody selection:
Use antibodies targeting specific phosphorylation sites on GPCRs known to be GRK5 substrates
For receptors without available phospho-specific antibodies, consider using general phospho-serine/threonine antibodies after immunoprecipitation of the receptor
Temporal dynamics:
Specificity controls:
Agonist selection and concentration:
Receptor model systems:
GRK5 phosphorylates various GPCRs including adrenergic receptors, muscarinic acetylcholine receptors (particularly Gi-coupled M2/M4 subtypes), dopamine receptors, and opioid receptors
Choose appropriate cell models expressing the receptor of interest
Consider using overexpression systems with tagged receptors for easier detection
Readout methodology:
Direct phosphorylation detection via Western blot with phospho-specific antibodies
Functional assays measuring desensitization (reduced cAMP/Ca2+ responses upon repeated stimulation)
Receptor trafficking assays (internalization following phosphorylation)
Arrestin recruitment assays (BRET/FRET-based) following GRK5-mediated phosphorylation
When interpreting results, remember that GRK5 is just one of several kinases that can phosphorylate GPCRs. Careful experimental design with appropriate controls is essential to attribute observed phosphorylation specifically to GRK5 activity.
Detecting GRK5 in neuronal tissues presents unique challenges due to its relatively lower abundance in brain compared to other tissues and its dynamic subcellular localization. Consider these optimization strategies:
Tissue preparation and fixation:
Test different fixation protocols (4% PFA, methanol, or combinations)
Optimize fixation duration (typically 10-20 minutes for PFA)
For brain tissues, perfusion fixation usually provides better preservation than immersion fixation
Antigen retrieval optimization:
Signal amplification strategies:
Utilize fluorophore-conjugated secondary antibodies with bright, photostable dyes
Consider tyramide signal amplification for low-abundance targets
Try biotin-streptavidin amplification systems
Antibody selection and protocol:
Reduction of autofluorescence:
Include Sudan Black B treatment to reduce lipofuscin autofluorescence
Consider using TrueBlack® or similar reagents to quench background
Use confocal microscopy with narrow bandpass filters
Controls and validation:
For subcellular localization studies, high-resolution confocal or super-resolution microscopy may be necessary to accurately distinguish between membrane, cytoplasmic, and nuclear GRK5 populations.
Accurate quantification of GRK5 expression changes in disease models requires robust methodological approaches:
Western blot quantification:
Use internal loading controls appropriate for your sample type (GAPDH, β-actin, or tubulin)
Employ LI-COR Odyssey® or similar fluorescent detection systems for more accurate linear quantification
Run standard curves with recombinant GRK5 to establish quantification limits
Normalize to total protein loading using stain-free gels or Ponceau S staining
ELISA-based approaches:
Quantitative immunohistochemistry:
Use automated image analysis software to quantify DAB staining intensity
Include standardized positive controls in each batch
Apply tissue microarray approaches for high-throughput analysis
Use stereological methods for unbiased quantification
mRNA quantification as complementary approach:
Perform RT-qPCR for GRK5 transcript levels in parallel with protein studies
Use droplet digital PCR for absolute quantification
Consider RNA-seq for broader pathway analysis
Mass spectrometry-based quantification:
Use targeted proteomics approaches (MRM/PRM) for absolute quantification
Apply AQUA peptide standards specific to GRK5
Combine with phospho-enrichment to simultaneously measure GRK5 expression and activity
When studying disease models, particularly important considerations include:
GRK5 deficiency in Alzheimer's disease: Total GRK5 levels decrease in AD autopsy samples and animal models
Subcellular redistribution: Changes in localization may occur without changes in total expression
Temporal dynamics: Monitor GRK5 levels across disease progression, particularly in early/prodromal stages
Regional variations: Focus on regions particularly relevant to your disease model (e.g., limbic system in AD)
Each quantification method has strengths and limitations, so using multiple complementary approaches provides the most robust assessment of GRK5 expression changes.
Emerging microscopy technologies offer powerful new approaches for studying GRK5 dynamics:
Super-resolution microscopy:
STED (Stimulated Emission Depletion) microscopy can resolve GRK5 membrane association with significantly improved resolution over confocal microscopy
STORM/PALM techniques allow single-molecule localization of GRK5, potentially revealing clustering or organization patterns at the membrane
SIM (Structured Illumination Microscopy) provides enhanced resolution while maintaining live-cell compatibility
Live-cell imaging approaches:
CRISPR-Cas9 knock-in of fluorescent tags at the endogenous GRK5 locus for physiological expression levels
Photoactivatable or photoconvertible fluorescent protein fusions to track specific populations of GRK5 molecules
FRAP (Fluorescence Recovery After Photobleaching) to measure GRK5 mobility and membrane association kinetics
Proximity detection technologies:
FRET sensors to detect GRK5 interactions with receptors or downstream effectors
BioID or APEX2 proximity labeling to identify the GRK5 interactome in specific subcellular compartments
Split-fluorescent protein complementation to visualize GRK5-substrate interactions
Correlative light-electron microscopy (CLEM):
Combine fluorescence imaging of GRK5 with electron microscopy ultrastructure
Particularly valuable for studying membrane microdomains where GRK5 may be enriched
Lattice light-sheet microscopy:
Enables long-term 3D imaging with minimal phototoxicity
Ideal for tracking GRK5 translocation events in response to stimuli over extended periods
Expansion microscopy:
Physical enlargement of specimens allowing super-resolution imaging on conventional microscopes
Particularly useful for crowded cellular regions where GRK5 and interacting proteins may be difficult to resolve
These advanced techniques could reveal key insights about GRK5 biology, including:
The dynamics of GRK5 membrane-cytosol shuttling in response to Ca2+/calmodulin binding
The specific membrane microdomains where GRK5 concentrates during GPCR activation
The kinetics of GRK5 nuclear translocation in response to specific stimuli
The impact of disease-associated mutations or polymorphisms (like GRK5-Gln41Leu) on GRK5 localization dynamics
GRK5 antibodies are increasingly valuable tools in translational and clinical research contexts:
Biomarker development:
GRK5 deficiency has been associated with mild cognitive impairment (MCI) and Alzheimer's disease
Antibody-based assays could potentially detect changes in GRK5 levels or localization in accessible biospecimens
Quantitative assessment of GRK5 in conjunction with other biomarkers might improve early detection of neurodegenerative conditions
Therapeutic target validation:
GRK5 modulation represents a potential therapeutic strategy for conditions including cardiovascular disease and neurodegeneration
Antibodies enable precise localization and quantification of GRK5 in preclinical models and patient samples
Validation of target engagement for GRK5-directed therapeutics
Patient stratification approaches:
Different GRK5 expression levels or polymorphisms may predict treatment response
Immunohistochemical analysis of GRK5 in patient-derived samples could guide personalized treatment strategies
The GRK5-Gln41Leu polymorphism associates with lower risk of late-onset AD and could be used for risk stratification
Drug discovery applications:
High-content screening using GRK5 antibodies to identify compounds that modulate GRK5 localization or activity
Validation of mechanism of action for compounds targeting GRK5 or GRK5-regulated pathways
Assessment of on-target vs. off-target effects in drug development pipelines
Mechanism-based diagnostics:
Combined analysis of GRK5 with its substrates may provide mechanistic insights into disease processes
Multi-parameter imaging approaches to simultaneously assess GRK5 and interacting partners