SNRK antibodies target the SNRK protein, a 765-amino acid nuclear kinase belonging to the AMP-activated protein kinase (AMPK) family . These antibodies enable precise detection in techniques such as:
Western blot (WB)
Immunoprecipitation (IP)
Immunofluorescence (IF)
Key commercial variants include:
Antibody Name | Host Species | Clonality | Reactivity | Applications |
---|---|---|---|---|
SNRK Antibody (A-3) | Mouse | Monoclonal | Human, Mouse, Rat | WB, IP, IF, ELISA |
SNRK Antibody (NBP1-83668) | Rabbit | Polyclonal | Human, Mouse, Rat | IHC, ICC/IF, WB |
SNRK is cardioprotective by suppressing inflammation via:
AKT Signaling Modulation: SNRK knockdown in cardiomyocytes (CMs) increases proinflammatory proteins (p-p65, TNF-α, IL-6) and reduces anti-inflammatory IL-10 . AKT inhibition reverses these effects, suggesting SNRK-AKT axis regulation .
NF-κB Pathway Suppression: SNRK interacts with phosphorylated p65 (NF-κB subunit), preventing nuclear translocation and proinflammatory gene activation. Angiotensin II (Ang II) exacerbates inflammation in SNRK-deficient CMs .
In Snrk ecKO mice, Ang II infusion increased cardiac TNF-α (1.8-fold) and IL-6 (2.1-fold) versus controls .
HL-1 cardiomyocytes with Snrk siRNA showed elevated NF-κB activity (GFP/RFP ratio: 2.4 vs. 1.1 in controls) .
Anti-SNRK antibodies predict therapeutic response to tofacitinib:
Diagnostic Performance:
Clinical Correlation: Higher baseline anti-SNRK levels correlate with caspase-1 (ρ=0.42) and IL-18 (ρ=0.38) . Post-treatment, levels decline significantly in responders (Δ= −43%, p<0.01) .
Orthogonal Validation: Novus’s NBP1-83668 confirms SNRK expression in human brain endothelial cells (nuclear localization) and rodent cell lines (NIH-3T3, NBT-II) .
Dilution Guidelines:
SNRK antibodies facilitate research into:
SNRK (SNF-related kinase) is a member of the CAMK Ser/Thr protein kinase family with a molecular weight of approximately 84.3 kDa and 765 amino acid residues in its canonical human form. The protein predominantly localizes to the nucleus and exists in up to two different isoforms. SNRK is expressed in hematopoietic progenitor cells and leukemic cell lines, suggesting a potential role in hematopoietic cell proliferation or differentiation. Recent research has revealed SNRK's significant function in cardiac homeostasis, particularly in regulating TGFβ signaling and preventing cardiac fibrosis . The protein undergoes post-translational modifications, notably phosphorylation, which may regulate its activity and function in different cellular contexts. Due to its involvement in these critical physiological processes, SNRK has become an important research target, making SNRK-specific antibodies valuable tools for investigating its expression, localization, and function.
When investigating the different SNRK isoforms, researchers typically employ antibodies targeting specific regions of the protein that differ between isoforms. Western blot analysis often reveals two distinct bands when probing for SNRK - a lower band at approximately 74 kDa that matches the expected SNRK size and an upper band that shows variation across different tissue samples . To distinguish between these isoforms, researchers should select antibodies that recognize epitopes unique to each variant or use a combination of antibodies targeting different protein regions. When analyzing human samples, it's important to note that SNRK expression patterns vary between atrial and ventricular tissues, with atrial tissue typically showing predominantly the lower band while ventricular tissue displays both bands . For optimal isoform distinction, western blotting conditions should be carefully optimized to resolve these closely migrating protein bands.
When conducting literature searches or designing experiments targeting SNRK, researchers should be aware of several synonyms used to describe this protein. SNRK may be referenced as SNF-related serine/threonine-protein kinase, SNF-1 related kinase, SNF1-related kinase, or HSNFRK in various research publications and databases . For comparative or evolutionary studies, it's valuable to note that SNRK has documented orthologs across multiple species, including mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken . These orthologs may exhibit varying degrees of sequence conservation and potentially distinct functional properties. When selecting antibodies for cross-species experiments, researchers should verify the conservation of the target epitope in the species of interest and confirm antibody cross-reactivity through manufacturer specifications or validation experiments. Understanding these nomenclature variations and evolutionary relationships facilitates comprehensive literature reviews and appropriate experimental design when studying SNRK.
In cardiovascular research, SNRK antibodies have proven particularly valuable for investigating chamber-specific expression patterns and the protein's role in cardiac fibrosis prevention. Western blot analysis represents the most widely used application, allowing researchers to quantify SNRK expression differences between atrial and ventricular tissues, as well as between healthy and pathological samples . Immunofluorescence studies using anti-SNRK rabbit polyclonal antibodies (typically at 1:100 dilution) enable visualization of SNRK's distribution in cardiac tissues, revealing its higher expression in atrial cardiomyocytes compared to ventricular cells . For co-localization studies, researchers can pair SNRK antibodies with cardiac troponin T antibodies to specifically identify cardiomyocyte populations . ELISA can be employed to quantify SNRK expression levels in tissue or cell lysates with high sensitivity. When investigating SNRK's relationship with TGFβ1 signaling, researchers should design experiments that simultaneously assess both proteins, as SNRK expression inversely correlates with TGFβ1 levels in cardiac tissues . These applications collectively provide complementary approaches to elucidate SNRK's function in cardiovascular physiology and pathology.
When designing siRNA experiments to investigate SNRK function in cardiac cells, researchers should first select an appropriate cardiac cell model that expresses SNRK, such as HL-1 adult immortalized mouse atrial cells which have proven effective in previous studies . The experimental design should include:
siRNA design targeting conserved regions of SNRK mRNA, avoiding sequences with homology to other kinases or off-target effects
Appropriate transfection controls (non-targeting siRNA) and SNRK expression verification through both mRNA (RT-qPCR) and protein (western blot) analyses
Optimized transfection conditions specific to cardiac cell types, as these can be challenging to transfect
Time-course analysis to determine the optimal time window for observing phenotypic effects after SNRK knockdown
For investigating SNRK's role in TGFβ1 regulation, researchers should collect both cell lysates and supernatants, as previous studies have shown that SNRK knockdown in HL-1 cells results in increased TGFβ1 secretion (approximately 48 pg/mL compared to 7 pg/mL in control conditions) . ELISA assays for TGFβ1 quantification in supernatants and western blots for analyzing downstream Smad 2/3 signaling represent essential readouts. To study paracrine effects on cardiac fibroblasts, co-culture systems with SNRK-knockdown cardiomyocytes and cardiac fibroblasts, monitoring α-SMA as a fibroblast activation marker, provide valuable insights into SNRK's role in intercellular communication .
When performing immunofluorescence studies with SNRK antibodies in cardiac tissue, several technical considerations are crucial for obtaining reliable and interpretable results. Tissue fixation and processing significantly impact antibody accessibility to epitopes, with 4% paraformaldehyde fixation and 7 μm-thick paraffin-embedded sections proving effective in previous studies . Antigen retrieval methods should be optimized, as SNRK epitopes may be masked during fixation. Most studies successfully employ anti-SNRK rabbit polyclonal antibodies (such as those from Genetex) at a 1:100 dilution in 1X PBS, incubated overnight at 4°C .
SNRK exhibits marked chamber-specific expression patterns in the heart, with significantly higher levels in atrial cardiomyocytes compared to ventricular cardiomyocytes across multiple species, including mice, rats, and humans . This differential expression appears functionally significant, as it inversely correlates with TGFβ1 levels and fibrotic tendencies. In human samples without heart failure, SNRK expression is notably higher in atrial tissue compared to ventricular tissue, though this pattern is disrupted in heart failure patients where SNRK expression is lost in approximately 83% (5 out of 6) of atrial samples .
The chamber-specific expression of SNRK has critical implications for cardiac fibrosis research. Conditional knockout of SNRK in mouse cardiomyocytes (Snrk cmcKO) leads to more pronounced fibrosis in atrial chambers compared to ventricular chambers under basal conditions . This chamber-selective fibrotic response mirrors observations from earlier studies with constitutively active TGFβ1 expression, which similarly produced selective atrial fibrosis despite expression in both chambers . For researchers investigating cardiac fibrosis mechanisms, these findings suggest that: (1) atrial and ventricular fibrotic processes may involve distinct regulatory mechanisms, (2) SNRK represents a critical chamber-specific regulator of fibrosis, and (3) therapeutic approaches targeting fibrosis may need to consider chamber-specific molecular contexts. When designing studies, researchers should analyze atrial and ventricular tissues separately rather than using whole-heart homogenates that might mask these important differences.
SNRK functions as a critical negative regulator of TGFβ1 expression and signaling in cardiac tissue, establishing an important mechanistic link to cardiac fibrosis development. In cardiomyocyte-specific SNRK knockout mice (Snrk cmcKO), TGFβ1 protein levels are significantly elevated in heart tissue under basal conditions and increase further with Angiotensin II stimulation compared to control mice . At the cellular level, siRNA-mediated knockdown of SNRK in HL-1 adult mouse atrial cells results in dramatically increased TGFβ1 secretion (approximately 7-fold increase from 7 pg/mL to 48 pg/mL) .
Mechanistically, SNRK appears to repress TGFβ1 signaling through inhibition of Smad 2/3 pathways in atrial cardiomyocytes . This inhibition prevents paracrine activation of cardiac fibroblasts, as evidenced by reduced α-SMA expression (a marker of fibroblast activation) in co-culture experiments . The relationship between SNRK and TGFβ1 has translational relevance, as human heart failure samples show an inverse correlation between SNRK expression and TGFβ1 levels .
For researchers investigating this pathway, experimental designs should include:
Analysis of both intracellular and secreted TGFβ1 (via cell/tissue lysates and supernatants/serum)
Assessment of downstream Smad 2/3 phosphorylation status
Evaluation of paracrine effects on fibroblast activation markers
Consideration of intervention points in the SNRK-TGFβ1 axis for potential therapeutic development
This SNRK-TGFβ1 relationship explains the chamber-specific fibrotic phenotypes observed in various models and offers a mechanistic framework for understanding atrial fibrosis in heart failure and atrial fibrillation.
Analyzing SNRK expression changes in heart failure models requires careful consideration of model selection, tissue sampling, and analytical techniques. Researchers should consider both genetic models (such as cardiomyocyte-specific Snrk knockout mice) and induced models (such as Angiotensin II infusion) to comprehensively understand SNRK's role in heart failure progression . When collecting tissue samples, chamber-specific analysis is essential, as SNRK expression and its effects differ significantly between atrial and ventricular tissues .
For protein-level analysis, western blotting represents the primary technique, though researchers should be aware that SNRK antibodies may detect multiple bands in cardiac tissue (particularly in ventricular samples) that require careful quantification . The characteristic lower band at approximately 74 kDa corresponds to the expected SNRK size, while upper bands may represent isoforms or post-translationally modified variants . Normalization to appropriate loading controls (such as GAPDH) is critical for accurate quantification.
Immunohistochemistry and immunofluorescence provide valuable spatial information about SNRK expression changes, particularly when combined with cardiomyocyte markers like cardiac troponin T for cell-type-specific analysis . For human samples, researchers should carefully classify specimens based on heart failure status, using established markers like CD68 (macrophage infiltration) and collagen staining (fibrosis) to verify disease state .
When analyzing results, researchers should consider SNRK's relationship with TGFβ1 expression, as these proteins show inverse correlation in heart failure progression . Longitudinal sampling in animal models can provide insights into the temporal dynamics of SNRK expression changes during heart failure development. For translational relevance, findings from animal models should be validated in human heart failure samples whenever possible.
Co-immunoprecipitation (co-IP) using SNRK antibodies represents a powerful approach for identifying novel protein interactions that might illuminate SNRK's regulatory mechanisms. When designing co-IP experiments, researchers should first select SNRK antibodies that effectively capture the native protein conformation, preferably validated for immunoprecipitation applications. The experimental workflow should begin with careful cell or tissue lysis under non-denaturing conditions to preserve protein-protein interactions, using buffers containing mild detergents like NP-40 or Triton X-100.
Cross-linking approaches using membrane-permeable reagents like DSP (dithiobis[succinimidyl propionate]) prior to lysis can stabilize transient or weak interactions. For cardiac tissue specifically, chamber-specific analysis is advisable given SNRK's differential expression and potential function between atrial and ventricular tissues . After immunoprecipitation with SNRK antibodies, researchers can identify interacting partners through techniques including:
Targeted western blotting for suspected interactors (particularly components of the TGFβ signaling pathway given SNRK's established role)
Mass spectrometry-based proteomics for unbiased discovery of the complete SNRK interactome
Functional validation through reciprocal co-IPs and proximity ligation assays
Since SNRK undergoes phosphorylation as a post-translational modification , phosphorylation-state specific antibodies may reveal condition-dependent interactions. Researchers should also consider comparing the interactome between different cardiac chambers and between healthy and pathological states to identify context-specific SNRK binding partners that might explain its chamber-specific effects on cardiac fibrosis .
When encountering contradictory findings regarding SNRK function in different cardiac pathologies, researchers should implement systematic approaches to reconcile these discrepancies. First, a comprehensive analysis of experimental models is essential, as differences between genetic knockout approaches (global versus tissue-specific), inducible versus constitutive systems, and acute versus chronic disease models can yield apparently contradictory results. The chamber-specific expression and function of SNRK necessitates careful consideration of whether studies focused on atrial pathology, ventricular pathology, or whole-heart analysis, as findings from one chamber may not generalize to the other.
Methodological variations in SNRK detection and quantification can contribute to discrepancies. Researchers should note whether studies used antibodies targeting different epitopes, which might detect distinct SNRK isoforms or post-translationally modified variants . The timing of analysis relative to disease progression is particularly important, as SNRK's role may evolve throughout pathogenesis—from initial compensatory responses to later maladaptive changes.
To systematically address contradictions, researchers should:
Design experiments that simultaneously assess multiple cardiac pathologies using consistent methodology
Perform time-course analyses to capture the dynamic nature of SNRK expression and function
Employ multiple, complementary techniques to measure SNRK levels and activity
Consider genetic background effects that might influence SNRK function
Evaluate SNRK's context-dependent interactions with the TGFβ signaling pathway
Through these approaches, apparent contradictions may resolve into a more nuanced understanding of SNRK's multifaceted and context-dependent roles in cardiac physiology and pathology.
The development of therapeutic strategies targeting the SNRK pathway for cardiac fibrosis represents an emerging research direction with translational potential. Anti-SNRK antibodies can contribute to this effort in several ways beyond their conventional research applications. Given SNRK's established role as a repressor of TGFβ1 expression and cardiac fibrosis , therapeutic approaches would likely aim to enhance or preserve SNRK function rather than inhibit it.
Researchers can use anti-SNRK antibodies to develop screening platforms for identifying compounds that stabilize or increase SNRK expression, particularly in atrial tissue where its levels are naturally higher and functionally significant . Such screens might employ high-content imaging with fluorescently-labeled anti-SNRK antibodies to quantify protein levels in response to compound libraries. Alternatively, ELISA-based screening approaches using anti-SNRK antibodies could provide quantitative readouts of protein levels in response to potential therapeutic candidates.
For more advanced therapeutic development, researchers might explore antibody-directed drug delivery, where anti-SNRK antibodies could guide payloads specifically to SNRK-expressing cardiomyocytes. This approach would require extensive modification and validation of antibodies to ensure they retain binding specificity while gaining cell-penetrating properties. Therapeutic strategies might also target the SNRK-TGFβ1 regulatory axis at various points:
Compounds that enhance SNRK expression or stability in cardiomyocytes
Molecules that promote SNRK's inhibitory effect on TGFβ1 signaling
Interventions that block the downstream effects of reduced SNRK expression
Given the chamber-specific expression patterns of SNRK , therapeutic approaches might need to be tailored to specifically address atrial fibrosis in conditions like atrial fibrillation, where SNRK's regulatory role appears particularly important.
Selecting the optimal SNRK antibody requires systematic evaluation of several criteria matched to the specific experimental application. For western blot applications, researchers should prioritize antibodies validated to detect the expected 84.3 kDa SNRK protein and any relevant isoforms. When working with cardiac tissue specifically, antibodies should be verified to detect both the lower band (~74 kDa) and upper band patterns observed in previous studies . For immunohistochemistry or immunofluorescence, researchers should select antibodies specifically validated for these applications, preferably with demonstrated performance in paraformaldehyde-fixed, paraffin-embedded tissues at defined dilutions (typically 1:100) .
The epitope location represents a critical consideration, as antibodies targeting different regions of SNRK may yield varying results. Those targeting the middle region may detect multiple isoforms, while those targeting terminal regions might be more isoform-specific . Species reactivity must align with the experimental model, with most commercially available antibodies showing reactivity to human and rat SNRK, though cross-reactivity with mouse and other species varies .
Polyclonal antibodies offer broader epitope recognition but potentially greater batch-to-batch variability, while monoclonal antibodies provide consistency at the potential cost of more limited epitope recognition. For quantitative applications, researchers should verify the antibody's linear detection range through dilution series experiments. When studying SNRK's relationship with TGFβ1 signaling, selecting antibodies compatible with multiplexed detection of both proteins is advantageous . Finally, researchers should verify that selected antibodies have been used successfully in peer-reviewed publications, particularly in similar experimental contexts to their planned applications.
Rigorous validation of SNRK antibodies is essential for ensuring experimental reliability and reproducibility, particularly when studying this protein in cardiovascular disease contexts. Primary validation should include positive and negative controls: positive controls might utilize recombinant SNRK protein or lysates from tissues known to express high SNRK levels (such as atrial tissue) , while negative controls could include lysates from SNRK knockout models or cells treated with SNRK-targeting siRNA .
For western blot applications, researchers should verify antibody specificity through:
Detection of bands at the expected molecular weight (~84.3 kDa for canonical SNRK, with potential additional bands at higher molecular weights in ventricular tissue)
Disappearance or reduction of these bands in SNRK-depleted samples
Pre-absorption controls using recombinant SNRK protein to confirm binding specificity
For immunohistochemistry or immunofluorescence applications, validation should include:
Absence of signal in SNRK knockout tissues or with isotype control antibodies
Co-localization with known SNRK interacting partners
Subcellular localization consistent with SNRK's reported nuclear distribution
Comparison of staining patterns using multiple antibodies targeting different SNRK epitopes
When employing SNRK antibodies for novel applications or in understudied tissues, additional validation through complementary techniques is advisable. For instance, correlating protein detection via antibodies with mRNA expression via RT-qPCR provides orthogonal validation. Finally, researchers should maintain detailed records of antibody lot numbers, dilutions, and experimental conditions to facilitate reproducibility and troubleshooting across experiments.
When working with SNRK antibodies in complex tissue samples like cardiac tissue, researchers may encounter several technical challenges requiring systematic troubleshooting approaches. For weak or absent signal in western blots, researchers should first optimize protein extraction methods, considering that SNRK's nuclear localization may require specialized nuclear extraction protocols. Loading higher protein amounts (50-100 μg) may help detect low abundance SNRK, particularly in ventricular samples where expression is lower than in atrial tissue . Extended exposure times and more sensitive detection systems (such as chemiluminescent substrates with longer emission durations) can enhance detection of weak signals.
For multiple band patterns, particularly in ventricular samples where both lower (~74 kDa) and higher molecular weight bands appear , researchers should consider whether these represent isoforms, post-translational modifications, or non-specific binding. Comparing patterns across different antibodies targeting distinct SNRK epitopes can help distinguish genuine SNRK-related bands from non-specific signals. Phosphatase treatment of samples prior to electrophoresis can determine whether higher molecular weight bands represent phosphorylated SNRK forms.
In immunohistochemistry applications, background staining can obscure specific SNRK signal. Optimization approaches include:
Extended blocking steps (2-3 hours) with 5-10% serum matching the secondary antibody species
Titration of primary antibody concentration (testing dilutions from 1:50 to 1:500)
Implementation of antigen retrieval methods optimized for nuclear antigens
Use of more specific detection systems like polymer-HRP conjugates rather than biotinylated secondaries
For chamber-specific SNRK analysis in cardiac tissue, careful tissue dissection and processing are essential to prevent cross-contamination between atrial and ventricular samples that could obscure the established expression differences . Finally, researchers should consider that SNRK expression varies with disease states, particularly in heart failure where atrial SNRK expression may be significantly reduced or lost , potentially requiring adjusted detection approaches for pathological samples.