TNF a Canine

Tumor Necrosis Factor-Alpha Canine Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of Canine TNF-α

Canine TNF-α is a 233-amino acid protein organized into three domains:

  • 35-amino acid cytoplasmic domain

  • 21-amino acid transmembrane segment

  • 177-amino acid extracellular domain (ECD)

The functional form is a noncovalent homotrimer with high structural conservation:

FeatureCanine TNF-αHuman TNF-α
Quaternary StructureTrimerTrimer
ECD Sequence Identity84%-94% (vs human)Reference
Crystal Resolution1.85 Å 2.1-2.8 Å (typical)

The ECD contains conserved receptor-binding regions critical for interaction with TNF receptors I/II .

Biological Functions and Pathological Roles

As a pleiotropic cytokine, canine TNF-α mediates:

  • Acute-phase inflammatory responses

  • Apoptotic signaling pathways

  • Immune cell differentiation

Clinical correlations demonstrate:

  • 10-14.7 pg/mL serum concentrations in babesiosis-associated renal failure

  • 0.05 nM IC₅₀ for TNF neutralization by Humivet-R1 antibody

  • 14% detection rate in osteoarthritic synovial fluid vs 0% in healthy joints

Antibody Therapeutics:

  • Humivet-R1: Chimeric antibody demonstrating:

    • 0.416 nM EC₅₀ binding affinity

    • 95% structural epitope overlap with human TNF-α

Clinical Trial Outcomes:

TreatmentStudy TypeOutcomeSource
caTNFR-Fc (IA injection)OA pilot studyNo gait improvement (p=0.62)
Anti-TNF mAbStructural studyNeutralization capacity validated

Key Research Findings

  1. Canine-Human Epitope Conservation:

    • 85% structural similarity enables cross-reactive antibody design

    • Adalimumab grafting maintains <0.5 nM potency in chimeric formats

  2. Disease Associations:

    • Babesiosis: Serum TNF-α correlates with:

      • Hypotension (r=-0.72 systolic BP)

      • Renal failure markers (FEₙₐ +0.71)

    • Osteoarthritis:

      • SF TNF-α: 0.62 pg/mL (OA) vs undetectable (healthy)

  3. Pharmacokinetic Challenges:

    • IA-administered caTNFR-Fc showed no systemic absorption

    • Plasma detection limit: <0.1 ng/mL

Product Specs

Introduction
Tumor necrosis factor (TNF), a cytokine crucial for systemic inflammation, belongs to a family of cytokines that trigger the acute phase reaction. Primarily produced by macrophages, TNF exerts pleiotropic effects, including apoptosis, cell proliferation, differentiation, inflammation, tumorigenesis, and viral replication. Additionally, it participates in lipid metabolism and coagulation. The regulation of immune cells stands as TNF's central role. Dysregulation and excessive production of TNF are implicated in various human diseases, encompassing autoimmune disorders, insulin resistance, and cancer.
Description
Recombinant Canine TNF-a, produced in E.Coli, is a single, non-glycosylated polypeptide chain comprising 157 amino acids. With a molecular mass of 17.3 kDa, it is purified using proprietary chromatographic techniques.
Physical Appearance
The product appears as a sterile, filtered white powder, freeze-dried for preservation.
Formulation
The product is filtered through a 0.2µm filter and subsequently lyophilized from a concentrated solution (1mg/ml) in 1x PBS at pH 7.4.
Solubility
For reconstitution, it is advised to dissolve the lyophilized Tumor Necrosis Factor-alpha in sterile 18M-cm H2O at a minimum concentration of 100µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
While lyophilized Tumor Necrosis Factor-a remains stable at room temperature for up to 3 weeks, storage in desiccated conditions below -18°C is recommended. After reconstitution, TNF-a should be kept at 4°C for a period of 2-7 days. For prolonged storage, freezing below -18°C is advised. To enhance stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Repeated freeze-thaw cycles should be avoided.
Purity
The purity exceeds 95.0% as determined by: (a) Reverse-Phase High Performance Liquid Chromatography (RP-HPLC) analysis. (b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Biological Activity
Exhibiting a specific activity greater than 3.3 x 10⁵ IU/mg, the biological activity is determined by assessing the cytolysis of murine L929 cells in the presence of Actinomycin D.
Synonyms
Tumor necrosis factor, Cachectin, TNF-alpha, Tumor necrosis factor ligand superfamily member 2, TNF-a, TNF, TNFA, TNFSF2.
Source
Escherichia Coli.
Amino Acid Sequence
VKSSSRTPSD KPVAHVVANP EAEGQLQWLS RRANALLANG VELTDNQLIV PSDGLYLIYS QVLFKGQGCP STHVLLTHTI SRFAVSYQTK VNLLSAIKSP CQRETPEGTE AKPWYEPIYL GGVFQLEKGD RLSAEINLPN YLDFAESGQV YFGIIAL.

Q&A

What is the biological role of TNF-alpha in canines and how does it compare to human TNF-alpha?

TNF-alpha in canines functions as a multifunctional proinflammatory cytokine belonging to the tumor necrosis factor superfamily, primarily secreted by macrophages. It binds to receptors TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR, playing crucial roles in immune cell regulation, cell proliferation, differentiation, apoptosis, lipid metabolism, and coagulation . Structurally, canine TNF-alpha shares 91% homology with human TNF-alpha, making it a valuable comparative model, though this homology does not guarantee cross-reactivity in all detection assays . Research has demonstrated neuroprotective functions of TNF-alpha in canines, similar to those observed in knockout studies in mice, suggesting conserved functional mechanisms across species . Despite structural similarities, species-specific differences in receptor binding affinity and downstream signaling pathways necessitate caution when extrapolating findings between human and canine systems.

What are the established reference ranges for TNF-alpha in healthy canines?

Currently, no universally established reference ranges exist for TNF-alpha concentrations in healthy canines, presenting a significant challenge for researchers interpreting experimental results . Published values show considerable variation across studies using different detection methodologies, breeds, and sample types. In healthy Beagles, plasma TNF-alpha has been reported at approximately 3.00 pg/mL (below detection limit to 3.00 pg/mL) . In various canine breeds, serum TNF-alpha measurements range from 11.9 pg/mL (9.44-27.0 pg/mL) to values frequently below detection limits . In mixed-breed racing sled dogs, plasma TNF-alpha averaged 0.81 pg/mL (0.49-1.35 pg/mL) . These variations highlight the importance of including appropriate breed-matched, age-matched, and sex-matched controls in experimental designs, as these factors significantly influence baseline cytokine measurements . Researchers should consider establishing internal reference ranges specific to their laboratory methods, sample processing protocols, and target populations rather than relying on published values from heterogeneous sources.

How do breed, age, and sex influence TNF-alpha expression in canines?

Research demonstrates that breed, age, and sex significantly impact baseline TNF-alpha expression in canines, necessitating careful experimental design . Multiple studies have documented statistically significant differences in cytokine measurements based on these factors, with breed-specific variations being particularly prominent . For example, data compiled from various studies show distinct TNF-alpha profiles among Beagles, Labrador Retrievers, and mixed-breed dogs . Age-related differences are also apparent, with developmental stage influencing inflammatory cytokine expression patterns, while sex-based variations may reflect hormonal influences on immune function . Even brain lateralization has been associated with differences in cytokine measurements in dogs, suggesting complex neuroimmunoendocrine interactions . These findings emphasize the critical importance of restricting study populations by breed, age, and sex when measuring TNF-alpha, or alternatively, ensuring experimental and control groups are precisely matched for these variables to minimize confounding factors. Researchers investigating TNF-alpha should report detailed demographic information about their canine subjects and consider these variables during data interpretation.

What are the optimal methods for detecting and quantifying canine TNF-alpha in different sample types?

The optimal detection methods for canine TNF-alpha vary based on sample type, required sensitivity, and available resources. Enzyme-linked immunosorbent assays (ELISAs) designed specifically for canine TNF-alpha remain the gold standard for quantification in serum, plasma, and cell culture supernatants . Canine-specific sandwich ELISAs utilizing matched antibody pairs demonstrate superior specificity compared to cross-reactive human TNF-alpha assays . The Quantikine® standard ELISA has been validated for canine samples, while the Immulite® automated ELISA, despite its convenience, failed to detect canine TNF-alpha despite the 91% homology between human and canine forms . For comprehensive cytokine profiling, multiplex assays allow simultaneous measurement of TNF-alpha alongside other inflammatory mediators, though sensitivity may vary . Sample preparation significantly impacts results—plasma and serum values differ systematically, with some studies reporting higher cytokine recovery from serum . Researchers should validate their chosen assay with appropriate positive controls, determine the limit of detection for their specific sample types, and maintain consistent sample collection, processing, and storage protocols throughout a study. Pre-analytical variables including time from collection to processing, storage temperature, and freeze-thaw cycles should be standardized and reported in publications.

How can researchers address the high variability and frequent below-detection results in canine TNF-alpha measurements?

Addressing the high variability and frequent below-detection results in canine TNF-alpha measurements requires a multifaceted approach to experimental design and data analysis. First, researchers should select assays with appropriate sensitivity for canine samples, preferably using canine-specific reagents rather than those designed for human samples, as demonstrated by the failure of Immulite® automated ELISA to detect canine TNF-alpha despite high sequence homology . Second, implementing rigorous pre-analytical standardization is crucial—controlling for collection time, immediate processing, consistent storage conditions, and minimizing freeze-thaw cycles . Third, researchers should consider sample enrichment techniques for low-abundance samples, such as optimizing protein extraction methods or employing signal amplification strategies. For statistical analysis of below-detection samples, specialized approaches such as maximum likelihood estimation or multiple imputation methods are preferable to simple substitution with zero or half the detection limit . Including positive control samples (e.g., LPS-stimulated whole blood cultures) can validate assay functionality when test samples yield below-detection results. Finally, researchers should report detailed methodological parameters including the assay's lower limit of detection, the percentage of samples below this threshold, and the specific statistical handling of these values to improve reproducibility across studies.

What statistical approaches are recommended when analyzing TNF-alpha data with significant inter-individual variation?

When analyzing canine TNF-alpha data with significant inter-individual variation, several statistical approaches can improve validity and interpretability of results. Non-parametric methods (e.g., Wilcoxon matched pairs test, Mann-Whitney U test) are often more appropriate than parametric tests given the typically non-normal distribution and heteroscedasticity of cytokine data . For longitudinal measurements, such as monitoring TNF-alpha during disease progression or treatment, mixed-effects modeling can account for both within-subject and between-subject variability while handling missing data points . Log transformation of cytokine concentrations frequently normalizes distributions and stabilizes variance, making parametric testing more valid after transformation . When examining relationships between TNF-alpha and categorical variables (such as disease stages or antigen levels), Fisher's exact test may be more appropriate than chi-squared tests for small sample sizes, as demonstrated in heartworm disease research . Additionally, researchers should consider using multivariate approaches that adjust for confounding factors including breed, age, and sex, which significantly impact cytokine measurements . Power calculations should account for the high variability observed in TNF-alpha measurements, typically requiring larger sample sizes than might be expected for more stable biomarkers. Finally, comprehensive reporting of all statistical methods, including specific handling of outliers and below-detection values, is essential for reproducibility.

How does TNF-alpha expression differ across various canine diseases compared to healthy controls?

TNF-alpha expression demonstrates disease-specific patterns in canines, providing insights into pathophysiological mechanisms. In babesiosis, TNF-alpha was detectable in 54.5% (6/11) of affected dogs with an average concentration of 14.7 pg/mL, suggesting moderate inflammatory activation . Dogs with heartworm disease (Dirofilaria immitis infection) showed detectable TNF-alpha in 64.3% (9/14) of cases with a mean concentration of 7.21±12.44 pg/mL, exhibiting significant individual variation . In contrast, canine lymphoma presents a different profile—only 12% (3/25) of affected dogs had detectable TNF-alpha at diagnosis, and levels became undetectable following complete or partial remission, suggesting limited utility as a tumor marker in this malignancy despite TNF-alpha's theoretical role in lymphomagenesis . The variability in TNF-alpha expression across these conditions reflects disease-specific inflammatory mechanisms, with some conditions producing robust, consistent elevation while others show more heterogeneous responses. This differential expression underscores the importance of disease-specific controls rather than generalized "healthy" reference ranges when studying inflammatory cytokines in canine models. Additionally, temporal dynamics of TNF-alpha expression may vary by condition, necessitating careful consideration of sampling time points relative to disease onset, progression, and treatment phases.

What is the role of TNF-alpha in the pathogenesis of canine heartworm disease and how does it correlate with disease severity?

In canine heartworm disease (HWD) caused by Dirofilaria immitis, TNF-alpha's pathogenic role appears complex and incompletely understood. Research shows detectable TNF-alpha in 64.3% (9/14) of naturally infected dogs with mean concentrations of 7.21±12.44 pg/mL, but significant changes in TNF-alpha levels were not observed during alternative therapy for HWD . Importantly, no statistically significant correlation was found between TNF-alpha concentration and antigen (Ag) levels (categorized as Ag++, Ag+, Ag 0), suggesting TNF-alpha alone may not directly reflect parasite burden or disease severity . Changes in both TNF-alpha concentration and Ag level during therapy did not demonstrate a mutual relationship, indicating that TNF-alpha quantification in isolation has limited value for monitoring treatment response . These findings suggest that while TNF-alpha may contribute to the inflammatory cascade in HWD, its role might be modulated by other cytokines or dependent on TNF receptor expression patterns rather than absolute cytokine levels. Researchers have concluded that quantification of TNF-alpha alone, without concurrent analysis of its receptors, does not substantially contribute to diagnosis or treatment monitoring in canine HWD . Future investigations should consider analyzing TNF receptors (TNFRs) together with TNF-alpha to better understand the cytokine's role in disease pathogenesis and potential implementation in diagnostic, prognostic, and therapeutic approaches.

How reliable is TNF-alpha as a biomarker in canine lymphoma and other cancers?

TNF-alpha demonstrates limited reliability as a standalone biomarker in canine lymphoma despite its theoretical involvement in lymphomagenesis. Research indicates that only 12% (3/25) of dogs with lymphoma had detectable TNF-alpha at diagnosis, with levels becoming undetectable following complete or partial remission . Furthermore, TNF-alpha was not detectable in any control dogs, suggesting low sensitivity as a diagnostic marker . These findings led researchers to conclude that "serum TNF-α appears to have limited value as a tumour marker in dogs with lymphoma" . The low detection rate might reflect several biological factors: TNF-alpha may function primarily in the tumor microenvironment with minimal systemic release; it has a short half-life in circulation; or lymphoma cells may primarily produce membrane-bound rather than soluble TNF-alpha. In contrast, modified forms of TNF-alpha show greater promise in therapeutic contexts—PEGylated TNF-alpha (PEG-TNF) demonstrated biological activity and antitumor effects in dogs with spontaneous cancers, including melanoma, squamous cell carcinoma, mammary carcinoma, and angiosarcoma . These divergent findings highlight the complex, context-dependent role of TNF-alpha in canine oncology—limited as a diagnostic biomarker but potentially valuable in therapeutic applications. Researchers should consider TNF-alpha as part of a broader panel of biomarkers rather than as an independent indicator in canine cancer investigations.

What methodological considerations are important when designing experimental studies involving TNF-alpha manipulation in canines?

Designing experimental studies involving TNF-alpha manipulation in canines requires careful attention to several methodological considerations. First, breed standardization is crucial given the documented variations in cytokine profiles across breeds—researchers should either restrict studies to a single breed or ensure balanced distribution across experimental groups . Second, the choice of TNF-alpha formulation significantly impacts pharmacokinetics and toxicity profiles, as demonstrated with PEGylated TNF-alpha, which showed improved half-life (15.3 ± 4.9 hours) and reduced toxicity compared to native TNF-alpha . Third, dose determination should follow rigorous phase I protocols—the established maximum tolerated dose of PEG-TNF in dogs (26.7 μg/kg) provides a starting reference, though this may vary based on specific formulations and target conditions . Fourth, comprehensive toxicity monitoring is essential, with particular attention to vascular leak syndrome, hypotension, and coagulopathy, which were dose-limiting toxicities in canine studies . Fifth, researchers should implement multimodal assessment of biological effects, including clinical parameters (fever, leukopenia), tissue analysis (inflammation, necrosis), and advanced imaging techniques such as dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to quantify changes in tumor blood flow . Finally, the temporal dynamics of TNF-alpha administration and sampling must be carefully planned, considering both the acute effects and potential delayed responses in target tissues. These methodological considerations ensure robust, reproducible, and translatable results from experimental manipulation of TNF-alpha in canine models.

How can dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) be utilized to evaluate TNF-alpha effects on tumor vasculature in canine cancer models?

Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) represents an advanced methodology for non-invasively evaluating TNF-alpha effects on tumor vasculature in canine cancer models. In preclinical investigations of PEGylated tumor necrosis factor alpha (PEG-TNF), DCE-MRI successfully detected significant increases in tumor blood flow following administration, providing objective quantification of TNF-alpha's vascular effects . This imaging modality offers several advantages for TNF-alpha research: it enables serial assessments in the same subject over time, allowing each animal to serve as its own control; it provides spatial information about heterogeneous responses across different tumor regions; and it generates quantitative parameters that can be statistically analyzed across treatment groups . When implementing DCE-MRI protocols, researchers should standardize contrast agent administration (type, dose, injection rate), image acquisition parameters (temporal resolution, spatial coverage, sequence type), and pharmacokinetic modeling approaches to ensure reproducibility. Correlation with histopathological findings is essential—researchers should collect tumor biopsies at timepoints coordinated with imaging to validate DCE-MRI findings with direct microscopic evidence of vascular changes, inflammatory cell infiltration, and necrosis . Additionally, integration of DCE-MRI with other functional imaging techniques such as diffusion-weighted imaging or hypoxia-specific tracers can provide complementary information about tumor microenvironment changes following TNF-alpha administration, creating a more comprehensive assessment of biological effects.

What insights does canine TNF-alpha research provide for developing improved immunotherapies for both veterinary and human applications?

Canine TNF-alpha research provides valuable insights for immunotherapy development across species boundaries, functioning as a critical translational bridge between murine models and human applications. The demonstrated efficacy of PEGylated TNF-alpha (PEG-TNF) in dogs with spontaneous cancers—including melanoma, squamous cell carcinoma, mammary carcinoma, and angiosarcoma—provides proof-of-concept for TNF-alpha modification strategies that balance bioactivity with toxicity . The established maximum tolerated dose, elimination half-life (15.3 ± 4.9 hours), and dose-limiting toxicities in canines inform human trial design more accurately than murine data alone, as dogs more closely approximate human body size, physiology, and immune system complexity . Importantly, canine studies have demonstrated that biologically effective doses of PEG-TNF can be administered safely while maintaining antitumor activity, addressing a fundamental challenge in TNF-alpha therapeutics . The 91% homology between human and canine TNF-alpha enables structural insights for designing next-generation TNF-alpha variants with optimized receptor binding profiles . Additionally, the observed limitations of TNF-alpha as a biomarker in canine lymphoma suggest similar constraints might apply in human lymphoid malignancies, guiding biomarker strategy . From a veterinary perspective, these studies establish dosing, safety, and efficacy parameters specific to canine patients, addressing the critical need for species-specific evidence rather than extrapolated human protocols. Together, these bidirectional insights accelerate immunotherapy development for both species while reducing research costs through shared translational knowledge.

What are the key challenges in standardizing TNF-alpha measurements across different laboratories and how can they be addressed?

Standardizing TNF-alpha measurements across laboratories presents multiple challenges requiring systematic solutions. First, assay selection inconsistency significantly impacts results—the reported failure of Immulite® automated ELISA to detect canine TNF-alpha despite high sequence homology with human TNF-alpha (91%) highlights the critical need for canine-specific, validated assays . Second, reference standards vary between commercial kits, creating significant inter-laboratory variation—implementing common calibrators traceable to international standards would improve comparability . Third, pre-analytical variables including sample type (serum versus plasma), processing delays, storage conditions, and freeze-thaw cycles introduce systematic biases—detailed reporting of these parameters and adherence to standardized protocols would reduce this variability . Fourth, detection thresholds vary considerably between assays, with TNF-alpha frequently falling below detection limits in healthy samples—standardized approaches to statistical handling of below-detection values would improve analytical consistency . To address these challenges, researchers should: participate in inter-laboratory validation studies using shared sample sets; report comprehensive methodological details including specific reagents, detection limits, and quality control measures; develop consensus guidelines for canine cytokine measurement modeled after human clinical laboratory standards; and establish shared biorepositories of reference samples with assigned values to calibrate local assays. Implementation of these strategies would substantially improve reproducibility and cross-study comparability in canine TNF-alpha research.

How do TNF-alpha receptor expression patterns influence cytokine activity in different canine tissues and disease states?

TNF-alpha receptor expression patterns significantly influence cytokine activity across canine tissues and disease states, often providing more insight than absolute cytokine levels. TNF-alpha mediates its biological effects primarily through two receptors: TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR, with differential expression patterns determining tissue-specific responses . Research on canine heartworm disease suggests that quantification of TNF-alpha without concurrent receptor analysis provides incomplete information, as changes in receptor expression may modulate cytokine activity independent of concentration changes . This phenomenon likely explains why TNF-alpha levels alone failed to correlate with disease severity or treatment response in several studies . Tissue microenvironments further complicate this relationship—receptor density, localization (membrane-bound versus soluble), and relative ratio of TNFR1 (predominantly mediating inflammatory and apoptotic signaling) to TNFR2 (primarily mediating tissue regeneration and immune modulation) determine net biological effects . Additionally, post-receptor signaling pathways likely vary between tissues and disease states, creating context-dependent responses to equivalent receptor stimulation. Future research should implement comprehensive approaches including: quantification of both membrane-bound and soluble TNF receptors; analysis of receptor distribution through immunohistochemistry or flow cytometry; examination of downstream signaling pathway activation; and correlation of these parameters with clinical outcomes. These integrated approaches would provide more nuanced understanding of TNF-alpha biology than cytokine measurement alone.

What emerging technologies might improve detection, quantification, and functional assessment of TNF-alpha in canine research models?

Emerging technologies promise to transform TNF-alpha research in canine models through enhanced detection sensitivity, improved quantification accuracy, and advanced functional assessment capabilities. Single-molecule array (Simoa) technology offers ultrasensitive detection—potentially 100-1000 times more sensitive than conventional ELISAs—enabling reliable quantification of previously undetectable TNF-alpha levels in healthy canine samples and resolving the persistent challenge of below-detection measurements . Mass spectrometry-based approaches, including targeted proteomics (MRM-MS) and data-independent acquisition (DIA), provide absolute quantification of TNF-alpha without antibody cross-reactivity limitations, while simultaneously measuring TNF-alpha proteoforms and processing variants . For functional assessment, proximity extension assays (PEA) and proximity ligation assays (PLA) enable simultaneous detection of TNF-alpha and its binding partners within tissue contexts, providing insights into cytokine-receptor interactions that govern biological activity . Cell-based reporter systems expressing canine-specific TNF receptors offer quantitative measurement of bioactivity rather than just protein concentration, addressing the critical distinction between immunoreactive and biologically active cytokine. Advanced imaging approaches, including intravital microscopy with fluorescent TNF-alpha probes, allow real-time visualization of cytokine dynamics in living tissues . Finally, computational methods integrating multi-omic data (transcriptomics, proteomics, metabolomics) with TNF-alpha pathway modeling enable systems-level understanding of cytokine networks. These emerging technologies collectively address current limitations in canine TNF-alpha research and promise more comprehensive insights into its complex biology.

How do TNF-alpha reference ranges compare across different canine breeds and assay methodologies?

TNF-alpha reference ranges demonstrate substantial variation across canine breeds and detection methodologies, necessitating careful interpretation of research findings. The following table synthesizes published TNF-alpha measurements in healthy dogs from multiple studies using various assay platforms:

StudyBreedSample TypeSample SizeTNF-α (pg/mL)
Bastien 2015BeaglePlasman = 183.00 (bd - 3.00)
Frank 2015Labrador RetrieverSerumn = 300.90 (0.67 - 1.21)
Nielsen 2013Various breedsSerumn = 1811.9 (9.44 - 27.0)
O'Neill 2012Various breedsSerumn = 95.60 (5.20 - 5.90)
Kilpatrick 2014Various breedsPlasman = 250.38 (bd - 7.00)
von Pfeil 2015Mixed breed racing sled dogsPlasman = 200.81 (0.49 - 1.35)
Zois 2012Cavalier King Charles SpanielSerumn = 15bd

bd = below detection limit

This data reveals several important patterns: First, breed-specific variations are evident, with Labrador Retrievers showing consistently lower TNF-alpha levels than mixed-breed populations . Second, sample type influences measurements—plasma and serum values differ systematically even within the same population . Third, assay methodology significantly impacts results, with some studies reporting predominantly below-detection values while others detect measurable concentrations in all samples . These variations underscore the importance of establishing internal reference ranges specific to the experimental conditions rather than relying on literature values. Researchers should consider these factors when designing studies, interpreting results, and comparing findings across publications.

What is the comparative efficacy of different TNF-alpha detection methods in canine samples?

The comparative efficacy of TNF-alpha detection methods in canine samples varies significantly, with important implications for research methodology selection. Standard sandwich ELISAs designed specifically for canine TNF-alpha, such as the Quantikine® ELISA, have demonstrated reliable detection capability in multiple studies and serve as the current gold standard . In direct comparison studies, the Immulite® automated ELISA failed to detect canine TNF-alpha despite 91% sequence homology with human TNF-alpha, highlighting the critical importance of species-specific validation . Multiplex assays offer the advantage of simultaneous measurement of multiple cytokines including TNF-alpha, though sensitivity may be compromised relative to single-analyte ELISAs . Quantitative comparison of detection methods reveals significant disparities in sensitivity—lower limits of detection range from <0.10 pg/mL for high-sensitivity assays to >3.0 pg/mL for standard platforms . When evaluating TNF-alpha in canine lymphoma, standard ELISA methodology detected the cytokine in only 12% (3/25) of affected dogs, suggesting either true biological absence or insufficient assay sensitivity . Beyond technical performance parameters, practical considerations including throughput, cost, sample volume requirements, and availability of validated reagents influence method selection. Researchers should implement validation protocols including spike-recovery experiments, dilution linearity testing, and comparison with established reference methods when introducing new TNF-alpha detection platforms for canine samples.

How does TNF-alpha response to therapeutic intervention compare across different canine disease models?

TNF-alpha response to therapeutic intervention demonstrates disease-specific patterns across canine models, reflecting underlying pathophysiological mechanisms and potential clinical utility. In canine heartworm disease (HWD), significant changes in TNF-alpha concentration were not observed during alternative therapy, and TNF-alpha levels did not correlate with antigen (Ag) level changes during treatment . These findings led researchers to conclude that "quantification of TNF-α solely, without insight on its receptors, does not contribute to the diagnosis and treatment of HWD in dogs" . Conversely, in canine lymphoma, TNF-alpha was detected in 12% (3/25) of dogs at diagnosis but became undetectable following complete or partial remission, suggesting potential utility as a treatment response marker in the subset of patients with detectable levels at baseline . In experimental settings, administration of PEGylated TNF-alpha (PEG-TNF) to dogs with spontaneous cancers produced biological effects including transient fever, leukopenia, increased tumor inflammation, and necrosis across multiple tumor types . Minor or transient antitumor responses were observed in dogs with melanoma, squamous cell carcinoma, and mammary carcinoma, while a partial response was achieved in a dog with angiosarcoma . These differential patterns highlight the context-dependent nature of TNF-alpha in disease pathophysiology and treatment response. Researchers should consider disease-specific mechanisms, baseline TNF-alpha detectability, and potential receptor modulation when evaluating this cytokine as a therapeutic target or biomarker in canine disease models.

What key research gaps remain in understanding canine TNF-alpha biology and function?

Despite advances in canine TNF-alpha research, significant knowledge gaps persist, limiting comprehensive understanding of this cytokine's biology. First, the regulatory mechanisms controlling TNF-alpha expression in different canine tissues remain poorly characterized, including transcriptional control, post-translational modifications, and epigenetic regulation . Second, the interplay between TNF-alpha and its soluble versus membrane-bound receptors requires further investigation, particularly how receptor shedding modulates cytokine bioavailability and activity in different disease states . Third, breed-specific differences in TNF-alpha responses need systematic characterization beyond the current fragmentary data, including potential genomic variants affecting TNF pathway components . Fourth, the contribution of TNF-alpha to age-related inflammatory processes in canines—potentially relevant to cognitive decline and degenerative conditions—remains largely unexplored . Fifth, comprehensive mapping of downstream signaling pathways activated by TNF-alpha in different canine cell types would enhance understanding of tissue-specific responses and identify potential intervention points . Sixth, the complex interactions between TNF-alpha and other cytokines in coordinating immune responses require network-level analysis rather than isolated cytokine studies . Finally, longitudinal studies tracking TNF-alpha dynamics throughout disease progression and treatment would provide valuable insights into temporal patterns and predictive value. Addressing these research gaps would substantially advance understanding of canine TNF-alpha biology and potentially reveal new therapeutic targets and biomarker applications.

How might advanced gene editing technologies contribute to TNF-alpha research in canine models?

Advanced gene editing technologies, particularly CRISPR-Cas9 systems, offer transformative potential for TNF-alpha research in canine models through precise genetic manipulation previously impossible in this species. These technologies enable several innovative research approaches: First, targeted modification of the TNF-alpha gene itself could create reporter systems where endogenous TNF-alpha is tagged with fluorescent proteins, allowing real-time visualization of cytokine production in living tissues . Second, generation of canine cell lines or primary cells with knockout or knockdown of TNF receptors (TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR) would enable detailed dissection of receptor-specific signaling pathways and biological responses . Third, precise engineering of TNF-alpha promoter regions could identify critical regulatory elements controlling cytokine expression in different contexts, including disease states . Fourth, creation of canine cell lines with receptor-specific reporter systems would facilitate high-throughput screening of compounds modulating TNF-alpha signaling with species-specific relevance . Fifth, for in vivo applications, somatic gene editing could potentially modify TNF-alpha expression in specific tissues of canine disease models, providing insights into localized cytokine function without systemic manipulation . While technical and ethical considerations limit wholescale genetic modification of canines, targeted approaches in primary cells, organoids, and possibly localized somatic editing represent feasible applications of these technologies. These approaches would complement traditional methods while providing unprecedented precision in understanding TNF-alpha biology in this important translational species.

Product Science Overview

Canine TNF-α

In the context of canines, TNF-α is produced by various cells, including macrophages, monocytes, T and B cells, neutrophils, and endothelial cells . It has a key regulatory role in the canine immune response, where it modulates the expression of numerous cytokines, growth factors, inflammatory mediators, and acute phase proteins .

Recombinant Canine TNF-α

Recombinant Canine TNF-α is a form of TNF-α that is produced through recombinant DNA technology. This involves inserting the gene that encodes TNF-α into a host organism, such as E. coli, which then produces the protein . The recombinant protein is typically purified to high levels of purity and tested for biological activity.

Production and Purification

The recombinant canine TNF-α protein is usually expressed in E. coli and purified using methods such as High-Performance Liquid Chromatography (HPLC) and SDS-PAGE . The protein is often lyophilized and can be reconstituted in sterile PBS for use in various applications .

Applications

Recombinant canine TNF-α is used in a variety of research applications, including:

  • Cytotoxicity Assays: It is used to measure the cytotoxic effects on cells, often in the presence of metabolic inhibitors like actinomycin D .
  • Immunological Studies: It helps in studying the immune response and the role of TNF-α in inflammation and other immune processes .
  • Drug Development: It is used in the development and testing of drugs that target TNF-α pathways, which are relevant in treating inflammatory diseases .
Stability and Storage

The stability and storage conditions for recombinant canine TNF-α are crucial for maintaining its biological activity. It is typically stored at -20 to -70°C and should be reconstituted in sterile conditions to avoid contamination .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.