TNFA Bovine

Tumor Necrosis Factor-alpha Bovine Recombinant
Shipped with Ice Packs
In Stock

Description

Immune and Inflammatory Responses

  • TNF-α is rapidly released during bacterial infections (e.g., Escherichia coli, Mycoplasma bovis) and activates macrophages, lymphocytes, and endothelial cells to drive inflammation .

  • It induces apoptosis in infected cells and upregulates proinflammatory cytokines (IL-6, IL-8, COX-2/PGE2) in bovine fibroblast-like synoviocytes .

  • High-affinity decoy receptors (TNFR2-Ig) suppress TNF-α-induced cell death by competitively inhibiting TNF-α/membrane receptor interactions .

Metabolic and Reproductive Roles

  • TNF-α disrupts lipid metabolism and insulin signaling, contributing to metabolic disorders like acidosis .

  • In the estrous cycle, low doses (0.1–1 μg) induce luteolysis by increasing prostaglandin F2α (PGF2α) and nitric oxide (NO), while high doses (10–50 μg) prolong luteal function via progesterone (P4) and PGE2 synthesis .

In Vitro and Therapeutic Studies

  • TNFR-Ig Fusion Proteins: TNFR1-Ig and TNFR2-Ig (soluble TNF receptor-Fc fusions) block TNF-α bioactivity, improving cell viability by 60–80% in apoptosis assays . TNFR2-Ig shows higher binding affinity and efficacy at lower concentrations .

  • Metabolic Profiling: TNF-α reprograms bovine synoviocyte metabolism, increasing glycolysis intermediates (e.g., glucose) while reducing TCA cycle metabolites (malate, fumarate) :

Metabolite ClassKey Changes (TNF-α vs. Control)Functional Impact
Carbohydrates↑ Glucose, ↓ Galactose 1-phosphateEnhanced glycolytic flux
Fatty Acids↓ Stearic, palmitic, myristic acidsReduced lipid storage/oxidation
Amino Acids↑ Isoleucine, lysine, tyrosineIncreased protein turnover

Diagnostic Tools

  • ELISA kits (e.g., Bovine TNF-α DuoSet ELISA DY2279) detect TNF-α concentrations in plasma/serum with high sensitivity .

Therapeutic Potential and Challenges

  • TNFR2-Ig: Reduces inflammatory cytokine production (IL-6, IL-1β) by 40–60% in vitro, suggesting utility in treating mastitis or endotoxic shock .

  • Limitations: TNFR2-Ig does not eliminate pathogens and requires adjunct therapies for disease resolution .

Regulatory Status

Recombinant bovine TNF-α is labeled "For research purposes only" due to unresolved safety and efficacy profiles in vivo .

Product Specs

Introduction
Tumor necrosis factor (TNF) is a cytokine that plays a crucial role in the acute phase reaction and systemic inflammation. Primarily secreted by macrophages, TNF exerts pleiotropic effects on cells, including apoptotic cell death, proliferation, differentiation, inflammation, tumorigenesis, and viral replication. Moreover, TNF is implicated in lipid metabolism and coagulation. Dysregulation and excessive production of TNF are associated with various human diseases, including autoimmune disorders, insulin resistance, and cancer.
Description
Recombinant Bovine Tumor Necrosis Factor Alpha (TNFA), expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 158 amino acids (78-234 a.a.). This protein variant has a molecular weight of 17.5 kDa. TNFA undergoes purification using proprietary chromatographic methods to ensure high purity.
Physical Appearance
Sterile, colorless solution free from particulate matter.
Formulation
TNFA is supplied as a 1 mg/ml solution in phosphate-buffered saline (pH 7.4) containing 10% glycerol.
Stability
For short-term storage (up to 4 weeks), store the vial at 4°C. For extended storage, freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Biological Activity
The biological activity, measured by a cytotoxicity assay using L929 mouse fibrosarcoma cells in the presence of actinomycin D, is characterized by an ED50 value of less than 15 ng/ml.
Synonyms

TNF-alpha, Tumor necrosis factor ligand superfamily member 2, TNF-a, Cachectin, DIF, TNFA, TNFSF2.

Source

Escherichia Coli.

Amino Acid Sequence

MLRSSSQASS NKPVAHVVAD INSPGQLRWW DSYANALMAN GVKLEDNQLV VPADGLYLIY SQVLFRGQGC PSTPLFLTHT ISRIAVSYQT KVNILSAIKS PCHRETPEWA EAKPWYEPIY QGGVFQLEKG DRLSAEINLP DYLDYAESGQ VYFGIIAL. 

Q&A

What is bovine TNF-α and what are its primary biological functions?

Bovine tumor necrosis factor alpha (TNF-α) is a potent lymphoid factor and pleiotropic cytokine that plays critical roles in regulating growth, differentiation, inflammation, viral replication, tumorigenesis, and autoimmune diseases in cattle. TNF-α exerts cytotoxic effects on a wide range of tumor cells and is involved in tumor necrosis, tumor metastasis, viral replication, septic shock, fever, inflammation, and various pathological conditions including Crohn's disease, rheumatoid arthritis, and graft-versus-host disease . As a prototypic member of the TNF superfamily, bovine TNF-α is produced immediately upon pathogen invasion, particularly in bacterial infections, and induces various immune responses such as cell proliferation and inflammation .

How does bovine TNF-α compare structurally with TNF-α from other species?

Bovine TNF-α shares varying degrees of amino acid sequence identity with TNF-α from other species. For instance, mouse TNF-α shares 70-77% amino acid sequence identity with bovine TNF-α within the extracellular domain . This moderate homology indicates conservation of core functional domains while allowing for species-specific variations that may affect receptor binding affinities and downstream signaling. The structural similarities enable some cross-reactivity in experimental systems but also necessitate species-specific reagents for optimal research outcomes.

What are the primary cellular sources of TNF-α in cattle?

In bovine systems, TNF-α is primarily produced by activated macrophages and T cells . During infection or inflammatory challenge, these immune cells rapidly secrete TNF-α as part of the innate immune response. Additionally, bovine CD14+ monocytes and CD4+/CD8+ lymphocytes have been identified as significant sources of intracellular TNF-α , demonstrating the involvement of multiple leukocyte populations in TNF-α production. Various bacterial pathogens, particularly Escherichia coli and Mycoplasma bovis, can trigger prompt release of TNF-α from these cells .

What are the current methods for detecting and quantifying bovine TNF-α in research samples?

Several methodologies have been developed for detecting and quantifying bovine TNF-α:

  • Monoclonal antibody-based assays: Specific monoclonal antibodies (e.g., clones 197-1 and 65-2, murine IgG1 isotypes) have been developed to detect both native and recombinant bovine TNF-α. These antibodies can be used in various immunological assays and lack cross-reactivity with other bovine cytokines .

  • Fluorescent bead-based assays: These assays use anti-TNF-α monoclonal antibodies with a detection range of 0.2 to 620 ng/mL, suitable for quantifying native bovine TNF-α in cell culture supernatants and plasma samples .

  • Flow cytometry: For intracellular TNF-α detection in bovine leukocyte populations, including CD14+ monocytes and CD4+/CD8+ lymphocytes .

  • ELISA-based methods: For detection of TNF-α in plasma, cell culture supernatants, and tissue homogenates.

  • Cell-based bioassays: Measuring the cytotoxic effects of TNF-α on susceptible cell lines, with cytotoxicity on porcine PK-15 cells showing an ED50 of 0.05-0.15 ng/mL .

How can I validate the specificity of antibodies used for bovine TNF-α detection?

Validation of antibody specificity for bovine TNF-α should follow these methodological steps:

  • Test for cross-reactivity with related bovine cytokines (e.g., IFN-γ, IL-10, CCL5) and cytokines from other species that might contaminate reagents (e.g., equine IL-4) .

  • Confirm detection of both recombinant and native bovine TNF-α in positive control samples such as supernatants from stimulated peripheral blood mononuclear cells (PBMC).

  • Perform spike-and-recovery tests in different matrices (culture medium, serum, plasma) to evaluate matrix effects. Recovery rates should ideally fall within 80-120% .

  • Conduct serial dilutions of positive samples to confirm linearity of detection across the assay's dynamic range.

  • Include appropriate negative controls (unstimulated cells) and isotype control antibodies for flow cytometry applications.

What are the optimal sample collection and storage conditions for preserving bovine TNF-α activity?

To maintain bovine TNF-α stability and activity:

  • For recombinant proteins:

    • Store lyophilized preparations at -20°C to -80°C

    • After reconstitution, aliquot and store at -20°C to -80°C

    • Avoid repeated freeze-thaw cycles

    • Reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin for optimal stability

  • For biological samples:

    • Process blood samples within 2-4 hours of collection

    • Separate plasma/serum promptly and store at -80°C

    • For longer-term storage, consider adding protease inhibitors to prevent degradation

    • When preparing cell culture supernatants, centrifuge to remove cellular debris before freezing

How can I establish a bovine TNF-α-induced inflammation model in vitro?

To establish a bovine TNF-α-induced inflammation model:

  • Cell preparation: Isolate primary bovine cells (e.g., chondrocytes, synoviocytes) or use established bovine cell lines. For primary chondrocytes, harvest bovine cartilage from young cattle (4-6 months), isolate cells, expand in monolayer, and transfer to 3D pellet culture .

  • TNF-α stimulation: After an adaptation period (e.g., one week in free-swelling conditions for 3D cultures), supplement medium with recombinant bovine TNF-α. A concentration range of 0.1-100 ng/mL is recommended for dose-response studies, with 48 hours being a common stimulation period .

  • Experimental design: Include multiple biological replicates (different donors) and technical replicates. Design a concentration gradient to establish dose-dependent effects.

  • Inflammatory response assessment: Measure various parameters including:

    • Gene expression of inflammatory markers (qPCR)

    • Nitric oxide (NO) production (Griess reaction assay)

    • Glycosaminoglycan (GAG) release (Blyscan™ assay)

    • Cell viability/apoptosis (ELISA for mono- and oligonucleosomes)

    • Pro-inflammatory cytokine secretion (IL-6, IL-8, IL-1β)

    • Metabolic changes (GC/MS metabolomic analysis)

What concentrations of recombinant bovine TNF-α are optimal for different experimental applications?

Optimal concentrations vary by application:

Experimental ApplicationRecommended TNF-α ConcentrationNotes
Cytotoxicity assays using PK-15 cells0.05-0.15 ng/mL (ED50)Lower concentrations effective for sensitive bioassays
General inflammatory stimulation0.025-0.1 μg/mL (ED50)Standard range for most inflammation models
Chondrocyte inflammation model0.1-100 ng/mLUsed in dose-response studies; typically 48h exposure
Metabolic reprogramming studies in bFLS10-50 ng/mLEffective for inducing glycolytic shift
In vivo inflammation modelsVaries by experimental designDaily administration may be required for sustained effects

Titration experiments are recommended for each specific cell type and experimental endpoint to determine optimal concentration.

How do I design metabolic flux experiments to study bovine TNF-α effects on cellular metabolism?

Metabolic flux experiments examining bovine TNF-α effects should follow these methodological steps:

  • Cell preparation: Culture bovine cells (e.g., fibroblast-like synoviocytes) until reaching appropriate confluence.

  • TNF-α treatment: Treat cells with bovine TNF-α at determined concentrations (based on pilot studies).

  • Isotope labeling: Incubate cells with isotopically labeled substrates (e.g., D-glucose-13C6) for sufficient time (approximately 12 hours) to reach isotopic steady state conditions .

  • Metabolite extraction: Extract intracellular metabolites using appropriate solvents and protocols optimized for metabolomics.

  • GC/MS analysis: Analyze samples using gas chromatography-mass spectrometry to determine multiple ion detections (MIDs) and correct for natural isotope abundance .

  • Data interpretation: Calculate the relative abundance of different isotopologs (e.g., M+0, M+2, M+3) for key metabolites like pyruvate, citrate, and malate. An increase in pyruvate M+3 with decreased citrate and malate M+2 isotopologs suggests a redirecting of glucose carbon flow away from the TCA cycle .

  • Comparison: Compare metabolic patterns between TNF-α-treated and control cells to identify metabolic rewiring events.

How does bovine TNF-α contribute to inflammatory conditions in cattle?

Bovine TNF-α plays multiple roles in inflammatory conditions:

  • Direct pro-inflammatory effects: TNF-α stimulates production of other inflammatory mediators, including IL-6, IL-8, IL-1β, and COX-2/PGE2 in bovine cells .

  • Metabolic reprogramming: TNF-α induces a shift toward glycolysis in bovine cells, characteristic of inflammatory activation. This is evidenced by increased glucose utilization, decreased TCA cycle activity, and altered fatty acid metabolism .

  • Joint inflammation: Elevated TNF-α levels in synovial fluid are associated with arthritis in cattle. TNF-α activates fibroblast-like synoviocytes, contributing to joint inflammation and damage .

  • Laminitis: Serum TNF-α concentrations are elevated in cattle with lameness and foot rot. In vitro studies show increased TNF-α release from bovine hoof dermal cells following LPS stimulation, supporting its role in laminitis pathogenesis .

  • Metabolic disorders: During the transition period, higher serum TNF-α concentrations are observed in lame cows compared to healthy animals, suggesting a link between inflammation and metabolic health .

What is the relationship between bovine TNF-α and metabolic disorders in dairy cattle?

Bovine TNF-α significantly impacts metabolic functions in dairy cattle:

  • Impaired gluconeogenesis: TNF-α administration reduces liver gluconeogenic gene expression in lactating cows, potentially contributing to hypoglycemia .

  • Increased lipid mobilization: The resulting hypoglycemia can increase adipose tissue triglyceride mobilization, exacerbating negative energy balance in early-lactation cows .

  • Altered nutrient partitioning: TNF-α administration during early lactation impairs milk production, suggesting redirected nutrient utilization from mammary gland to inflammatory processes .

  • Metabolic stress amplification: Inflammatory challenges via TNF-α exacerbate metabolic stress in early-lactation cows, creating a vicious cycle that adversely affects production and health status .

  • Transition period vulnerability: The natural metabolic adaptations during the transition period make dairy cows particularly susceptible to TNF-α's detrimental metabolic effects, potentially predisposing them to conditions like ketosis and fatty liver .

How can I design experiments to investigate bovine TNF-α's role in specific disease conditions?

For investigating bovine TNF-α in disease conditions:

  • In vitro disease models:

    • Simulate joint inflammation by treating bovine synoviocytes with TNF-α and measuring inflammatory mediator production and metabolic changes

    • Model laminitis by treating bovine hoof dermal cells with TNF-α and/or LPS

    • Create hepatic steatosis models by exposing bovine hepatocytes to TNF-α and measuring effects on lipid accumulation and glucose metabolism

  • Ex vivo approaches:

    • Collect tissues from healthy and diseased animals to compare TNF-α expression and signaling pathway activation

    • Use whole blood stimulation assays to assess TNF-α production capacity in different disease states

  • In vivo studies:

    • Design controlled trials with recombinant bovine TNF-α administration to healthy animals, monitoring physiological parameters, production outcomes, and health status

    • Consider dose, duration, and timing relative to physiological state (e.g., early vs. late lactation)

    • Include appropriate control groups and sufficient animal numbers for statistical power

  • Disease correlation studies:

    • Measure TNF-α levels in biological samples from animals with naturally occurring diseases

    • Correlate TNF-α concentrations with disease severity, metabolic parameters, and production outcomes

What are the potential therapeutic approaches targeting bovine TNF-α for inflammatory diseases?

Several approaches for targeting bovine TNF-α show therapeutic potential:

  • Soluble TNF receptor fusion proteins: Bovine sTNFR1 and sTNFR2 IgG1 Fc-fusion proteins (TNFR1-Ig and TNFR2-Ig) can function as decoy receptors, competitively inhibiting TNF-α binding to membrane TNF receptors .

  • Anti-TNF-α antibodies: Monoclonal antibodies that specifically neutralize bovine TNF-α could be developed for therapeutic applications, similar to those used in human medicine.

  • Small molecule inhibitors: Compounds that inhibit TNF-α production or signaling could be developed or adapted from human medicine for veterinary applications.

  • Targeted delivery approaches: Liposomal or nanoparticle-based delivery systems could improve the pharmacokinetics and tissue targeting of anti-TNF-α therapeutics.

Research suggests TNFR2-Ig may be particularly effective for attenuating excessive inflammation in cattle, with demonstrated in vitro efficacy in inhibiting inflammatory cytokine production .

How effective are soluble TNF receptor fusion proteins in neutralizing bovine TNF-α activity?

Studies on soluble TNF receptor fusion proteins (TNFR-Ig) show:

  • Biochemical properties: TNFR1-Ig, TNFR2-Ig form dimers with molecular weights of approximately 50 kDa and 70 kDa, respectively .

  • Binding activity: Both TNFR1-Ig and TNFR2-Ig competitively inhibit TNF-α/membrane TNFR interactions, with TNFR2-Ig showing superior efficacy in most experimental systems .

  • Anti-inflammatory effects: TNFR2-Ig can inhibit inflammatory cytokine production, thereby attenuating excessive inflammation in in vitro models .

  • Therapeutic limitations: TNFR-Ig treatment alone cannot eradicate diseases as it targets inflammation rather than pathogen elimination. Therefore, it is best suited for attenuating acute and severe inflammatory responses rather than as a standalone treatment .

  • Application scope: These proteins show promise for controlling inflammatory diseases in cattle, particularly those involving bacterial infections like E. coli and Mycoplasma bovis, which trigger TNF-α release .

What methodological considerations are important when evaluating anti-TNF-α interventions in bovine systems?

When evaluating anti-TNF-α interventions:

  • Model selection:

    • Consider both in vitro systems using relevant bovine cell types and in vivo models

    • Include bacterial antigens or live bacteria in models, as disease progressions involve complex interactions between pathogens and host immunity

    • Use appropriate bovine primary cells (e.g., mammary epithelial cells for mastitis studies)

  • Efficacy assessment:

    • Measure direct neutralization of TNF-α activity

    • Evaluate downstream effects on inflammatory mediators

    • Assess functional outcomes like cell viability or tissue damage

    • Consider combination therapies with antimicrobials for infectious conditions

  • Experimental design:

    • Include proper controls (negative control proteins like Cont-Ig)

    • Test dose-dependency and time-course effects

    • Consider prophylactic versus therapeutic administration timing

    • Evaluate potential compensatory mechanisms

  • Translation considerations:

    • Address pharmacokinetic and pharmacodynamic properties in bovine systems

    • Consider practical administration routes and frequencies for field applications

    • Evaluate cost-effectiveness for agricultural implementation

    • Assess potential immunogenicity of biological therapeutics

How does bovine TNF-α induce metabolic reprogramming in target cells?

Bovine TNF-α induces significant metabolic reprogramming, particularly in inflammatory contexts:

  • Glycolytic shift: GC/MS metabolomic analysis reveals increased glucose levels in TNF-α-treated bovine fibroblast-like synoviocytes (bFLS), indicating enhanced glucose uptake .

  • TCA cycle suppression: TNF-α treatment decreases TCA cycle intermediates including malate, fumarate, and α-ketoglutarate (α-KG) in bovine cells .

  • Altered carbon flow: Metabolic flux experiments using D-glucose-13C6 demonstrate increased pyruvate M+3 isotopologs with decreased citrate and malate M+2 isotopologs in TNF-α-treated cells, indicating reduced flow of glucose carbons into the TCA cycle .

  • Fatty acid metabolism changes: Significant decreases in fatty acid methyl esters (FAMEs), including stearate, palmitate, and laurate, occur in TNF-α-treated bFLS, suggesting altered lipid metabolism .

  • Amino acid metabolism: TNF-α treatment increases certain amino acids (isoleucine, lysine, tyrosine, leucine, and valine) while reducing others (ornithine and methionine), indicating complex effects on protein metabolism and potentially alternative carbon sources for cellular energetics .

Metabolite CategoryChanges in TNF-α-treated bFLSSignificance
GlucoseIncreasedEnhanced glycolytic activity
TCA cycle intermediatesDecreasedReduced oxidative metabolism
Fatty acidsDecreasedAltered lipid metabolism
Amino acidsVariable (some increased, some decreased)Complex effects on protein metabolism

What are the signaling mechanisms mediating bovine TNF-α effects in different cell types?

Bovine TNF-α activates multiple signaling pathways, with some similarities to human systems but also species-specific aspects:

  • NF-κB pathway activation: Bovine TNF-α induces the expression of pro-inflammatory cytokines such as IL-6 and IL-8 via the NF-κB pathway in bovine fibroblast-like synoviocytes .

  • MAPK signaling: TNF-α likely activates p38 MAPK, ERK, and JNK pathways, contributing to inflammatory gene expression and metabolic changes.

  • Cell death signaling: In susceptible cells, bovine TNF-α can induce apoptosis through death domain-containing receptors, though the exact molecular components may differ from human systems.

  • Metabolic signaling: The metabolic reprogramming observed in TNF-α-treated cells suggests involvement of metabolic regulators like mTOR, AMPK, and HIF-1α, though these pathways need further characterization in bovine systems.

  • Cell type-specific responses: Different bovine cell types (immune cells, epithelial cells, fibroblasts) may exhibit distinct signaling patterns and sensitivity thresholds to TNF-α stimulation.

How can single-cell analysis technologies advance our understanding of bovine TNF-α biology?

Single-cell technologies offer powerful approaches for bovine TNF-α research:

  • Cellular heterogeneity: Single-cell RNA sequencing (scRNA-seq) can identify subpopulations of bovine immune cells with differential TNF-α production capacities or responsiveness.

  • Dynamic response mapping: Time-resolved single-cell analyses can track the temporal evolution of TNF-α-induced transcriptional programs in bovine cells.

  • Spatial context: Spatial transcriptomics can reveal tissue-specific patterns of TNF-α expression and response in disease states, preserving the anatomical context.

  • Multi-omics integration: Combining single-cell transcriptomics with proteomics or metabolomics can provide comprehensive understanding of how TNF-α reshapes cellular function.

  • Methodological considerations:

    • Sample preparation must preserve viability of bovine cells

    • Antibody panels for CyTOF or flow cytometry must be validated for bovine antigens

    • Bioinformatic pipelines may need adaptation for bovine genome and transcriptome

    • Reference datasets for healthy bovine tissues should be established as benchmarks

Implementing these advanced technologies requires careful optimization for bovine systems but offers unprecedented insights into TNF-α biology in cattle.

What are the key unresolved questions in bovine TNF-α research?

Several important questions remain in bovine TNF-α research:

  • Receptor-specific signaling: How do bovine TNFR1 and TNFR2 differentially contribute to inflammatory and metabolic effects in various tissues?

  • Species-specific mechanisms: What unique aspects of bovine TNF-α signaling differ from better-characterized human systems?

  • Physiological states: How does the TNF-α response network change across different physiological states in cattle (e.g., pregnancy, lactation, growth phases)?

  • Genetic variation: What genetic polymorphisms in the bovine TNF-α pathway influence disease susceptibility and production traits?

  • Microbiome interactions: How does the bovine microbiome influence TNF-α production and activity, particularly in gastrointestinal and mammary health?

  • Therapeutic optimization: What are the optimal formulations, doses, and administration regimens for anti-TNF-α therapeutics in cattle?

  • Long-term adaptations: What compensatory mechanisms develop during chronic TNF-α elevation or prolonged anti-TNF-α treatment?

What novel technologies might advance bovine TNF-α research in the next decade?

Emerging technologies with potential to transform bovine TNF-α research include:

  • CRISPR/Cas9 gene editing: Creating precise genetic modifications in bovine cells or potentially whole animals to study TNF-α pathway components.

  • Organoid models: Developing bovine tissue-specific organoids to study TNF-α effects in complex 3D microenvironments that better mimic in vivo conditions.

  • Advanced imaging techniques: Implementing intravital microscopy and other advanced imaging to visualize TNF-α signaling dynamics in living tissues.

  • Systems biology approaches: Applying computational modeling to integrate multi-omics data and predict TNF-α network behaviors under various conditions.

  • Nanobody and aptamer technologies: Developing smaller, more stable alternatives to conventional antibodies for TNF-α targeting with improved tissue penetration.

  • Extracellular vesicle analysis: Investigating how TNF-α influences the cargo and function of extracellular vesicles in intercellular communication.

  • Precision livestock technologies: Integrating wearable sensors and continuous monitoring to correlate real-time physiological data with TNF-α biomarkers.

How might bovine TNF-α research inform comparative immunology across species?

Bovine TNF-α research contributes valuable comparative immunology insights:

  • Evolutionary conservation: Understanding conserved versus divergent aspects of TNF-α biology across species illuminates evolutionary pressures on inflammatory pathways.

  • Disease modeling: Bovine inflammatory conditions may serve as naturally occurring models for human disorders, offering comparative insights into pathogenesis.

  • Cross-species therapeutic applications: Successful anti-TNF-α approaches in cattle might inspire novel therapeutic strategies for human diseases, and vice versa.

  • Host-pathogen interactions: Bovine-specific responses to pathogens through TNF-α pathways may reveal unique immune defense mechanisms.

  • Metabolic-immune interfaces: The prominent metabolic effects of TNF-α in cattle during physiological transitions provide a window into integrated metabolic-immune regulation.

  • Tissue-specific responses: Comparative analysis of tissue-specific TNF-α effects across species could reveal specialized adaptations in different anatomical contexts.

  • Environmental adaptations: Understanding how bovine TNF-α responses adapt to various environmental challenges may inform broader concepts in eco-immunology.

Product Science Overview

Introduction

Tumor Necrosis Factor-alpha (TNF-α) is a crucial cytokine involved in systemic inflammation and is part of the body’s immune response. It is a member of the TNF superfamily, which consists of various cytokines that can cause cell death (apoptosis). TNF-α is known for its role in inflammation, immune system development, apoptosis, and lipid metabolism .

Structure and Function

TNF-α is a pleiotropic molecule, meaning it has multiple effects on different types of cells. It is produced primarily by macrophages, a type of white blood cell that engulfs and digests cellular debris and pathogens. TNF-α can bind to two receptors: TNFR1 and TNFR2. The binding of TNF-α to these receptors can trigger various cellular responses, including cell death, survival, proliferation, and differentiation .

Recombinant Bovine TNF-α

Recombinant bovine TNF-α is a form of TNF-α that has been genetically engineered for use in research and therapeutic applications. This recombinant protein is produced by inserting the gene that encodes bovine TNF-α into a suitable expression system, such as bacteria or yeast, which then produces the protein in large quantities .

Applications in Research and Medicine

Recombinant bovine TNF-α is used extensively in research to study its effects on various cell types and its role in different diseases. For instance, it has been shown to play a significant role in inflammatory diseases in cattle, such as mastitis, which is an inflammation of the mammary gland . By understanding how TNF-α functions, researchers can develop new therapeutic strategies to treat these diseases.

In addition, recombinant bovine TNF-α has been used to study the effects of TNF-α on cell death and inflammatory cytokine kinetics. For example, studies have shown that TNF-α can induce cell death in certain cell types, and this effect can be modulated by using decoy receptors that bind to TNF-α and prevent it from interacting with its natural receptors .

Therapeutic Potential

The therapeutic potential of recombinant bovine TNF-α lies in its ability to modulate the immune response. By blocking the activity of TNF-α, it is possible to reduce inflammation and improve outcomes in diseases characterized by excessive inflammation. For example, decoy receptors for TNF-α have been developed and shown to be effective in reducing inflammation in vitro .

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.