Recombinant Human Lymphotoxin-alpha protein (LTA) (Active)

Shipped with Ice Packs
In Stock

Description

Biological Functions and Receptor Interactions

LTA mediates immune responses through:

  • Homotrimeric Binding: Activates TNFR1 (TNFRSF1A), TNFR2 (TNFRSF1B), and HVEM (TNFRSF14) .

  • Heterotrimeric Binding: Forms complexes with LT-beta to activate LT-beta R (TNFRSF3), crucial for lymphoid organ development .

Key Roles:

  • Lymphoid Organ Development: Essential for lymph node and spleen structure .

  • Inflammation: Induces adhesion molecules (ICAM-1, E-selectin) and chemokines (RANTES, MCP-1) in endothelial cells .

  • Cytotoxicity: Kills tumor cells in vitro and in vivo .

  • Autoimmunity: Elevated in rheumatoid arthritis (RA) synovial tissue and serum .

Table 1: Functional Assay Data

AssayTargetEC₅₀ RangeSource
ELISA (Binding)TNFR14.41–6.80 ng/mL
TNFR21.63–2.70 ng/mL
CytotoxicityTumor cells4–20 pg/mL (ED₅₀)

Applications:

  • Immune Studies: Used to model lymphoid tissue organization and inflammation .

  • Cancer Research: Evaluates tumor cell cytotoxicity and immunotherapy strategies .

  • Autoimmune Disease Models: Tested in collagen-induced arthritis (CIA) to assess therapeutic blockade efficacy .

Therapeutic Potential and Clinical Relevance

LTA’s dual role in inflammation and immune regulation positions it as a target for therapies:

  • RA Treatment: Anti-LTA antibodies improved outcomes in TNF-alpha-refractory patients by depleting LTA-expressing T cells .

  • Lymphangiogenesis: Promotes lymphatic vessel formation in inflammation, relevant to cancer metastasis .

  • NK Cell Activity: Enhances natural killer (NK) cell recruitment and antitumor responses .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4, with 0.02% Tween-20
Description

Recombinant Human Lymphotoxin-alpha protein (LTA) (Active) is a partial-length protein expressed without a tag in *E. coli*. Its expression region corresponds to amino acids 35-205 of human LTA protein. Purity was determined by SDS-PAGE and reached up to 96%, exhibiting a molecular mass band around 18.7 kDa on the gel. Biological activity was validated through functional ELISA analyses. The endotoxin level of this recombinant protein is less than 1.0 EU/µg as determined by the LAL method. This recombinant LTA protein is in stock and available for immediate purchase.

LTA is a cytokine that, in its homotrimeric form, binds to TNFRSF1A/TNFR1, TNFRSF1B/TNFBR, and TNFRSF14/HVEM. In its heterotrimeric form with LTB, it binds to TNFRSF3/LTBR. Lymphotoxin is produced by lymphocytes and is cytotoxic for a wide range of tumor cells *in vitro* and *in vivo*. Diseases associated with LTA include Psoriatic Arthritis and Leprosy.

Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by several factors, including storage conditions, buffer ingredients, storage temperature, and the stability of the protein itself.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
LTalpha; DAMA-25N12.13-004; DIF; LT alpha; LT; LT-alpha; Lta; Lymphotoxin alpha (TNF superfamily, member 1); Lymphotoxin alpha; Lymphotoxin-alpha; MGC117668; OTTHUMP00000037612; OTTHUMP00000037613; TNF B; TNF superfamily member 1; TNF, lymphocyte-derived; TNF-beta; TNFB; TNFB_HUMAN; TNFbeta; TNFSF 1; TNFSF1; TNLG1E; Tumor necrosis factor beta; tumor necrosis factor ligand 1E; Tumor necrosis factor ligand superfamily member 1; tumor necrosis factor superfamily member 1
Datasheet & Coa
Please contact us to get it.
Expression Region
35-205aa
Mol. Weight
18.7 kDa
Protein Length
Full Length of Mature Protein
Purity
>96% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
E.coli
Species
Homo sapiens (Human)
Target Names
LTA
Uniprot No.

Target Background

Function

Cytokine that in its homotrimeric form binds to TNFRSF1A/TNFR1, TNFRSF1B/TNFBR and TNFRSF14/HVEM. In its heterotrimeric form with LTB binds to TNFRSF3/LTBR. Lymphotoxin is produced by lymphocytes and is cytotoxic for a wide range of tumor cells *in vitro* and *in vivo*.

Gene References Into Functions
  1. LTA levels were significantly increased in SZ patients, but no significant difference in the mRNA levels of LTA was observed between SZ patients and NC subjects. PMID: 28476335
  2. There are independent and additive interactions between LTalpha +252 G/G genotype, chronic viral hepatitis, and habits of substance use on the risk of hepatocellular carcinoma. PMID: 28738973
  3. Independent and additive interaction between polymorphisms of TNFalpha -308 and lymphotoxin alpha +252 on the risk of hepatocellular carcinoma related to hepatitis B has been reported. PMID: 28865603
  4. The C allele for rs1042522 in p53 was genetically associated with a higher risk for RD but not for PVR in this cohort. This is the first association study attempting replication of PVR-associated risk alleles in a nonwhite population. PMID: 28106707
  5. The results of this study showed that neither -252 G/A nor -804 C/A polymorphism of the LTA gene was found to be associated with overall stroke as well as any subtype of IS excluding SVD in the North Indian population. PMID: 26707826
  6. This study found an association between the single nucleotide polymorphism LTA +252 with the development of sepsis in Colombian patients. PMID: 27592234
  7. Frequencies of polymorphic allele and genotypes for the lymphotoxin-alpha gene, position +252 (rs909253), in Brazilian women with preeclampsia. PMID: 26561241
  8. The allelic frequencies of LTA SNPs were found to be significantly associated with the risk of oral cancer and precancerous lesions. PMID: 27312561
  9. rs909253 in LTA was identified to be significantly associated with ankylosing spondylitis susceptibility in this study population, but no significant association was found between ankylosing spondylitis risk and LTA rs2239704 or rs2229094. PMID: 28489756
  10. The study showed that the heterozygous variant (AG) genotype of TNF-beta was associated with persistent primary immune thrombocytopenia, when compared with controls. PMID: 26761582
  11. A positive correlation of HLA-DRB1*12 and a negative correlation of HLA-DRB1*13 with younger patients. TNFb4 allele's negative association with older patients displaying higher PSA levels, higher GS and positive surrounding tissue involvement; positive association of TNFb5 allele for both older and younger patients. PMID: 27102235
  12. The present study suggests LTa +252G as a risk allele for disease susceptibility associated with higher serum levels of LTa and concomitant discrete clinical features among Indian systemic lupus erythematosus patients in India. PMID: 27838362
  13. This study shows that TNF-b is elevated in wound exudates of patients with hidradenitis suppurativa. PMID: 27819528
  14. LTA -804C/A gene polymorphism is not found to be associated with the susceptibility of ischemic stroke in both Asian as well as in the Caucasian population. PMID: 27411794
  15. After multivariable analysis, TNF-alpha polymorphism showed no consistent association with leukemia. CONCLUSIONS: This meta-analysis suggests that the LT-alpha +252 AA polymorphism is associated with the risk of leukemia. PMID: 27647233
  16. Tumor Necrosis Factor and Lymphotoxin in Regulation of Intestinal Inflammation. PMID: 27914457
  17. Joint effects of these CRP, TNF-alpha, and LTA risk alleles with physical inactivity in elders were observed, suggesting that physical activity may modulate effects of genotypes on handgrip strength. PMID: 27056089
  18. No association was found between two promoter variants of TNF and LTA, and diabetic retinopathy in a large cohort of Caucasian patients with type 1 diabetes and type 2 diabetes. PMID: 26821796
  19. Polymorphism +252 LT gene did not show any significant association with COPD. PMID: 26932696
  20. TNFB+252G/A was associated with the presence of immune thrombocytopenia. PMID: 26076780
  21. LTalpha plays a role in malignant angiogenesis and disease progression in Cutaneous T cell lymphoma and may serve as a therapeutic target in this disease. PMID: 25915535
  22. LTA polymorphism is associated with genetic susceptibility of systemic lupus in Asians, but not in rheumatoid arthritis. PMID: 25931031
  23. Studied the association between immuno-modulatory gene polymorphisms (including LTA) and risk for nasal NK/T-cell lymphoma in a Chinese population. Found The LTA +252 GA + AA genotypes were associated with increased risk for NK/T-cell lymphoma. PMID: 26108796
  24. TNF-beta NcoI polymorphism, by itself, was not associated with increased stroke susceptibility. But the homozygous genotype for the allele TNFB2 was associated with higher expression of classical inflammatory and metabolic markers of stroke. PMID: 25063351
  25. These results support an association between ANKK1 and LTA genetic markers and vulnerability to schizophrenia and show the potential influence of just one copy of the mutant C or G allele in the Egyptian population. PMID: 26114114
  26. Studied six SNP loci: (rs2279115 of BCL2 gene, rs804270 of NEIL2 gene, rs909253 of LTA gene, rs2294008 of PSCA gene, rs3765524 and rs10509670 of PLCE1 gene) to evaluate gastric cancer risk using magnetic nanoparticles and universal tagged arrays. PMID: 26554163
  27. TNF-alpha (-308G/A), TNF-beta (+252A/G) and IL-10 (-1082G/A, -819C/T and -592C/A) polymorphisms are associated with the susceptibility of oral lichen planus. PMID: 26221924
  28. Elite controllers of HIV infection present marginal zone-like B-cell populations which IL-10 and LT-alpha expression profiles may favor homeostasis of immune responses and lymphoid microenvironments. PMID: 25003989
  29. Biotin-pulldown assay showed that HuR specifically interacts with the 3'-untranslated region (3'-UTR) of the LTalpha mRNA. PMID: 25980610
  30. The AA genotype of LT-alpha + 252 was significantly associated with shortened overall or leukemia-free survival in patients with myelodysplastic syndrome with excess blasts. PMID: 23931336
  31. MTHFR, TGFbeta1, and TNFB polymorphisms are not significantly associated with the risk of osteoporosis in rheumatoid arthritis. PMID: 25981594
  32. Statistical evidence for the interactions between 1) TNF/LTA SNP rs2229094 and depression symptoms for average pain intensity and duration and 2) IL1B two SNP diplotype and kinesiophobia for average shoulder pain intensity. PMID: 24598699
  33. The TNFB2/B2 genotype of TNF-beta NcoI polymorphism was associated with increased inflammatory and metabolic markers and this association was different according to sex of MS patients. PMID: 25173940
  34. Review/Meta-analysis: Suggest lymphotoxin-alpha +252A/G gene polymorphism is a risk factor for NHL in North Americans, and this polymorphism may contribute to diffuse large B cell lymphoma susceptibility. PMID: 24610621
  35. Combinations of cytokine gene network polymorphic markers as potential predictors of myocardial infarction. PMID: 25735143
  36. The findings appear to support the hypothesis that genetic variability of 252A>G polymorphism in the TNF-beta region may modulate the risk of migraine in the population of Asian ancestry. PMID: 24959879
  37. Studies indicate that tumor necrosis factors TNF-alpha/TNF-beta and their receptor TNFR1 and TNFR2 play a regulatory role of different immune cells. PMID: 24896334
  38. This indicates an association between polymorphism of the TNF-beta gene and post-operative sepsis, suggesting the TNFB2/B2 genotype as a high-risk factor for the development of sepsis after elective surgery. PMID: 24796628
  39. IL-6-174 SNP is rare or not seen in the Han population in Guangzhou, so SNP at this locus cannot be selected for disease association analysis. PMID: 25140780
  40. Our findings suggest that Hodgkin and Reed Sternberg cell-derived LTalpha is an important mediator that contributes to T cell recruitment into lesional lymph nodes in Hodgkin lymphoma. PMID: 25139349
  41. TNFB2 allele was associated with the presence of multiple sclerosis independently of HLA-DRB1 in white patients and the TNFB2/B2 genotype was associated with increased TNF-alpha levels in white and brown patients. PMID: 24696164
  42. TNFB +252A/G and exon 3 C/A polymorphisms are associated with vitiligo susceptibility. PMID: 24312346
  43. A significant association was detected between LT-alpha +249A/G and increased risk of diabetes, in particular for young-onset patients. PMID: 24233435
  44. TNF and LTA genetic polymorphisms contribute to SLE susceptibility in the Egyptian population and are associated with disease characteristics. PMID: 24420856
  45. Polymorphisms of the LTA gene can probably be used with other genetic markers together to identify individuals at high susceptibility to myocardial infarction. PMID: 24642747
  46. These results suggest for the first time that TNF-beta is involved in microenvironment inflammation in chondrocytes during rheumatoid arthritis. PMID: 24283517
  47. Our meta-analyses suggested that the LTA rs1041981, rs2239704 and rs2229094 polymorphisms contributed to the increased risk of cancers. PMID: 24349304
  48. This study provides evidence for rs1799724 at the LTA/TNFalpha locus as a susceptibility factor for idiopathic achalasia. PMID: 24259423
  49. TNF-alpha -308 A/G and LT-alpha +252 A/G polymorphisms are associated with susceptibility to sarcoidosis in an European population. [META-ANALYSIS] PMID: 24197700
  50. The TNF-beta gene A252G polymorphism might be a potential risk factor for the development of sarcoidosis. (Meta-analysis) PMID: 24244632

Show More

Hide All

Database Links

HGNC: 6709

OMIM: 153440

KEGG: hsa:4049

STRING: 9606.ENSP00000403495

UniGene: Hs.36

Involvement In Disease
Psoriatic arthritis (PSORAS)
Protein Families
Tumor necrosis factor family
Subcellular Location
Secreted. Membrane. Note=The homotrimer is secreted. The heterotrimer is membrane-associated.

Q&A

What is the molecular structure of recombinant human LTA protein?

Recombinant human Lymphotoxin-alpha/TNF-beta is a 22 kDa protein derived from E. coli expression systems. The protein consists of amino acids Leu35-Leu205 with an N-terminal methionine. Biologically active LTA forms homotrimers that bind to and activate specific receptors including TNF RI/TNFRSF1A, TNF RII/TNFRSF1B, and HVEM/TNFRSF14. The protein shares approximately 73% amino acid sequence identity with mouse and rat LTA/TNF-beta, which is important to consider when designing cross-species experiments .

What cell types express LTA and under what conditions?

LTA is primarily expressed by activated T and B lymphocytes. Expression is tightly regulated and typically occurs during immune activation. In research contexts, understanding the activation conditions that promote LTA expression is crucial for experimental design. While constitutive expression is limited, various activation protocols using cytokines, mitogens, or receptor-mediated signaling can induce LTA expression in lymphocytes .

What is the optimal reconstitution protocol for recombinant human LTA?

For carrier-free formulations (211-TBB/CF), reconstitute the lyophilized protein at 100 μg/mL in PBS. For formulations containing carrier protein (211-TBB), reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin. After reconstitution, aliquot the protein to avoid repeated freeze-thaw cycles that can compromise activity. Store reconstituted protein at -20°C to -80°C, and use a manual defrost freezer to maintain protein integrity .

How should I determine the appropriate dose of LTA for in vitro cytotoxicity assays?

The effective dose for LTA-induced cytotoxicity varies by application and cell type. For the L-929 mouse fibroblast cell line, the ED50 for cytotoxicity in the presence of actinomycin D is 4-20 pg/mL for the newer formulation (211-TBB) and 0.1-0.4 ng/mL for the older formulation (211-TB). When designing cytotoxicity assays, establish a dose-response curve (0.1 pg/mL to 10 ng/mL) to determine optimal concentrations for your specific cell system. Include actinomycin D (typically 1 μg/mL) as a metabolic inhibitor to enhance cytotoxic effects, and implement appropriate controls including TNF-alpha as a comparative standard .

What experimental controls should be included when studying LTA-mediated effects?

When designing experiments with LTA, include the following controls:

Control TypePurposeImplementation
Vehicle controlControls for buffer effectsInclude same volume of reconstitution buffer without LTA
Positive controlConfirms assay functionalityTNF-alpha at equivalent molar concentration
Neutralizing antibodyConfirms specificityPre-incubate LTA with anti-LTA antibody before adding to cells
Receptor antagonistIdentifies receptor involvementBlock individual receptors (TNF RI, TNF RII, HVEM) to determine pathway specificity
Heat-inactivated LTAControls for non-specific effectsHeat protein at 95°C for 10 minutes before use

These controls help differentiate specific LTA-mediated effects from non-specific or assay-related outcomes .

How can I design experiments to distinguish between LTA homotrimer and LTA/LT-beta heterotrimer signaling?

Distinguishing between signaling pathways activated by LTA homotrimers versus LTA/LT-beta heterotrimers requires careful experimental design. Use receptor-specific blocking antibodies to differentiate between pathways: LTA homotrimers signal through TNF RI/II and HVEM, while LTA/LT-beta heterotrimers signal through LT-beta R.

For advanced studies, implement the following approaches:

  • Use receptor-specific reporter cell lines expressing individual receptors

  • Employ CRISPR/Cas9 to knock out specific receptors in your cell system

  • Use recombinant soluble receptors as competitive inhibitors

  • Compare effects of recombinant LTA alone versus co-culture with LT-beta-expressing cells

Verification can be performed using receptor-specific phosphorylation assays and downstream signaling analysis by Western blot or phospho-flow cytometry .

What methodologies are optimal for studying LTA's role in lymphoid organ development?

Investigating LTA's role in lymphoid organ development requires integrating in vitro and in vivo approaches:

  • Organoid culture systems: Establish lymphoid organoids incorporating stromal cells, lymphocytes, and controlled LTA exposure.

  • 3D tissue engineering: Create artificial lymphoid structures with controlled gradients of LTA.

  • Conditional knockout models: Use Cre-lox systems for cell type-specific and temporally controlled LTA deletion.

  • Adoptive transfer experiments: Transfer LTA-sufficient or LTA-deficient lymphocytes to appropriate recipient models.

  • High-resolution imaging: Employ two-photon microscopy to visualize LTA-dependent cellular interactions in developing lymphoid tissues.

Analysis should incorporate comprehensive immune phenotyping, spatial transcriptomics, and functional evaluation of the developing structures .

How does human LTA cross-reactivity with mouse receptors impact translational research models?

  • Validate receptor binding: Confirm binding affinity of human LTA to mouse TNF-RI, TNF-RII, and LT-beta R using surface plasmon resonance or cellular binding assays.

  • Compare signaling potency: Perform parallel dose-response analyses in human and mouse cells to establish equivalent functional doses.

  • Account for species-specific differences: Some downstream pathways may show divergent responses due to species-specific adapter proteins or signaling intermediates.

  • Consider humanized models: For highly translational studies, use mouse models with humanized TNF receptors.

Quantify these differences by comparing ED50 values between species and adjust dosing accordingly to ensure translational relevance .

What strategies can resolve inconsistent results in LTA-induced cytotoxicity assays?

Inconsistent cytotoxicity results can stem from multiple factors:

  • Protein activity loss: Recombinant LTA can lose activity due to improper storage or handling. Ensure proper aliquoting to avoid freeze-thaw cycles and verify protein integrity by SDS-PAGE before critical experiments.

  • Cell line variability: L-929 and other reporter cell lines may change sensitivity over passages. Maintain low-passage cells and periodically verify sensitivity using standard curves.

  • Metabolic inhibitor effectiveness: The potency of actinomycin D is critical for observing cytotoxic effects. Ensure fresh preparation and appropriate concentration (typically 1 μg/mL).

  • Media components interference: Serum factors can neutralize LTA activity. Consider titrating serum concentration or using serum-free conditions during treatment periods.

  • Cell density effects: Cytotoxicity is influenced by cell density at treatment time. Standardize seeding density and treatment timepoint relative to plating.

Implementing a standard operating procedure with these considerations can significantly reduce variability in cytotoxicity assays .

What analytical methods should be used to confirm the integrity and activity of recombinant human LTA?

Multiple complementary methods should be employed to confirm LTA integrity:

  • Structural analysis:

    • SDS-PAGE under reducing conditions (should show a band at 19 kDa)

    • Size exclusion chromatography to confirm trimeric assembly

    • Circular dichroism spectroscopy to verify secondary structure

  • Functional assessment:

    • L-929 cytotoxicity assay with actinomycin D (ED50 4-20 pg/mL)

    • Reporter cell lines expressing TNF receptors coupled to luciferase or other reporters

    • Binding assays using surface plasmon resonance with purified receptors

  • Immunological confirmation:

    • Western blot with specific anti-LTA antibodies

    • ELISA to quantify protein concentration

For critical experiments, performing both structural and functional assessments is recommended to ensure reproducible results .

How can researchers distinguish between direct LTA effects and secondary cytokine responses?

Distinguishing primary LTA effects from secondary cytokine responses requires temporal and mechanistic controls:

  • Temporal analysis: Perform detailed time-course studies capturing both early (minutes to hours) and late (hours to days) responses.

  • Transcriptional inhibition: Use actinomycin D at sub-cytotoxic doses or other transcriptional inhibitors to block secondary cytokine production.

  • Translational blockade: Apply cycloheximide to inhibit protein synthesis and prevent secondary cytokine effects.

  • Receptor occupancy analysis: Use competitive binding assays with labeled LTA to determine receptor occupancy kinetics.

  • Cytokine neutralization: Implement comprehensive cytokine neutralization using antibody cocktails to eliminate secondary effects.

  • Single-cell analysis: Employ single-cell transcriptomics or proteomics to identify directly responding cells versus those responding to secondary signals.

These approaches can be combined for robust differentiation between direct LTA signaling and downstream cytokine cascades .

How can multiparametric analysis be optimized to study LTA-induced inflammatory pathways?

Optimizing multiparametric analysis requires integration of complementary technologies:

  • Phospho-flow cytometry: Simultaneously evaluate multiple phosphorylation events (p38 MAPK, JNK, ERK, NF-κB) in heterogeneous cell populations following LTA stimulation.

  • Multiplex cytokine analysis: Employ bead-based multiplex assays to quantify up to 50 cytokines simultaneously from culture supernatants.

  • Transcriptomic profiling: Use RNA-seq or targeted NanoString panels to identify transcriptional networks activated by LTA.

  • Pathway inhibitor panels: Systematically apply specific kinase inhibitors in a matrix format to delineate signaling dependencies.

  • CRISPR screens: Implement focused CRISPR libraries targeting inflammatory pathway components to identify essential mediators of LTA responses.

Data integration across these platforms using computational approaches such as principal component analysis or pathway enrichment can reveal the multidimensional nature of LTA-induced inflammatory responses .

What methodologies enable investigation of LTA's role in TNF superfamily cross-talk?

Investigating cross-talk between LTA and other TNF superfamily members requires specialized approaches:

  • Sequential and simultaneous stimulation protocols: Apply LTA before, simultaneously with, or after other TNF family ligands to identify synergistic, additive, or antagonistic effects.

  • Receptor complex immunoprecipitation: Use proximity ligation assays or co-immunoprecipitation to identify receptor complex formation and composition.

  • FRET/BRET analysis: Employ fluorescence or bioluminescence resonance energy transfer to study receptor clustering and heterotypic interactions.

  • Competitive binding assays: Quantify displacement of labeled ligands to determine binding competition at shared receptors.

  • Chimeric receptors: Engineer domain-swapped TNF receptors to identify regions mediating cross-talk.

  • Bispecific reagents: Develop bispecific antibodies or fusion proteins targeting multiple TNF receptors to modulate cross-talk experimentally.

These approaches can reveal how LTA signaling is influenced by and influences other TNF superfamily members in complex immune responses .

How can researchers accurately model the role of LTA in autoimmune disease pathogenesis?

Modeling LTA's role in autoimmune pathogenesis requires multilevel approaches:

  • Patient-derived materials: Analyze LTA expression and polymorphisms in samples from autoimmune disease patients, correlating with clinical parameters.

  • Humanized mouse models: Develop models with human immune system components to better reflect human LTA biology.

  • Tissue-specific conditional expression: Use inducible promoters to express LTA in relevant tissues mimicking autoimmune conditions.

  • Ex vivo tissue models: Culture affected tissues (synovium for rheumatoid arthritis, CNS for multiple sclerosis) with controlled LTA exposure.

  • Systems biology integration: Combine genomic, transcriptomic, and proteomic data to create predictive models of LTA-dependent disease progression.

  • Therapeutic intervention models: Test LTA-pathway inhibitors at different disease stages to identify optimal intervention points.

These approaches can help define LTA's precise contributions to specific autoimmune conditions and identify targeted therapeutic strategies .

How should researchers interpret differences in potency between older (211-TB) and newer (211-TBB) recombinant human LTA formulations?

The significant potency difference between older (211-TB, ED50: 0.1-0.4 ng/mL) and newer (211-TBB, ED50: 4-20 pg/mL) formulations represents an approximately 25-fold increase in specific activity. This difference should be interpreted considering:

  • Protein folding and trimeric assembly: The newer formulation likely maintains more native trimeric structure.

  • Experimental design implications: Protocols developed for the older formulation require dose adjustment when transitioning to the newer product.

  • Historical data comparison: When comparing new results with published literature using older formulations, normalize to biological activity rather than protein mass.

  • Receptor binding kinetics: Perform comparative receptor binding assays to determine if higher potency correlates with increased receptor affinity.

  • Data reproducibility: Higher specific activity may improve signal-to-noise ratio and data consistency in functional assays.

When publishing results, always specify the exact formulation used and include standardized bioactivity measurements to facilitate cross-study comparisons .

What analytical frameworks help distinguish between LTA's direct cytotoxic effects and its immunomodulatory functions?

Distinguishing between direct cytotoxicity and immunomodulation requires systematic analytical frameworks:

  • Cell type-specific response patterns:

Cell TypeDirect CytotoxicityImmunomodulation
L-929 fibroblastsPrimary endpoint (with actinomycin D)Minimal
Primary T cellsMinimalCytokine production, proliferation
Dendritic cellsResistantMaturation, cytokine secretion
NK cellsVariableEnhanced cytotoxic function
  • Temporal discrimination: Direct cytotoxicity typically manifests within 6-24 hours, while immunomodulatory effects may develop over days.

  • Concentration-dependent effects: Plot complete dose-response curves, as cytotoxic effects often require higher concentrations than immunomodulatory functions.

  • Pathway-specific inhibitors: Apply selective inhibitors of death receptor signaling (caspase inhibitors) versus immunomodulatory pathways (NF-κB inhibitors).

  • Genetic manipulation: Compare effects in cells with genetic deficiencies in apoptotic versus immunomodulatory signaling components.

This analytical framework provides a structured approach to differentiate LTA's diverse functional effects across experimental systems .

How can researchers effectively compare and integrate LTA data across different experimental models and species?

Cross-model and cross-species data integration requires standardized frameworks:

  • Activity normalization: Convert all doses to biological activity units rather than mass concentrations to account for potency differences.

  • Receptor expression profiling: Quantify TNF receptor expression levels across models to normalize for receptor availability.

  • Pathway activation mapping: Develop standardized readouts of key signaling nodes (e.g., NF-κB, MAPK, caspase activation) across models.

  • Temporal alignment: Adjust timepoints based on model-specific kinetics of receptor expression and signaling.

  • Comparative transcriptomics: Identify conserved gene modules responsive to LTA across species and models.

  • Meta-analysis frameworks: Apply statistical methods designed for heterogeneous data integration with appropriate weighting of model relevance.

  • Visualization tools: Implement dimensionality reduction techniques like PCA or t-SNE to visualize relationships between datasets from different models.

These approaches facilitate meaningful comparisons across diverse experimental systems while acknowledging model-specific contexts and constraints .

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.