TNFRSF8 Mouse mediates immune responses through:
Immune Cell Regulation: Expressed on antigen-stimulated Th cells and B cells .
Th2 Bias: Costimulates Th2 cytokine secretion (e.g., IL-13) and suppresses Th1 responses .
Apoptosis Regulation: Promotes thymic negative selection of CD4+CD8+ T cells .
Ligand Interaction: Binds CD30 Ligand (TNFSF8), triggering NF-κB activation via TRAF2/5 .
NF-κB Activation: Drives proliferation and cytokine production in B cells .
Soluble CD30 (sCD30): Shed via TACE-mediated cleavage; inhibits membrane-bound CD30 signaling .
Studies using TNFRSF8 Mouse knockout models reveal conflicting roles:
Lymphomas: Overexpressed in Hodgkin’s disease (Reed-Sternberg cells) and anaplastic large cell lymphoma .
Autoimmunity: Linked to chronic inflammation and autoimmune disorders .
Brentuximab Vedotin: FDA-approved antibody-drug conjugate targeting CD30, effective in relapsed lymphomas .
MAB8521 (R&D Systems): Used in Luminex assays to detect plasma cytokines post-irradiation .
AF852 (R&D Systems): Detects CD30 in mouse splenocytes via fluorescent ICC .
Application | Target | Result/Use Case | Source |
---|---|---|---|
Luminex Development | Plasma cytokines | Post-irradiation cytokine profiling in mice | |
IHC-P (Whole Tissue) | CD30 in splenocytes | Localization in immune cells |
Tumor Necrosis Factor Receptor Superfamily, Member 8, Lymphocyte Activation Antigen CD30, CD30L Receptor, Ki-1 Antigen, D1S166E, CD30, Cytokine Receptor CD30, CD30 Antigen, Ki-1
Sf9, Baculovirus cells.
FPTDRPLKTT CAGDLSHYPG EAARNCCYQC PSGLSPTQPC PRGPAHCRKQ CAPDYYVNED GKCTACVTCL PGLVEKAPCS GNSPRICECQ PGMHCCTPAV NSCARCKLHC SGEEVVKSPG TAKKDTICEL PSSGSGPNCS NPGDRKTLTS HATPQAMPTL ESPANDSARS LLPMRVTNLV QEDATELVKV PESSSSKARE PSPDPGNAEK NMTLELPSPG TLPDISTSEN SKEPASTAST LEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGKHHHHHH
Mouse CD30 (TNFRSF8) is a 120 kDa type I transmembrane glycoprotein belonging to the tumor necrosis factor receptor superfamily. The mature mouse CD30 protein consists of three distinct domains: a 264 amino acid extracellular domain (ECD) containing three cysteine-rich repeats, a 27 amino acid transmembrane segment, and a 190 amino acid cytoplasmic domain. Unlike mouse CD30, human CD30 contains an additional 90 amino acids in the ECD and features six cysteine-rich repeats instead of three. Within shared regions of the ECD, mouse CD30 exhibits 53% amino acid sequence identity with human CD30 and 80% with rat CD30 .
When designing cross-species experiments, these structural differences must be considered, particularly for antibody selection, as epitope recognition may vary significantly between species. Researchers should validate antibody cross-reactivity before conducting comparative studies.
Under physiological conditions, CD30/TNFRSF8 expression is predominantly restricted to activated T and B lymphocytes. It is not typically expressed on resting lymphocytes. Expression is induced following antigen stimulation of both Th cells and B cells . In experimental settings, CD30 expression can be detected in mouse splenocytes using immunofluorescence techniques with specific anti-CD30 antibodies .
For tissue-specific expression studies, researchers should consider using multiple detection methods including flow cytometry, immunohistochemistry, and mRNA analysis to comprehensively map expression patterns across different activation states and tissue microenvironments.
Mouse CD30/TNFRSF8 primarily exerts its effects through engaging with its ligand, CD30L/TNFSF8, which is expressed on activated Th cells, monocytes, granulocytes, and medullary thymic epithelial cells. This interaction triggers several important functional outcomes:
Costimulation of antigen-induced Th0 and Th2 proliferation and cytokine secretion
Promotion of Th2-biased immune responses
Induction of IL-13 expression in T cells, even in the absence of antigenic stimulation
Contribution to thymic negative selection through apoptotic cell death of CD4+CD8+ T cells
For investigating these signaling pathways, researchers should employ techniques such as western blotting for downstream phosphorylation events, gene expression analysis for cytokine production, and cell viability assays to assess apoptotic outcomes.
Several validated methodologies exist for detecting mouse CD30/TNFRSF8:
ELISA: Sandwich ELISA using specific capture and detection antibodies allows quantification of soluble CD30 in serum, plasma, and cell culture media. The typical detection range for commercial ELISA kits is 19.5-1250 pg/mL .
Immunofluorescence: Detection of CD30 on cellular surfaces can be performed using fluorochrome-conjugated antibodies. For example, CD30 has been successfully detected in immersion-fixed mouse splenocytes using goat anti-mouse CD30/TNFRSF8 antibodies followed by fluorescent secondary antibodies .
Flow Cytometry: For quantitative analysis of CD30 expression on specific cell populations, flow cytometry using validated anti-CD30 antibodies is recommended.
When selecting a detection method, researchers should consider the specific experimental question, sample type, and required sensitivity. For low-abundance samples, more sensitive techniques such as ELISA may be preferable over immunohistochemistry.
Proper storage and handling of CD30/TNFRSF8 research reagents is crucial for experimental success:
Recombinant Proteins:
Antibodies:
Use a manual defrost freezer and avoid repeated freeze-thaw cycles
Store at -20°C to -70°C for up to 12 months from receipt date (as supplied)
After reconstitution, store at 2-8°C under sterile conditions for up to 1 month
For longer storage after reconstitution, maintain at -20°C to -70°C under sterile conditions for up to 6 months
Researchers should always follow manufacturer-specific instructions as optimal storage conditions may vary between product formulations.
Robust experimental design for CD30/TNFRSF8 analysis should include several types of controls:
Positive Controls:
Activated mouse T cells known to express CD30
Cell lines transfected with mouse CD30
Recombinant mouse CD30 protein (for ELISA)
Negative Controls:
Resting lymphocytes that do not express CD30
Samples from CD30 knockout mice
Isotype control antibodies to assess non-specific binding
Technical Controls:
Standard curves using recombinant proteins of known concentration
Internal reference standards to normalize across experimental batches
Dilution linearity tests to confirm detection within the quantitative range
Each experimental method requires specific controls. For example, ELISA experiments should include blank wells and standard curves, while flow cytometry should include fluorescence-minus-one (FMO) controls and isotype controls.
While mouse CD30/TNFRSF8 shares functional similarities with human CD30, several important differences must be considered in translational research:
Structural Differences: Mouse CD30 contains three cysteine-rich repeats in its ECD compared to six in human CD30, and shares only 53% sequence identity in common regions . These differences may affect ligand binding properties and downstream signaling dynamics.
Expression Patterns: Although both mouse and human CD30 are expressed on activated lymphocytes and upregulated in certain pathological conditions, tissue-specific and temporal expression patterns may differ.
Signaling Outcomes: While both mouse and human CD30 promote Th2-biased responses, the magnitude and specific cytokine profiles induced may vary between species.
Distinguishing between membrane-bound and soluble CD30 requires specific methodological approaches:
For Membrane-bound CD30:
Flow cytometry using anti-CD30 antibodies on intact cells
Immunofluorescence microscopy of non-permeabilized cells
Cell surface biotinylation followed by immunoprecipitation
For Soluble CD30:
ELISA-based detection in cell-free supernatants, serum, or plasma
Western blotting of concentrated supernatants using antibodies against the extracellular domain
Size-exclusion chromatography to separate soluble CD30 from membrane-bound forms in vesicles
When studying the relationship between these forms, researchers can employ techniques such as inhibitors of metalloproteinases to block shedding, or stimulate shedding with phorbol esters to assess the dynamics of conversion from membrane-bound to soluble forms.
Several experimental approaches can be used to manipulate CD30 signaling:
Genetic Approaches:
CD30 knockout mice to study loss-of-function effects
Conditional knockout models using Cre-loxP systems for tissue-specific deletion
Transgenic overexpression models to study gain-of-function effects
Pharmacological Approaches:
Agonistic anti-CD30 antibodies to stimulate signaling
Blocking antibodies to inhibit CD30-CD30L interactions
Recombinant soluble CD30 as a competitive inhibitor
Small molecule inhibitors of downstream signaling components
Cellular Approaches:
Adoptive transfer of CD30-deficient or CD30-overexpressing cells
Ex vivo stimulation of CD30 before cell transfer
Co-culture systems with CD30L-expressing cells
These approaches can be combined with readouts such as cytokine production, proliferation assays, and in vivo disease models to comprehensively assess CD30's role in immune regulation.
When studying CD30/TNFRSF8 in disease models, several factors should be considered:
Model Selection:
For lymphoma studies, choose models that naturally express CD30 (e.g., certain ALK-positive anaplastic large cell lymphoma models)
For autoimmunity, consider models where Th2 responses play a significant role
Timing Considerations:
CD30 expression fluctuates during disease progression
Temporal analysis at multiple time points is crucial
Consider early intervention versus treatment of established disease
Readouts:
Multidimensional assessment including histopathology, flow cytometry, serum biomarkers
Functional tests relevant to the specific disease
Analysis of both CD30+ cell populations and effects on bystander cells
Controls:
Include CD30-deficient mice on the same background
Use isotype controls for antibody treatments
Consider sex-specific effects as immune responses may differ
Researchers frequently encounter several challenges when studying mouse CD30/TNFRSF8:
Low Expression Levels:
Solution: Use appropriate stimulation protocols (e.g., ConA, anti-CD3/CD28) to upregulate CD30 expression
Employ signal amplification techniques such as tyramine signal amplification for immunohistochemistry
Consider enrichment of CD30+ cells prior to analysis
Antibody Cross-Reactivity:
Solution: Validate antibody specificity using CD30 knockout tissues
Perform blocking experiments with recombinant CD30 protein
Use multiple antibody clones targeting different epitopes
Protein Degradation:
Solution: Add protease inhibitors to all buffers
Process samples quickly and maintain cold temperatures
For soluble CD30, stabilize with carrier proteins when appropriate
Glycosylation Variability:
Solution: Consider deglycosylation treatments for more consistent molecular weight analysis
Use multiple detection methods that are less affected by glycosylation
Include controls with known glycosylation patterns
Addressing these challenges requires careful optimization and validation of each experimental protocol for specific applications.
Detecting low-abundance CD30/TNFRSF8 requires specialized approaches:
Sample Preparation Optimization:
Fresh tissue preparation whenever possible
Optimal fixation protocols to preserve epitopes
Antigen retrieval optimization for fixed tissues
Signal Amplification Techniques:
Employ biotin-streptavidin amplification systems
Use tyramide signal amplification for immunohistochemistry
Consider rolling circle amplification for extremely low abundance targets
Detection Technology Selection:
Choose high-sensitivity ELISA kits with detection limits in the pg/mL range
Consider digital ELISA platforms for sub-pg/mL detection
Use spectral flow cytometry with bright fluorophores
Implement image cytometry for spatial context with high sensitivity
Enrichment Strategies:
Magnetic bead enrichment of CD30+ cells before analysis
Laser capture microdissection of regions with expected expression
Cell sorting of relevant populations followed by molecular analysis
Combinations of these approaches can significantly improve detection of low-abundance CD30 in complex tissue samples.
When encountering discrepancies between detection methods, consider these analytical approaches:
Method-Specific Biases:
Flow cytometry may detect only cell surface CD30, missing internalized protein
ELISA might detect soluble CD30 fragments not recognized by certain antibodies
Western blotting can be affected by protein denaturation altering epitope recognition
Reconciliation Strategies:
Use orthogonal methods targeting different epitopes or properties
Employ knockout controls to establish true background levels
Consider spike-in experiments with recombinant standards
Analyze correlation patterns rather than absolute values
Biological Interpretation:
Different forms of CD30 (membrane-bound, soluble, differentially glycosylated) may predominate in different contexts
Activation state may affect epitope accessibility
Technical variability must be distinguished from biological heterogeneity
Analyzing CD30/TNFRSF8 expression data requires appropriate statistical methods:
For Continuous Expression Data (e.g., ELISA, qPCR):
Parametric tests (t-test, ANOVA) if normality assumptions are met
Non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) for non-normal distributions
Consider log transformation for skewed distribution common in expression data
Use paired tests when comparing the same subjects under different conditions
For Categorical Data (e.g., percent positive cells):
Chi-square or Fisher's exact test for comparing proportions
McNemar's test for paired categorical data
For Complex Experimental Designs:
Mixed-effects models for repeated measures with missing data
ANCOVA when controlling for covariates
Multiple regression to assess relationships between CD30 and multiple factors
Multiple Testing Correction:
Bonferroni correction for strong control of family-wise error rate
Benjamini-Hochberg procedure for controlling false discovery rate
Consider the biological context when interpreting adjusted p-values
Importantly, power analysis should be conducted during experimental design to ensure sufficient sample sizes for detecting biologically meaningful differences in CD30 expression.
CD30L interacts with its receptor, CD30 (also known as TNFRSF8), which is a cell membrane protein of the TNF receptor superfamily . CD30 is expressed by activated, but not resting, T and B cells . The interaction between CD30L and CD30 plays a crucial role in regulating the proliferation of lymphocytes and may also be involved in human immunodeficiency virus (HIV) replication .
As a regulator of apoptosis, CD30 can induce cell death or proliferation depending on the cell type . It has been shown to limit the proliferative potential of autoreactive CD8 effector T cells, thereby protecting the body against autoimmunity . CD30 expression is upregulated in various hematological malignancies, including Reed-Sternberg cells in Hodgkin’s disease, anaplastic large cell lymphoma, and subsets of Non-Hodgkin’s lymphomas . Additionally, CD30 is linked to leukocytes in patients with chronic inflammatory diseases such as lupus erythematosus, asthma, rheumatoid arthritis, and atopic dermatitis .
Recombinant Mouse CD30 Ligand is produced using a mouse myeloma cell line, NS0-derived mouse CD30 Ligand/TNFSF8 protein . The recombinant protein is typically purified to a high degree, with a purity greater than 95% as determined by SDS-PAGE visualized with Silver Staining and quantitative densitometry by Coomassie® Blue Staining . The endotoxin level is kept below 0.10 EU per 1 μg of the protein by the LAL method .
The activity of the recombinant mouse CD30 Ligand is measured by its ability to stimulate IL-6 secretion by HDLM human Hodgkin’s lymphoma cells . The effective dose (ED50) for this effect is between 5-30 ng/mL in the presence of 10 µg/mL of a cross-linking antibody .
Recombinant Mouse CD30 Ligand is used in various research applications, including studies on cell signaling, apoptosis, and immune response . It is available in both carrier-free and carrier-containing formulations. The carrier-free version does not contain Bovine Serum Albumin (BSA), which can interfere with certain applications .
For optimal stability and storage, the lyophilized protein should be stored at -20 to -70 °C and reconstituted at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin . Once reconstituted, it should be stored under sterile conditions at 2 to 8 °C for up to one month or at -20 to -70 °C for up to three months .