CD8B Human

CD8B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of CD8B

CD8B is a type I transmembrane glycoprotein encoded by the CD8B gene located on human chromosome 2 (2p12). It shares structural homology with CD8α but differs in key functional domains:

DomainFeatures
ExtracellularContains an immunoglobulin variable (IgV)-like domain (residues 23–170) that binds MHC-I α3 region .
TransmembraneHelix (residues 171–191) with cysteine residues critical for dimerization .
CytoplasmicShort tail (residues 192–210) lacking Lck-binding motifs present in CD8α .

CD8B primarily forms heterodimers with CD8α (CD8αβ) but can also exist as homodimers (CD8ββ) in humans, unlike murine CD8β, which requires CD8α for surface expression .

Immune Synapse Formation

CD8αβ stabilizes TCR-MHC-I interactions, increasing binding affinity by 10- to 100-fold . This co-receptor recruits the tyrosine kinase Lck via CD8α, enabling phosphorylation of CD3 and ζ-chain immunoreceptor tyrosine-based activation motifs (ITAMs) .

Effector Functions

  • Cytotoxicity: Enhances CTL-mediated lysis of infected or cancerous cells .

  • Memory T-Cell Survival: CD8αβ expression correlates with long-lived memory T-cell populations .

  • Regulatory Roles: CD8β isoforms modulate T-cell activation thresholds and downregulation post-stimulation .

Isoforms and Splice Variants

Human CD8B undergoes alternative splicing, producing four isoforms (M1–M4) with distinct cytoplasmic tails:

IsoformExpressionKey Features
M1Naïve T cellsEquivalent to murine CD8β; lacks dihydrophobic motifs .
M4Effector memory T cellsContains a 36-amino-acid cytoplasmic tail with a leucine-based internalization motif .

The M4 isoform’s unique tail regulates surface expression and internalization, impacting T-cell activation dynamics .

Autoimmunity and Inflammation

  • Reduced CD4+CD8A+ T cells (co-expressing CD8B mRNA) are observed in inflamed intestinal tissue of Crohn’s disease patients .

  • CD8+ regulatory T cells (Tregs) expressing KIR or Foxp3 suppress autoimmune responses and graft-versus-host disease (GVHD) .

Therapeutic Targets

  • CAR-T Cells: CD8β incorporation improves persistence and cytotoxicity in chimeric antigen receptor T-cell therapies .

  • Checkpoint Blockade: Anti-CD45RC antibodies deplete effector T cells while sparing CD8+ Tregs, promoting transplant tolerance .

Cell Lines

The CD8α/CD8B Jurkat cell line (BPS Bioscience #78917) co-expresses both subunits, enabling studies on TCR signaling and cytotoxicity .

Antibodies

  • BV421 Anti-Human CD8b (BD Biosciences #568373): Used for flow cytometry to distinguish CD8β+ T-cell subsets .

  • 5F2 Antibody: Detects CD8β homodimers independent of CD8α .

Key Research Findings

  • Species-Specific Dimerization: Human CD8ββ homodimers bind MHC-I less efficiently than CD8αβ, suggesting a regulatory role in T-cell activation .

  • Palmitoylation: Post-translational lipid modification targets CD8β to lipid rafts, optimizing Lck recruitment .

  • Splice Variants: The M4 isoform’s internalization motif fine-tunes T-cell responses during chronic infections .

Product Specs

Introduction
The CD8 antigen's beta chain isoforms are encoded by the CD8B gene. The majority of cytotoxic T lymphocytes express CD8, a cell surface glycoprotein that makes it easier for cells to interact effectively within the immune system. The T-cell receptor on the T lymphocyte and the co-receptor CD8 antigen work together to identify antigens that an antigen-presenting cell (APC) has exposed in the presence of class I MHC molecules. A heterodimer made up of one alpha and one beta chain or a homodimer made up of two alpha chains can act as the functional co-receptor. Immunoglobulin variable light chains share significant homology with both alpha and beta chains.
Description
Recombinant CD8B Human is a single, non-glycosylated polypeptide chain produced in E. coli. It contains 172 amino acids (22-170), has a molecular mass of 19.2 kDa, and is fused to a 23 amino acid His-tag at its N-terminus.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The CD8B solution (0.5 mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
If the entire vial will be used within 2-4 weeks, store at 4°C. For longer storage periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Avoid multiple freeze-thaw cycles.
Purity
Greater than 80% as determined by SDS-PAGE.
Synonyms
T Lymphocyte Surface Glycoprotein Beta Chain, CD8 Antigen, Beta Polypeptide 1 (P37), CD8b Molecule, CD8b Antigen, T-Cell Surface Glycoprotein CD8 Beta Chain, CD8B1, LYT3, LY3, LEU2, P37.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSLQQTPAY IKVQTNKMVM LSCEAKISLS NMRIYWLRQR QAPSSDSHHE FLALWDSAKG TIHGEEVEQE KIAVFRDASR FILNLTSVKP EDSGIYFCMI VGSPELTFGK GTQLSVVDFL PTTAQPTKKS TLKKRVCRLP RPETQKGPLC SP

Q&A

What is CD8B and how does it function in human T cell biology?

CD8B (CD8 beta chain) is a critical component of the CD8 co-receptor found on cytotoxic T lymphocytes. It typically partners with CD8A (CD8 alpha chain) to form CD8αβ heterodimers, though CD8αα homodimers can also exist. The CD8αβ heterodimer plays an essential role in T cell receptor (TCR) recognition of peptide-MHC class I complexes, enhancing T cell sensitivity to antigens and subsequent activation.

Methodologically, researchers identify CD8B-expressing cells through flow cytometry using fluorescently-labeled antibodies specific to the CD8B protein, often in combination with CD8A detection to distinguish between cells expressing CD8αα homodimers versus CD8αβ heterodimers. Single-cell RNA sequencing approaches now enable the identification of CD8B expression at the transcript level, revealing previously unappreciated heterogeneity within CD8+ T cell populations .

How do tissue-resident memory CD8 T cells maintain CD8B expression?

Tissue-resident memory CD8 T cells (TRM) in barrier sites like the small intestine maintain CD8B expression through microenvironmental signals that regulate their transcriptional program. Research indicates that TRM cells occupy distinct spatial niches that influence their phenotype and function over time.

To study this maintenance mechanism, researchers employ spatial transcriptomic profiling techniques like Xenium technology, which can simultaneously detect the expression of hundreds of genes while preserving tissue architecture. This approach has revealed that CD8 T cells dynamically respond to tissue-derived signals that sustain their resident phenotype, including continued expression of CD8B . The methodology involves Xenium processing followed by immunohistochemical detection of CD8α and other markers, combined with haematoxylin and eosin (H&E) staining to assess tissue structures .

What methods accurately distinguish CD8B+ populations in human tissue samples?

Accurately distinguishing CD8B+ T cell populations in human tissues requires multiple complementary approaches:

  • Flow cytometry and cell sorting: Using antibodies against CD8A and CD8B to differentiate CD8αα+ from CD8αβ+ cells

  • Single-cell RNA sequencing: Detecting CD8B transcripts at the single-cell level to identify expression patterns and correlations with other genes

  • Spatial transcriptomics: Visualizing CD8B expression in situ while preserving tissue architecture

  • Multiplex immunohistochemistry/immunofluorescence: Simultaneously detecting CD8B with other markers to characterize cell phenotypes in tissue sections

Recent advances in spatial transcriptomics have been particularly valuable, as demonstrated by studies using Xenium technology with a custom multi-gene panel that enables detection of CD8B alongside hundreds of other relevant genes while maintaining spatial context . This methodology has revealed that CD8B expression patterns often correlate with distinct functional states and tissue locations.

What are the optimal strategies for integrating mouse and human CD8B data in comparative studies?

Integration of mouse and human CD8B data presents significant challenges due to species-specific differences in gene expression patterns and immune cell distributions. Researchers have developed several methodological approaches to address these challenges:

  • Fast gene set enrichment analysis (fGSEA): This computational approach identifies shared gene signatures between mouse and human CD8+ T cell populations, enabling cross-species functional comparisons despite differences in individual gene expression .

  • Single-cell integration algorithms: Tools that correct for batch effects and species-specific variability while preserving biologically meaningful differences.

  • Conserved transcriptional signature approach: Identifying evolutionarily conserved core gene sets that define functionally equivalent CD8+ T cell populations across species.

Research has shown that while there are important differences in CD8+ T cell populations between mice and humans (for example, MAIT cells are abundant in humans but rare in mice), integration of single-cell transcriptomic data can reveal functionally equivalent populations . The methodology involves performing parallel single-cell RNA-seq on both mouse and human intestinal samples, followed by computational integration using shared gene signatures .

How can spatial heterogeneity of CD8B+ T cells be quantitatively assessed in tissue specimens?

Quantitative assessment of CD8B+ T cell spatial heterogeneity requires sophisticated methodological approaches:

  • Dual-coordinate-axis system: This approach maps individual CD8+ T cells based on their proximity to the nearest epithelial cell and distance to the base of the muscularis, creating two-dimensional density representations called immune allocation plots (IMAPs) .

  • Spatial transcriptomics: Technologies like Xenium enable simultaneous detection of CD8B expression and hundreds of other genes while preserving spatial information .

  • Computational spatial statistics: Methods including nearest neighbor analysis, quadrat analysis, and spatial autocorrelation to quantify clustering patterns and association with tissue structures.

These approaches have revealed that CD8+ T cells (including those expressing CD8B) dynamically occupy different regions in tissues like the small intestine, with distinct populations forming along spatial axes such as the crypt-villus axis . The methodology involves tissue collection at different timepoints, immunohistochemical staining, spatial transcriptomic profiling, and computational analysis to generate quantitative spatial metrics .

What are the current technical limitations in single-cell analysis of CD8B expression?

Single-cell analysis of CD8B expression faces several technical challenges that researchers must address through methodological innovations:

  • mRNA detection sensitivity: CD8B transcript levels may be low in some populations, requiring high-sensitivity methods and appropriate normalization strategies.

  • Protein-transcript correlation: Discrepancies between CD8B mRNA and protein levels necessitate integrated approaches combining transcriptomics with protein detection methods.

  • Spatial context preservation: Traditional single-cell methods lose spatial information, requiring special preservation techniques or computational inference.

  • Sample dissociation bias: Enzymatic tissue digestion can alter surface protein expression or preferentially recover certain cell populations.

  • Data integration challenges: Combining datasets from different platforms, tissues, or disease states requires sophisticated computational methods to control batch effects while preserving biological variation.

Researchers address these limitations through methodological approaches including targeted gene panels in spatial transcriptomics (e.g., the 350-gene panel with CD8B detection described in ), combined protein and RNA detection methods, and computational integration strategies .

How does CD8B expression correlate with cytotoxic function in human T cells?

CD8B expression shows complex relationships with cytotoxic functionality in human T cells:

  • Cytotoxic gene correlation: Single-cell transcriptomic analysis reveals that CD8B expression often correlates with cytotoxic genes including GZMA (Granzyme A), GZMB (Granzyme B), and other effector molecules .

  • Functional heterogeneity: Despite this correlation, not all CD8B+ cells exhibit the same cytotoxic potential. Spatial transcriptomics has identified distinct subpopulations with varying levels of cytotoxic gene expression .

  • Microenvironmental influence: The cytotoxic function of CD8B+ cells appears to be modulated by tissue microenvironments, with more differentiated cells (high GZMA, GZMB, and ITGAE expression) found at the top of intestinal villi and progenitor-like cells (high TCF7 and SLAMF6 expression) around the crypt area .

Methodological approaches to study this correlation include cytotoxicity assays combined with CD8B phenotyping, transcriptional profiling of sorted CD8B+ populations, and spatial transcriptomics to correlate CD8B expression with cytotoxic gene expression patterns in situ .

What role does CD8B play in tissue-resident memory T cell formation and maintenance?

CD8B contributes to tissue-resident memory T cell (TRM) formation and maintenance through several mechanisms:

  • TCR signal modulation: CD8B enhances TCR signaling sensitivity, potentially influencing the initial priming events that determine TRM fate.

  • Spatial positioning: CD8B+ TRM cells show distinct spatial distribution patterns in tissues like the small intestine, occupying specific niches that influence their differentiation trajectory .

  • Differentiation state maintenance: CD8B expression correlates with TRM differentiation states, with spatial transcriptomics revealing two polarized states - a more differentiated state at the top of intestinal villi (high GZMA, GZMB, and ITGAE expression) and a progenitor-like state around the crypt area (high TCF7 and SLAMF6 expression) .

Methodologically, this has been studied using adoptive transfer experiments of traceable CD8+ T cells followed by spatial transcriptomic analysis at different timepoints post-infection . These approaches have revealed that niche-dependent signals contribute to differentiating incoming CD8+ T cells and maintaining polarized TRM states over time .

How does CD8B expression change in inflammatory bowel disease contexts?

CD8B expression patterns undergo significant alterations in inflammatory bowel disease (IBD), providing insights into disease pathophysiology:

  • Altered distribution: The proportion of certain CD8+ T cell populations (including those expressing CD8B) differs between inflamed and non-inflamed tissues in patients with Crohn's disease, with reduced CD27-CD4+CD8A+ T cells in inflamed tissues compared to non-inflamed regions from the same patients .

  • Transcriptional changes: Single-cell transcriptomics of intestinal samples from IBD patients reveals altered expression patterns of CD8B and associated genes compared to healthy controls .

  • Functional implications: These changes may reflect alterations in the tissue's immune regulatory capacity, as certain CD8B+ populations may have immunomodulatory functions.

Methodologically, these changes are studied through single-cell RNA sequencing of intestinal samples from IBD patients, differential expression analysis to identify disease-associated gene signatures, and correlation with clinical parameters . Researchers have used markers like CD27 to divide T cell populations and compared their abundance between inflamed and non-inflamed tissues .

What methodological approaches best capture CD8B+ T cell dynamics in disease progression?

Capturing CD8B+ T cell dynamics during disease progression requires integrated methodological approaches:

  • Longitudinal sampling: Serial biopsies or blood draws from patients at different disease stages, with consistent processing protocols to minimize technical variation.

  • Multi-modal analysis: Combining flow cytometry, transcriptomics, and functional assays to correlate CD8B expression with functional states and disease metrics.

  • Spatial analysis: Using technologies like Xenium spatial transcriptomics to map CD8B+ T cell distributions in relation to disease features like inflammatory foci or tissue damage .

  • Computational trajectory analysis: Applying pseudotime and RNA velocity methods to single-cell data to infer developmental relationships and state transitions of CD8B+ populations during disease progression.

  • Integration with clinical data: Correlating CD8B+ T cell features with clinical parameters, treatment responses, and disease outcomes.

These approaches have been applied in IBD research, revealing that CD8B+ T cell dynamics correlate with disease features and may have prognostic or therapeutic implications . The methodology involves careful patient stratification, standardized tissue processing, and integrated multi-omic analysis .

How do chemokine gradients influence CD8B+ T cell trafficking and retention in tissues?

Chemokine gradients play a crucial role in orchestrating CD8B+ T cell trafficking and tissue retention:

  • CXCL9/CXCL10 gradients: Research has shown that these chemokines create spatial gradients that guide CXCR3-expressing CD8+ T cells (which may include CD8B+ populations) to specific tissue locations .

  • Spatial organization: In the small intestine, CXCL9 and CXCL10 are enriched in the top half of the lamina propria during homeostasis but are induced in complement fibroblasts at the bottom of the muscularis after infection, creating a second potential attraction point for CXCR3-expressing CD8+ T cells .

  • Experimental manipulation: CRISPR-Cas9 approaches to induce CXCR3 deletion in CD8+ T cells have been used to test the role of these gradients in CD8+ T cell localization and differentiation .

Methodologically, these processes are studied using spatial transcriptomics to visualize chemokine expression patterns, adoptive transfer of genetically modified T cells, and dual-coordinate mapping of cell positions relative to tissue landmarks . These approaches have revealed the dynamic nature of chemokine-guided CD8+ T cell positioning and its impact on functional specialization.

What computational frameworks best integrate CD8B expression data with spatial and functional information?

Advanced computational frameworks for integrating CD8B expression with spatial and functional data include:

  • Immune allocation plots (IMAPs): Two-dimensional density representations that capture the distribution dynamics of CD8+ T cells in tissue structures like intestinal villi, revealing spatial separation of populations along anatomical axes .

  • Integrated multi-modal analysis: Frameworks that combine transcriptomic, proteomic, and spatial data to generate comprehensive views of CD8B+ T cell states and functions.

  • Spatial statistics and neighborhood analysis: Methods that quantify cell-cell interactions and spatial relationships between CD8B+ cells and other tissue components.

  • Trajectory inference with spatial constraints: Algorithms that incorporate spatial information to improve inference of developmental relationships between CD8B+ T cell states.

  • Graph-based representation learning: Approaches that model complex relationships between spatial location, gene expression, and functional states.

These computational approaches have been applied to datasets generated through spatial transcriptomics technologies like Xenium, which can simultaneously detect hundreds of genes in tissue sections while preserving spatial information . The methodology involves implementing dual-coordinate-axis systems based on cell proximity to tissue landmarks, generating spatial visualizations, and applying machine learning algorithms to identify patterns .

Product Science Overview

Structure and Function

CD8B is a member of the immunoglobulin superfamily and contains one Ig-like V-type (immunoglobulin-like) domain . The CD8 molecule can exist as either a homodimer composed of two alpha chains or a heterodimer composed of one alpha and one beta chain . Both alpha and beta chains share significant homology to immunoglobulin variable light chains .

The CD8 molecule acts as a co-receptor for the T-cell receptor (TCR) and is involved in the activation of T cells. It interacts with MHC class I molecules on APCs, facilitating the recognition of antigens by the TCR . This interaction is crucial for the immune response against intracellular pathogens, such as viruses, and for the elimination of cancerous cells .

Recombinant CD8B

Recombinant CD8B refers to the CD8 beta protein that has been produced using recombinant DNA technology. This involves inserting the gene encoding CD8B into an expression system, such as bacteria or mammalian cells, to produce the protein in large quantities . Recombinant CD8B is often used in research to study the function of the CD8 molecule and its role in the immune response .

Applications

Recombinant CD8B has several applications in biomedical research and clinical diagnostics. It is used in flow cytometry to identify and isolate CD8+ T cells from blood samples . It is also used in functional assays to study T cell activation and cytotoxicity . Additionally, recombinant CD8B can be used to generate antibodies against the CD8 molecule, which are valuable tools for immunological research .

Clinical Relevance

The CD8 molecule, including its beta subunit, is of significant clinical interest due to its role in immune responses. Alterations in CD8 expression or function can lead to immune deficiencies or contribute to the pathogenesis of various diseases . For example, CD8+ T cells are critical in controlling viral infections and tumor surveillance. Therefore, understanding the biology of CD8B and its interactions is essential for developing immunotherapies and vaccines .

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.