CD8 Biotin Antibody

CD8, Rat Anti-Mouse, Biotin
Shipped with Ice Packs
In Stock

Description

CD8 Biotin Antibody: Key Properties

Anti-CD8 biotinylated antibodies are monoclonal or polyclonal reagents engineered for high specificity. Notable clones include RFT8, MEM-31, and RPA-T8, each validated for distinct applications .

CloneIsotypeApplicationsHost SpeciesConcentration
RFT8 Mouse IgG1κFlow cytometry, immunohistochemistry, immunocytochemistryMouse (BALB/c)Lot-specific
MEM-31 Mouse IgGFlow cytometry, T-cell activation studiesMouse0.5–1.0 mg/mL
RPA-T8 Mouse IgG1κCyTOF, IHC, in vitro assaysMouseVaries by lot

Critical Note: MEM-31 recognizes a conformation-dependent extracellular epitope and does not react with formaldehyde-fixed cells, limiting its use in Western blotting .

Activation vs. Inhibition of CD8+ T Cells

  • OKT8 Antibody: Induces effector functions (e.g., cytokine release, cytotoxicity) in CD8+ T cells independent of TCR engagement .

  • SK1 and C8/144B Clones: Fail to activate T cells but enhance TCR/pMHCI binding kinetics, improving tetramer staining .

  • CT-CD8a/b (Mouse): Activates CD8+ T cells, contrasting with human-specific clones like DK25 .

Antibody CloneEffect on T CellsApplications
OKT8 Activates effector functionsT-cell activation studies
SK1 Enhances TCR/pMHCI bindingTetramer staining optimization
MEM-31 Blocks conjugate formationT-cell suppression assays

Therapeutic Implications

  • Autoimmune Disease: Autoreactive CD8+ T cells are highly CD8-dependent, making blocking antibodies (e.g., MEM-31) potential tools for selective immunosuppression .

  • Cancer Diagnostics: Biotinylated CD8 antibodies are used in clinical settings for tumor-infiltrating lymphocyte analysis (e.g., crefmirlimab) .

Flow Cytometry

  • Surface Staining: Detects CD8+ T cells in peripheral blood, with sensitivity validated across clones like RPA-T8 and RFT8 .

  • Tetramer Enhancement: Anti-CD8 antibodies (e.g., SK1) increase pMHCI tetramer binding avidity by up to 10-fold .

Immunohistochemistry (IHC)

  • Frozen vs. Paraffin Sections: RFT8 and RPA-T8 show robust staining in frozen sections but require optimization for paraffin-embedded tissues .

Functional Assays

  • Cytotoxicity Studies: OKT8-induced effector function is quantified via chromium release assays .

  • Depletion Protocols: Used to isolate CD8+ populations in mixed lymphocyte cultures .

Limitations and Considerations

  • Species Reactivity: Most clones are human-specific; cross-reactivity with mouse CD8 requires validation .

  • Fixation Sensitivity: MEM-31 and RFT8 lose reactivity in formaldehyde-fixed samples .

  • Lot Variability: Concentration and performance may differ between production batches .

Product Specs

Introduction
CD8 is a transmembrane glycoprotein present on the surface of most cytotoxic T lymphocytes (CTLs). It plays a crucial role in the immune system by mediating cell-cell interactions. Functioning as a co-receptor with the T-cell receptor (TCR), CD8 recognizes antigens presented by antigen-presenting cells (APCs) in the context of major histocompatibility complex class I (MHC I) molecules. CD8 exists as either a homodimer of two alpha chains or a heterodimer consisting of one alpha and one beta chain. Both alpha and beta chains share structural similarities with immunoglobulin variable light chains. The primary function of CD8 is to assist in the identification and elimination of cells displaying foreign antigens, such as virus-infected cells or tumor cells.
Formulation
Supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) following reconstitution.
Storage Procedures
For long-term storage, maintain the lyophilized antibody at 4 degrees Celsius. Upon reconstitution, it is recommended to aliquot the antibody and store it at -20 degrees Celsius if not intended for use within one month.
Solubility
To reconstitute the lyophilized antibody, add deionized water (H2O) to the vial. Gently mix the solution, ensuring that the contents on the sides of the vial are dissolved. Allow 30-60 seconds for complete reconstitution before use.
Applications
This antibody is suitable for various applications, including fluorescence-activated cell sorting (FACS) staining, immunohistochemistry (IHC), and Western blotting (WB). For optimal staining results, a concentration of 10 microliters per 1,000,000 cells is recommended.
Available Conjugates
Currently, this antibody is offered in both unconjugated and fluorescein isothiocyanate (FITC)-conjugated forms.
Synonyms
CD8, MAL, p32.
Purification Method
Ion Exchange Column.
Type
Rat Anti Mouse Monoclonal.
Clone
NYRmCD8.
Immunogen
Purified Mouse LN T cells.
Ig Subclass
Rat IgM.

Q&A

What is CD8 Biotin Antibody and how does it function in research applications?

CD8 Biotin Antibody (such as the RPA-T8 clone) is a monoclonal antibody that specifically binds to the CD8 alpha (CD8α) protein, a type I transmembrane glycoprotein and member of the immunoglobulin superfamily. The antibody is conjugated with biotin, enabling secondary detection through streptavidin-conjugated fluorochromes or other biotin-binding molecules. In research applications, it functions by specifically recognizing CD8α expressed on the majority of thymocytes, subpopulations of αβ T cells, γδ T cells, and some NK cells . The biotin conjugation allows for versatile experimental applications including flow cytometry, cell isolation, and immunohistochemistry, particularly when signal amplification is needed or when multicolor panels require strategic fluorochrome combinations .

How do CD8+ T cells differ structurally and functionally from other T cell populations?

CD8+ T cells express the CD8 glycoprotein either as disulfide-linked homodimers (CD8αα) or as heterodimers (CD8αβ) when disulfide-bonded to a CD8 beta chain (CD8β). While conventional CD8+ αβ T cells coexpress both CD8αα homodimers and CD8αβ heterodimers, some γδ T cells and NK cells exclusively express CD8αα homodimers .

Functionally, CD8+ T cells typically act as cytotoxic T lymphocytes that recognize antigenic peptides presented by MHC class I molecules. The extracellular immunoglobulin superfamily domain of CD8α binds to non-polymorphic determinants on HLA class I molecules (specifically the α3 domain), enabling CD8 to function as a co-receptor with MHC class I-restricted T cell receptors during antigen recognition. The cytoplasmic domain of CD8α associates with Lck, a Src family protein tyrosine kinase involved in intracellular signaling pathways crucial for T cell activation . Recent research has also revealed that CD8 T cells can sometimes acquire follicular helper-like properties, supporting B cell antibody class-switching in certain autoimmune contexts .

What are the critical parameters for optimal CD8 Biotin Antibody staining in flow cytometry?

For optimal CD8 Biotin Antibody staining in flow cytometry:

  • Cell concentration and antibody titration: Use approximately 1 × 10^6 cells in a 100-μl experimental sample. The pre-diluted reagent should be used at the recommended volume per test, but verification through antibody titration is advisable for each experimental system .

  • Isotype control: An isotype control should be used at the same concentration as the CD8 Biotin Antibody to account for non-specific binding .

  • Streptavidin conjugate selection: When using biotin-conjugated antibodies, select an appropriate streptavidin-fluorochrome conjugate that fits your panel design, considering spectral overlap with other fluorochromes.

  • Secondary staining protocol: After primary staining with CD8 Biotin Antibody, wash cells thoroughly before adding streptavidin conjugates to prevent non-specific binding.

  • Instrument settings: Refer to established guidelines for fluorochrome spectra and suitable instrument settings based on your flow cytometer configuration .

  • Buffer considerations: Use buffers containing sodium azide with caution as it yields highly toxic hydrazoic acid under acidic conditions. Always dilute azide compounds in running water before discarding .

How do aptamer-based CD8+ T cell isolation methods compare to traditional antibody-based methods?

Aptamer-based CD8+ T cell isolation represents an innovative alternative to traditional antibody-based methods, offering several distinct advantages:

FeatureAntibody-Based IsolationAptamer-Based Isolation
Isolation principleUses anti-CD8 antibodies conjugated to magnetic beadsUses DNA aptamers generated through cell-SELEX with affinity for CD8
Cell release mechanismGenerally requires harsh elution conditionsAllows traceless isolation through toehold-mediated strand displacement
Cell purityHigh purity (comparable to aptamer method)Average purity of ~95.6% CD8+ T cells in isolation fraction
Cell yieldHigh yield (~97.5% of CD8+ T cells from starting population)Combined yield of ~97.5%, with traceless isolation alone yielding ~72.3%
Effect on cell surfaceMay reduce detectable CD8 expression after isolationPreserves normal CD8 expression levels comparable to pre-isolation
Cost considerationsMore expensiveSignificantly lower cost (~$5-10 of aptamer for clinical-scale isolation)
Clinical applicationsCurrently approved for clinical-scale CAR-T manufacturingPromising for CAR-T cell manufacturing with comparable functionality

Research has demonstrated that CAR-T cells manufactured from aptamer-isolated CD8+ T cells exhibit identical performance to those generated from antibody-selected cells in anti-tumor effector function assays, suggesting that aptamer-based traceless cell isolation represents a practical selection strategy for advanced cell therapy applications .

What methodological considerations are important when using CD8 Biotin Antibody for isolating T cells for CAR-T cell production?

When using CD8 Biotin Antibody for isolating T cells for CAR-T cell production, several methodological considerations are critical:

  • Purity vs. Yield Tradeoff: While high purity of CD8+ T cells (>95%) is achievable with both antibody and aptamer-based methods, researchers must consider the tradeoff between purity and yield. For clinical applications requiring defined product compositions, prioritizing high purity may be more important than maximizing yield, especially since a typical apheresis product contains sufficient T cells for therapeutic manufacturing even with moderate isolation efficiency .

  • Label-free considerations: Traditional antibody-based isolation methods may leave residual antibodies on isolated cells, potentially affecting downstream functionality. When using biotin-conjugated antibodies, consider whether the biotin-streptavidin interaction might interfere with subsequent manipulations. Newer aptamer-based methods offer traceless isolation where selected cells are released label-free through complementary oligonucleotides .

  • Functional assessment: Post-isolation assessment is crucial. Research shows that CD8+ T cells isolated using aptamer-based methods demonstrate normal CD8 expression levels post-isolation, whereas antibody-isolated cells may show reduced CD8 staining .

  • Transduction efficiency: Studies indicate that CD8+ T cells isolated by either antibody or aptamer methods can be effectively transduced with lentiviral vectors encoding CARs (e.g., CD19scFv-41BB-CD3ζ), achieving high expression of transduction markers (>60% initially, increasing to >94% after expansion) .

  • Expansion protocols: Standardized 2-week stimulation protocols followed by rapid expansion with irradiated feeder cells can effectively expand isolated CD8+ T cells regardless of isolation method .

  • Functional equivalence validation: CAR-T cells generated from differently isolated CD8+ T cells should be compared for key functions including proliferation, phenotype, effector function, and anti-tumor activity in appropriate models .

What are the primary limitations of CD8+ T cell depletion studies in understanding T cell dynamics?

CD8+ T cell depletion studies using antibodies like anti-CD8β mAb have revealed important limitations and considerations:

  • Specificity of depletion reagents: Different anti-CD8 antibodies target distinct epitopes, affecting which cell populations are depleted. Anti-CD8β mAb specifically depletes classical MHC-Ia–restricted, TCR-αβ–expressing CD8αβ+ T cells while preserving CD8αα+ cell types, whereas CD8α-targeting can have broader effects .

  • Off-target immunological effects: CD8α-targeted cell depletion can cause unintended immunological consequences, including increased IL-15 bioactivity that drives activation, differentiation, and proliferation of residual cells, particularly CCR5-expressing CD4+ memory T cells. This can potentially increase CD4+ memory T cell viral susceptibility and viral reactivation in latently infected cells .

  • Variability in depletion efficacy: Complete depletion is challenging to achieve consistently across different anatomical compartments, with potential persistence of CD8+ T cells in certain tissues affecting result interpretation.

  • MHC influence on outcomes: Studies using animal models with defined MHC-Ia allomorphs associated with moderately effective (Mamu-A01) versus highly effective (Mamu-B08) CD8+ T cell responses have shown that genetic background significantly influences experimental outcomes .

  • Compartmentalization effects: CD8+ T cells are naturally excluded from some key sites of viral persistence, particularly CD4+ follicular helper T cells within B cell follicles, complicating the interpretation of depletion studies focused on viral rebound dynamics .

  • Functional heterogeneity: Not all CD8+ T cells have identical functionality; some may acquire follicular helper-like properties in certain contexts, promoting B cell antibody class-switching rather than cytotoxic functions .

How can researchers optimize multiparameter flow cytometry panels that include CD8 Biotin Antibody?

Optimizing multiparameter flow cytometry panels with CD8 Biotin Antibody requires careful consideration of several factors:

  • Strategic positioning in staining sequence: When using biotin-conjugated antibodies in multiparameter panels, apply a sequential staining approach where the biotin-conjugated primary antibody is applied first, followed by washing steps before adding streptavidin conjugates and additional directly labeled antibodies.

  • Fluorochrome selection for streptavidin conjugate: Select a streptavidin conjugate with a fluorochrome that optimally fits your panel design based on:

    • Expression level of CD8 (high expression allows use of dimmer fluorochromes)

    • Spectral properties of your cytometer

    • Potential spectral overlap with other markers in your panel

    • Refer to comprehensive spectral viewers available from manufacturers for optimal combinations

  • Panel balance: When designing panels including both CD4 and CD8 identification:

    • Consider pairing CD8 Biotin/streptavidin detection with direct conjugates for CD4

    • Avoid using spectrally overlapping fluorochromes for CD4 and CD8

    • Use dump channels to exclude unwanted populations (e.g., B cells, monocytes)

  • Titration for optimal signal-to-noise ratio: Even with pre-diluted reagents, verification of optimal concentration through titration is recommended to minimize background while maintaining robust positive signal .

  • Controls:

    • Include proper isotype controls conjugated to biotin at the same concentration

    • Use fluorescence-minus-one (FMO) controls to set accurate gates

    • Consider single-stained controls for compensation when using multiple fluorochromes

  • Instrument calibration: Ensure proper instrument setup, including tracking bead standardization and application-specific calibration protocols .

What are the critical factors that can influence CD8 Biotin Antibody binding specificity and how can they be addressed?

Several critical factors can influence CD8 Biotin Antibody binding specificity, with solutions to address potential issues:

How do researchers verify the functional activity of CD8+ T cells following isolation with biotin-conjugated antibodies?

Verifying the functional activity of CD8+ T cells following isolation with biotin-conjugated antibodies requires comprehensive assessment across multiple parameters:

  • Phenotypic analysis:

    • Assess post-isolation expression of key markers including CD8, CD3, TCR complexes, and activation markers (CD25, CD69)

    • Compare CD8 expression levels between isolated cells and the original population, as antibody binding may transiently mask or downregulate CD8 expression

  • Proliferation capacity:

    • Measure proliferative response to stimulation with anti-CD3/CD28 beads or specific antigens

    • Use CFSE dilution or proliferation markers (Ki-67) to track division kinetics

    • Compare proliferation rates to cells isolated using alternative methods (e.g., untouched isolation)

  • Cytokine production:

    • Assess production of key effector cytokines (IFN-γ, TNF-α, IL-2) following stimulation

    • Compare cytokine profiles to those from cells isolated by alternative methods

    • Consider using intracellular cytokine staining, ELISA, or multiplex cytokine assays

  • Cytotoxic function:

    • Measure cytotoxic activity against appropriate target cells

    • Assess granzyme and perforin expression and release

    • For CAR-T applications, compare specific lysis of target cells between different isolation methods

  • Genomic and transcriptomic profiling:

    • Consider RNA-seq analysis to compare global transcriptional profiles

    • Look for potential isolation-induced stress signatures or activation of unwanted pathways

  • In vivo functionality:

    • For cell therapy applications, compare in vivo persistence, trafficking, and anti-tumor activity in appropriate animal models

    • Assess potential differences in therapeutic efficacy between isolation methods

  • Exhaustion and activation analysis:

    • Evaluate expression of exhaustion markers (PD-1, TIM-3, LAG-3) that might be induced by isolation procedures

    • Determine baseline activation state to ensure isolation has not artificially activated the cells

Research comparing antibody and aptamer-based isolation methods has demonstrated that CAR-T cells manufactured from cells isolated using either approach show comparable performance in proliferation, phenotype, effector function, and anti-tumor activity, suggesting that both isolation methods can yield functionally equivalent therapeutic products .

How are CD8 Biotin Antibodies being adapted for single-cell analysis technologies?

CD8 Biotin Antibodies are being strategically adapted for cutting-edge single-cell analysis technologies through several innovative approaches:

  • Integration with mass cytometry (CyTOF): By conjugating streptavidin with rare earth metals, biotin-labeled CD8 antibodies can be incorporated into high-parameter CyTOF panels that simultaneously measure 40+ parameters without fluorescence spillover concerns. This enables comprehensive phenotyping of CD8+ T cell subsets at unprecedented resolution.

  • Single-cell RNA sequencing applications:

    • In CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing), biotinylated CD8 antibodies can be linked to oligonucleotide barcodes via streptavidin bridges, allowing simultaneous measurement of CD8 protein expression and whole-transcriptome analysis

    • For cell hashing applications, biotinylated CD8 antibodies can be used to identify and isolate specific populations prior to droplet-based single-cell analysis

  • Spatial transcriptomics integration: Biotin-conjugated CD8 antibodies can be used with streptavidin-linked fluorescent tags or enzymes for multiplexed imaging of tissue sections, enabling spatial mapping of CD8+ T cells in relation to their transcriptional profiles and tissue microenvironments.

  • Microfluidic applications: In microfluidic single-cell analysis platforms, biotinylated CD8 antibodies can be immobilized on streptavidin-coated surfaces for selective capture of CD8+ T cells prior to downstream analysis.

  • Multi-omic approaches: The versatility of the biotin-streptavidin system allows CD8 Biotin Antibodies to be incorporated into protocols that simultaneously assess transcriptomics, proteomics, and epigenetics from the same single cells.

  • Single-cell functional assays: Using microwell technologies, individual CD8+ T cells identified with biotin-labeled antibodies can be assessed for cytokine secretion, cytotoxicity, and proliferation at the single-cell level.

These adaptations enable researchers to comprehensively characterize heterogeneity within CD8+ T cell populations and correlate phenotypic markers with functional states at unprecedented resolution.

What novel roles for CD8+ T cells have been discovered beyond traditional cytotoxic functions?

Recent research has revealed several novel and unexpected roles for CD8+ T cells beyond their classical cytotoxic functions:

  • Follicular helper-like activity: In certain autoimmune contexts, CD8+ T cells can acquire a follicular helper-like phenotype, expressing CXCR5, Bcl6, and IL-21. These cells, sometimes termed CD8 follicular T cells (CD8 Tfc), can localize to B cell follicles and promote B cell differentiation and antibody isotype class-switching. This phenomenon has been observed in both IL-2-deficient and scurfy mouse models of autoimmunity .

  • Germinal center regulation: CD8+ T cells can express B cell co-stimulatory proteins and support B cell activity within germinal centers. In rheumatoid arthritis synovial ectopic follicles, CD8+ T cells constitute the majority of infiltrating T cells and express CD40L, which plays an important role in B cell germinal center reactions. These CD8+ T cells are required for the formation and maintenance of ectopic germinal centers .

  • CXCR5+ CD8+ T cell functions: Under chronic viral infection and inflammatory conditions, CD8+ T cells can express CXCR5, allowing them to localize to B cell follicles. In human tonsil, these CXCR5+ CD8+ T cells support B cell survival. In LCMV and SIV infections, CXCR5+ CD8+ memory T cells within germinal centers control viral load and express many genes associated with CD4+ T follicular helper differentiation and function .

  • Role in autoimmune pathogenesis: Beyond their direct cytotoxic functions, CD8+ T cells can contribute to autoimmune pathogenesis through follicular-like differentiation and functionality. Depletion of CD8+ T cells has been shown to mitigate autoimmune pathogenesis in IL-2-deficient mice, highlighting their pathogenic potential in certain contexts .

These findings expand our understanding of CD8+ T cell biology beyond the traditional view of cytotoxic effectors and suggest these cells have significant functional plasticity that may be harnessed or targeted in various disease settings.

How do different CD8+ T cell isolation methods impact downstream applications in cell therapy research?

The choice of CD8+ T cell isolation method can significantly impact downstream applications in cell therapy research through several mechanisms:

  • Cell surface marker modulation:

    • Antibody-based isolation can temporarily downregulate CD8 expression, potentially affecting early post-isolation functionality

    • Research comparing aptamer and antibody isolation has shown that CD8 staining of aptamer-isolated CD8+ T cells with the RPA-T8 antibody clone was comparable to that of CD8+ T cells in the starting population, whereas antibody-isolated CD8+ T cells showed lower CD8 staining intensity

  • Transduction efficiency and transgene expression:

    • While isolation method might theoretically affect cell receptiveness to viral transduction, comparative studies have shown high expression of transgenes (>60%) following transduction with lentiviral vectors encoding CARs in both antibody and aptamer-isolated CD8+ T cells

    • After enrichment and expansion periods, no significant differences in transgene expression were observed between cells isolated by different methods

  • Functional characteristics in manufactured cell products:

    • Despite initial differences immediately after isolation, CD8+ CAR-T cells generated from both antibody-isolated and traceless aptamer-isolated cells showed equivalent:

      • Expansion characteristics during manufacturing

      • Phenotypic profiles

      • Gene expression patterns

      • Effector functions in vitro

      • Anti-tumor activity in mouse models of B-cell lymphoma

  • Cost and scalability considerations:

    • Aptamer-based methods offer significant cost advantages for clinical-scale isolation (estimated $5-10 of aptamer needed) compared to antibody-based approaches

    • The trade-off of slightly lower yield (72.3% for traceless aptamer isolation vs. higher for antibody methods) for high purity (>95%), lower cost, and label-free selection has minimal practical consequences in therapeutic manufacturing

  • Regulatory and manufacturing implications:

    • Traceless isolation methods that leave no residual selection reagents on final cell products may offer regulatory advantages

    • Currently, antibody-based CD8 Microbeads remain the only selection technology approved for clinical-scale CAR-T cell manufacturing, though aptamer approaches show promise for future clinical applications

How might advancements in aptamer technology further improve CD8+ T cell isolation for research and therapeutic applications?

Advancements in aptamer technology hold significant promise for further improving CD8+ T cell isolation through several innovative approaches:

  • Development of aptamer cocktails: Creating combinations of aptamers targeting different epitopes of CD8 or multiple T cell markers simultaneously could enhance both specificity and yield. This multi-target approach might overcome limitations associated with single-target recognition.

  • Sequential, multi-marker isolation: By employing multiple aptamers with the corresponding complementary oligonucleotides, researchers could develop fully synthetic systems for sequential and traceless isolation of desired lymphocyte subsets from a single system . This would enable the isolation of highly specific T cell subpopulations.

  • Engineered aptamer modifications:

    • Incorporating chemical modifications to enhance aptamer stability and binding affinity

    • Developing aptamers with temperature or pH-dependent binding properties for controlled release

    • Creating photo-switchable aptamers that change conformation upon light exposure for gentler elution methods

  • Integration with microfluidic platforms: Combining aptamer-based isolation with microfluidic systems could enable high-throughput, automated isolation of CD8+ T cells with minimal sample requirements and operator intervention.

  • Closed-system manufacturing: Developing fully closed, GMP-compatible aptamer-based isolation systems specifically designed for therapeutic applications would streamline manufacturing workflows and reduce contamination risks.

  • Aptamer evolution for subset specificity: Using modified cell-SELEX procedures to generate aptamers specific for CD8+ T cell subsets (naïve, memory, effector, exhausted) would enable more precise isolation of functionally defined populations.

  • Direct aptamer-CAR linkage: Creating aptamers that can both isolate CD8+ T cells and deliver CAR-encoding genetic material could potentially streamline manufacturing processes by combining isolation and modification steps.

  • Dual-function aptamers: Developing aptamers that both isolate CD8+ T cells and activate specific signaling pathways could potentially enhance downstream expansion or functionality.

These advancements would address current limitations of CD8+ T cell isolation methods, potentially improving both research applications and clinical manufacturing of cell therapies.

What are the implications of CD8+ T cell functional heterogeneity for experimental design and interpretation?

The emerging understanding of CD8+ T cell functional heterogeneity has profound implications for experimental design and data interpretation:

  • Marker selection beyond CD8: Given the functional diversity within CD8+ populations, experiments should incorporate additional markers to distinguish:

    • Cytotoxic vs. helper-like CD8+ T cells

    • Tissue-resident vs. circulating memory cells

    • Exhausted vs. functional effector cells

    • Cells with follicular helper-like properties expressing CXCR5, Bcl6, and IL-21

  • Spatial context considerations: CD8+ T cells demonstrate distinct functionalities in different anatomical locations. Experiments should:

    • Consider localization to B cell follicles vs. extrafollicular regions

    • Assess potential exclusion from key sites (like B cell follicles in viral reservoirs)

    • Evaluate tissue-specific functions that may not be captured in blood samples

  • Genetic background influence: MHC alleles significantly impact CD8+ T cell functionality. Studies should:

    • Consider MHC-I allomorphs associated with different effector capacities (e.g., Mamu-A01 vs. Mamu-B08 in rhesus macaques)

    • Account for genetic heterogeneity when interpreting variable responses across experimental subjects

  • Temporal dynamics: CD8+ T cell functionality evolves over the course of responses. Experimental designs should:

    • Include appropriate time-course analyses

    • Consider that ART suppression leads to contraction of virus-specific CD8+ T cell responses and loss of effector differentiation

    • Account for potential functional adaptation during chronic stimulation

  • Depletion study limitations: When using CD8-depleting antibodies, researchers must consider:

    • Differential effects on CD8αβ vs. CD8αα populations

    • Potential off-target immunological effects, including increased IL-15 bioactivity

    • Variability in depletion efficacy across anatomical compartments

  • Functional readout diversity: Experiments should incorporate multiple functional readouts beyond cytotoxicity, including:

    • B cell helper functions and antibody class-switching promotion

    • Cytokine production profiles

    • Proliferative capacity and longevity

    • Metabolic characteristics

    • Transcriptional profiles

  • Target cell resistance mechanisms: Consider that cells harboring reactivating virus may be intrinsically resistant to CD8+ T cell–mediated cytolysis, affecting interpretation of therapeutic potential .

These considerations highlight the need for comprehensive experimental approaches that capture the functional complexity of CD8+ T cells across diverse contexts.

How will single-cell technologies transform our understanding of CD8+ T cell functionality in disease and therapy?

Single-cell technologies are poised to revolutionize our understanding of CD8+ T cell functionality through several transformative approaches:

  • Unraveling functional heterogeneity:

    • Single-cell RNA sequencing will reveal previously unappreciated transcriptional programs within CD8+ T cell populations

    • Integration of protein (CITE-seq) and transcriptomic data will connect surface phenotypes with functional states

    • Identification of novel CD8+ T cell subsets with specialized functions, including those with helper-like or follicular properties

  • Clonal tracking and evolution:

    • Single-cell TCR sequencing will enable tracking of specific T cell clones throughout disease progression and therapy

    • Combined TCR and transcriptome analysis will reveal how identical clones may adopt different functional states in different microenvironments

    • Tracking of barcoded viral clonotypes (like SIVmac239M with ~10,000 different barcodes) will allow discrimination of viral-specific responses at unprecedented resolution

  • Spatial context integration:

    • Spatial transcriptomics and high-parameter imaging technologies will map CD8+ T cell localization relative to other immune cells, target cells, and anatomical structures

    • Understanding of how tissue niches shape CD8+ T cell functionality

    • Visualization of CD8+ T cell exclusion or inclusion in specific anatomical compartments, such as B cell follicles

  • Therapeutic response prediction:

    • Identification of transcriptional signatures predictive of CD8+ T cell efficacy in adoptive cell therapies

    • Early detection of dysfunction or exhaustion markers that might limit therapeutic potential

    • Development of quality control metrics for cell therapy products based on single-cell profiles

  • Resistance mechanism elucidation:

    • Understanding of how target cells develop resistance to CD8+ T cell-mediated killing

    • Identification of checkpoint molecules and inhibitory pathways at single-cell resolution

    • Discovery of microenvironmental factors that compromise CD8+ T cell function in specific disease contexts

  • Dynamic response visualization:

    • Real-time tracking of CD8+ T cell activation, differentiation, and function

    • Monitoring of individual cell transitions between functional states during disease and therapy

    • Characterization of rare cellular events that may have disproportionate impacts on outcomes

  • Multi-omic integration:

    • Combining transcriptomics with epigenomics, proteomics, and metabolomics at single-cell level

    • Comprehensive mapping of regulatory networks governing CD8+ T cell functionality

    • Identification of targetable pathways for therapeutic enhancement

These technological advances will transform our ability to harness CD8+ T cells for therapeutic applications by providing unprecedented insights into the cellular and molecular determinants of their diverse functionalities.

Product Science Overview

Introduction

The “CD8, Rat Anti-Mouse, Biotin” antibody is a monoclonal antibody specifically designed to target the CD8 antigen in mice. This antibody is conjugated with biotin, a vitamin that allows for easy detection and purification of the antibody-antigen complex. The CD8 antigen is a cell surface glycoprotein found on most cytotoxic T lymphocytes, which play a crucial role in the immune response.

CD8 Antigen

The CD8 antigen, also known as Lyt-2 or Ly-2, consists of disulfide-linked alpha (α) and beta (β) chains that form either an α/β heterodimer or an α/α homodimer. The CD8 α/β heterodimer is predominantly expressed on the surface of most thymocytes and a subset of mature TCR α/β T cells. The CD8 α/α homodimer, on the other hand, is expressed on a subset of γ/δ TCR-bearing T cells, natural killer (NK) cells, intestinal intraepithelial lymphocytes, and lymphoid dendritic cells .

Function of CD8

CD8 serves as an antigen co-receptor on T cells, interacting with major histocompatibility complex (MHC) class I molecules on antigen-presenting cells or epithelial cells. This interaction is crucial for the activation of T cells, as it promotes the association of the T-cell receptor (TCR) complex with protein tyrosine kinase lck (p56lck), leading to T-cell activation and subsequent immune response .

Rat Anti-Mouse CD8 Antibody

The rat anti-mouse CD8 antibody is a monoclonal antibody that specifically binds to the CD8 antigen in mice. The antibody is produced by immunizing rats with mouse thymus or spleen cells, followed by the extraction and purification of the antibody from the rat’s serum. The antibody is then conjugated with biotin, which allows for easy detection and purification using streptavidin-based methods .

Applications

The biotin-conjugated rat anti-mouse CD8 antibody is widely used in various immunological assays, including:

  • Flow Cytometry: This technique allows for the analysis of cell surface markers, such as CD8, on individual cells within a mixed population. The biotin-conjugated antibody can be detected using streptavidin-conjugated fluorochromes, enabling the identification and quantification of CD8-expressing cells .
  • Cell Separation: The biotin-conjugated antibody can be used to isolate CD8-expressing cells from a mixed population using streptavidin-coated magnetic beads or columns .
  • Immunohistochemistry: This technique involves the staining of tissue sections to visualize the distribution and localization of CD8-expressing cells. The biotin-conjugated antibody can be detected using streptavidin-conjugated enzymes or fluorochromes .

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.