CD19 Human

CD19 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of CD19

CD19 is a 95 kDa Type I transmembrane glycoprotein in the immunoglobulin superfamily (IgSF). Its structure includes:

FeatureDetails
Gene LocationChromosome 16 (7.41 kb gene)
Domains- Extracellular: Two C2-type Ig-like domains (272 amino acids)
- Transmembrane: 22 amino acids
- Cytoplasmic: 243 amino acids with nine conserved tyrosine residues
Key ResiduesY391, Y482, Y513 (essential for signaling)
Post-Translational ModificationsN-linked glycosylation sites

CD19 forms a multimolecular complex with CD21, CD81, and CD225, enhancing B cell receptor (BCR) signaling .

Expression Profile

  • Developmental Stages: Expressed from pro-B cells through mature B cells, excluding plasma cells .

  • Regulation: Surface density increases threefold during B cell maturation .

Functional Roles

  • BCR Signaling:

    • Amplifies BCR responses by lowering activation thresholds via PI3K-AKT and BTK pathways .

    • Recruits SYK and BLNK to propagate survival signals .

  • TLR9 Integration:

    • CD19/PI3K/AKT/BTK axis is essential for TLR9-induced B cell activation .

  • MYC Regulation:

    • Stabilizes c-MYC protein to promote B cell proliferation, independent of BCR .

Immunodeficiencies

  • Mutations in CD19 or its complex partners (CD21, CD81) cause hypogammaglobulinemia and impaired antibody responses .

  • CD19-deficient B cells exhibit reduced MHC class II signaling and survival .

Autoimmunity

  • Overexpression correlates with systemic sclerosis (SSc) and multiple sclerosis (MS) .

  • Mouse models show CD19 deficiency attenuates autoimmune responses .

B Cell Malignancies

DiseaseCD19 ExpressionClinical Implications
B-ALL/CLLNormal/highTarget for CAR-T therapy
DLBCL/FLReducedPoor prognosis
Autoimmune DisordersElevated (e.g., SSc, MS)Therapeutic target for depletion

CAR-T Cell Therapy

  • Mechanism: CD19-targeted CAR-T cells reprogram cytotoxic T cells to eliminate CD19+ B cells .

  • Clinical Trials:

    • Phase I Trial (Hu19-CD841BBZ CAR-T):

      • Efficacy: 2 CR, 3 PR in NHL/CLL patients; 1 CR in B-ALL .

      • Toxicity: CRS and ICANS observed, consistent with other CAR-T therapies .

ParameterResults
Manufacturing Success100% (Prodigy® Device)
Dose Levels5e5–2e6 CAR-T cells/kg
Survival Outcomes1-year progression-free: 57% (NHL)

Targeted Inhibitors

  • PI3K/AKT/BTK inhibitors show efficacy in dampening CD19-driven signaling in autoimmune models .

Research Tools and Reagents

  • Recombinant Proteins:

    • CD19 Fc Chimera (Genscript): Used for binding assays .

    • GST-Tagged CD19 (Bio-Techne): For signaling studies .

Product Specs

Introduction

CD19, also referred to as B-Lymphocyte Surface Antigen B4, is a member of the immunoglobulin superfamily and is found on the surface of almost all B-lymphocytes, excluding fully matured plasma cells. Functioning as a signal-transducing receptor, CD19 plays a crucial role in regulating both B cell differentiation and proliferation. It acts as an adaptor protein, recruiting cytoplasmic signaling proteins to the cell membrane. Working in conjunction with the CD19/CD21 complex, it lowers the activation threshold of B cell receptor signaling pathways. Given its presence on most B cells, CD19 serves as a valuable biomarker in diagnosing lymphoma, understanding B lymphocyte development, and as a target for immunotherapies targeting leukemia.

Description

CD19, produced using Sf9 Baculovirus cells, is a single-chain polypeptide with glycosylation. It consists of 510 amino acids (21-291a.a.) and has a molecular weight of 57.0kDa. Note: On SDS-PAGE, the apparent molecular size will be approximately 50-70kDa. This CD19 protein is expressed with a 239 amino acid hIgG-His tag at its C-Terminus and is purified using proprietary chromatographic techniques.

Physical Appearance
A sterile, colorless solution that has been filtered.
Formulation

The provided CD19 protein solution has a concentration of 0.25mg/ml. It is prepared in a solution containing Phosphate Buffered Saline at a pH of 7.4 and 10% glycerol.

Stability
For short-term storage (up to 2-4 weeks), keep the CD19 protein solution refrigerated at 4°C. For longer storage, it should be stored frozen at -20°C. To ensure optimal protein stability during long-term storage, we recommend adding a carrier protein such as HSA or BSA to a final concentration of 0.1%. Repeated freezing and thawing of the protein solution should be avoided to maintain its integrity.
Purity

Analysis by SDS-PAGE indicates that the purity of this CD19 protein is greater than 85.0%.

Synonyms

CD19 Molecule, B-Lymphocyte Surface Antigen B4, T-Cell Surface Antigen Leu-12, Differentiation Antigen CD19,  CD19 Antigen, B-Lymphocyte Antigen CD19, CVID3, B4.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

EEPLVVKVEE GDNAVLQCLK GTSDGPTQQL TWSRESPLKP FLKLSLGLPG LGIHMRPLAI WLFIFNVSQQ MGGFYLCQPG PPSEKAWQPG WTVNVEGSGE LFRWNVSDLG GLGCGLKNRS SEGPSSPSGK LMSPKLYVWA KDRPEIWEGE PPCLPPRDSL NQSLSQDLTM APGSTLWLSC GVPPDSVSRG PLSWTHVHPK GPKSLLSLEL KDDRPARDMW VMETGLLLPR ATAQDAGKYY CHRGNLTMSF HLEITARPVL WHWLLRTGGW KLEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPGKHHHHHH

Q&A

What is CD19 and where is it typically expressed in humans?

CD19 is a 95-kDa type I transmembrane glycoprotein traditionally considered a B-cell lineage-specific surface marker. It functions as a critical co-receptor in B cell receptor (BCR) signaling. While CD19 has historically been characterized as exclusively expressed on B cells from early pre-B cell development until plasma cell differentiation, recent single-cell RNA sequencing and immunohistochemistry studies have revealed that CD19 is also expressed in human brain mural cells, including pericytes and vascular smooth muscle cells (vSMCs), which are critical for blood-brain-barrier integrity .

Methodologically, researchers can detect CD19 expression through:

  • Flow cytometry with fluorescently labeled anti-CD19 antibodies

  • Immunohistochemistry using validated antibodies (such as clone BT51E)

  • Single-cell RNA sequencing (scRNA-seq) to detect CD19 mRNA

  • Quantitative PCR for measuring CD19 transcript levels

When investigating CD19 expression in non-B cell populations, it's essential to employ multiple detection methods, as expression levels may be significantly lower than in B cells.

How is CD19 expression regulated during B cell development?

CD19 expression follows a tightly regulated pattern during B cell development:

Developmental StageCD19 ExpressionRegulatory Factors
Hematopoietic stem cellAbsentSilenced by DNA methylation
Pro-B cellInitial expressionPAX5, EBF1 activation
Pre-B cellHigh expressionContinued PAX5 activity
Immature B cellHigh expressionStabilized expression
Mature B cellHigh expressionMaintained by B cell program
Plasma cellDownregulatedRepressed by BLIMP1

To study CD19 regulation, researchers employ:

  • Chromatin immunoprecipitation (ChIP) to identify transcription factor binding

  • Reporter gene assays to map enhancer and promoter elements

  • CRISPR/Cas9 genome editing to modify regulatory regions

  • DNA methylation analysis using bisulfite sequencing

  • Single-cell RNA-seq to track expression changes across developmental stages

Interestingly, the regulation of CD19 expression in brain mural cells appears to differ from B cells, as these cells do not express B cell-specific transcription factors like PAX5. This suggests alternative regulatory mechanisms that require further investigation .

What are the most effective methods for detecting CD19 expression in human tissues?

Researchers employ various complementary techniques to detect CD19 expression:

MethodApplicationAdvantagesLimitations
Flow cytometryCell suspensionsQuantitative, multi-parameterRequires single-cell suspensions
Immunohistochemistry (IHC)Tissue sectionsPreserves tissue architectureSemi-quantitative
ImmunofluorescenceTissue sectionsMultiple markers, co-localizationPhotobleaching, autofluorescence
Western blotProtein lysatesProtein size confirmationLoses spatial information
qRT-PCRRNA extractsSensitive, quantitativeNo protein confirmation
Single-cell RNA-seqDissociated cellsUnbiased, cell type identificationExpensive, technical challenges
Mass cytometry (CyTOF)Cell suspensionsHigh-parameter, minimal spilloverExpensive, specialized equipment

When detecting CD19 in non-B cell populations such as brain mural cells, researchers should:

  • Use multiple detection methods for confirmation

  • Include appropriate positive controls (B cells) and negative controls

  • Validate antibody specificity with knockout experiments

  • Use co-staining with lineage-specific markers (e.g., CD248, PDGFRB for mural cells)

In research identifying CD19 in brain mural cells, immunohistochemistry using a clinically validated anti-human CD19 antibody (clone BT51E) was used to confirm protein expression in perivascular areas of the human brain .

What structural features of human CD19 are important for function and antibody recognition?

Human CD19 contains several key structural features critical for function:

DomainDescriptionFunction
Extracellular291 amino acids with two C2-type immunoglobulin-like domainsLigand binding and interaction with CD21
Transmembrane21 amino acidsAnchoring to cell membrane
Cytoplasmic242 amino acidsSignal transduction via tyrosine phosphorylation sites

Key structural elements include:

  • Multiple N-linked glycosylation sites that affect antibody recognition

  • Nine conserved tyrosine residues in the cytoplasmic domain, including three YxxM motifs that serve as binding sites for PI3K

  • Disulfide bonds maintaining structural integrity

  • Association with tetraspanin CD81, required for proper surface expression

The epitope recognized by most therapeutic antibodies and CAR-T cells is located in the extracellular domain. Importantly, the CD19 isoform expressed in brain mural cells contains the epitope targeted by clinical CAR-T cells and BiTEs, making these cells legitimate targets for these therapies .

How does CD19 function in normal B cell signaling?

CD19 serves as a critical co-receptor in B cell activation through multiple mechanisms:

  • Signal amplification: CD19 lowers the threshold for B cell activation by enhancing BCR signaling.

  • Co-receptor complex formation: CD19 forms a complex with CD21 (CR2), CD81 (TAPA-1), and CD225 (Leu-13).

  • Tyrosine phosphorylation cascade: Upon B cell activation, CD19's cytoplasmic domain becomes phosphorylated on tyrosine residues.

  • Signaling molecule recruitment: Phosphorylated CD19 recruits and activates:

    • PI3K, leading to Akt activation

    • Vav, activating Rac1 and cytoskeletal rearrangement

    • Lyn and other Src-family kinases

  • Calcium flux enhancement: CD19 signaling amplifies calcium mobilization during B cell activation

Methodological approaches to study these functions include:

  • Phospho-flow cytometry to measure CD19 phosphorylation states

  • CRISPR-based mutagenesis to identify critical functional domains

  • Calcium flux assays using fluorescent indicators

  • Proximity ligation assays to detect protein-protein interactions

  • Super-resolution microscopy to visualize signaling complexes

Whether CD19 in brain mural cells participates in similar signaling pathways remains unknown and represents an important area for future research .

What is the significance of CD19 expression in human brain mural cells?

The discovery of CD19 expression in human brain mural cells has profound implications:

  • Neurotoxicity mechanism: CD19 expression in mural cells provides a potential on-target mechanism for neurotoxicity observed in patients receiving CD19-directed immunotherapies such as CAR-T cells and bispecific T-cell engager (BiTE) antibodies .

  • Blood-brain barrier integrity: Mural cells (pericytes and vSMCs) are critical for maintaining blood-brain barrier (BBB) integrity. CD19-directed therapies may compromise BBB function by targeting these cells, explaining observed cerebral edema in severe cases .

  • Developmental biology: CD19 expression in brain mural cells begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions, suggesting potential developmental roles .

  • Regional heterogeneity: CD19 expression varies across brain regions, with higher expression observed in the hippocampus, insula, temporal lobe, frontal lobe, and parietal lobe compared to regions such as the pons and occipital lobe .

  • Target reassessment: This finding necessitates reevaluation of CD19 as a B-cell-specific target and demands more careful design of immunotherapeutics to minimize neurotoxicity.

To further investigate this phenomenon, researchers can employ:

  • Genetic knockdown of CD19 specifically in mural cells to assess functional consequences

  • Co-culture systems with CD19 CAR-T cells and mural cells to assess direct cytotoxicity

  • In vitro BBB models to assess impact on barrier integrity

  • Spatial transcriptomics to map CD19 expression across different brain vasculature regions

How does CD19 expression in mural cells contribute to neurotoxicity in CD19-directed immunotherapies?

CD19 expression in brain mural cells provides a mechanistic explanation for neurotoxicity in CD19-directed therapies:

  • Direct targeting mechanism: CD19-directed therapies (CAR-T cells, BiTEs) can recognize and target CD19-expressing mural cells in the brain, leading to their destruction .

  • Blood-brain barrier disruption: Mural cell depletion compromises BBB function, leading to increased permeability and cerebral edema .

  • T cell infiltration cascade: BBB disruption facilitates further T cell infiltration into the brain parenchyma, amplifying the inflammatory response .

  • Antigen density sensitivity: CD19 CAR-T cells are sensitive to even low levels of CD19 antigen density, making even low-level expression in mural cells clinically relevant .

  • Regional vulnerability patterns: Heterogeneous CD19 expression across brain regions may explain regional patterns of neurotoxicity and varying manifestations among patients .

  • Recognition of relevant epitope: The CD19 isoform expressed in brain mural cells contains the epitope targeted by clinical CAR-T cells and BiTEs .

Methodological approaches to investigate this mechanism include:

  • Patient cerebrospinal fluid analysis for markers of BBB disruption

  • Postmortem examination of brain tissue from patients with severe neurotoxicity

  • Development of humanized mouse models expressing human CD19 in mural cells

  • Live imaging of CAR-T cell interactions with brain vasculature

This mechanism could explain the higher incidence of neurotoxicity in CD19-directed immunotherapies compared to treatments targeting other B cell proteins, such as CD20 .

What are the differences in CD19 expression between humans and mouse models?

Significant species differences in CD19 expression have important implications for preclinical research:

AspectHuman CD19Mouse Cd19Implication
Expression in mural cellsPresent at detectable levelsLower levelsLimited predictive value of mouse models for neurotoxicity
Protein sequenceHuman-specificMouse-specificPotential differences in antibody/CAR recognition
Developmental patternEarly onset, persistentLess characterizedHuman developmental toxicity may not be predicted
Regional distributionHeterogeneous across brain regionsLess characterizedRegional toxicity patterns may differ
Isoform expressionContains clinical CAR-T target epitopeMay differSpecies-specific targeting effects

These differences highlight critical methodological considerations:

  • Humanized mouse models expressing human CD19 may better recapitulate clinical neurotoxicity

  • Validation in human cells and tissues is essential before clinical translation

  • Careful interpretation of preclinical safety data from conventional mouse models

  • Species-specific antibodies and detection methods must be employed

The lower expression of Cd19 in mouse mural cells explains why preclinical studies often fail to predict the severity of neurotoxicity observed in clinical trials . This finding highlights the importance of human single-cell atlases for designing immunotherapies with improved safety profiles.

How can researchers account for CD19 expression in non-B cell populations when designing CAR-T therapies?

To address CD19 expression in non-B cell populations when designing CAR-T therapies, researchers can employ several strategies:

  • Epitope engineering:

    • Identify unique epitopes on B cell CD19 not present or accessible on mural cell CD19

    • Design CARs targeting B cell-specific post-translational modifications

    • Methodological approach: Comparative epitope mapping between B cells and mural cells

  • Affinity modulation:

    • Develop lower-affinity CARs that recognize high CD19 expression on B cells but not low expression on mural cells

    • Methodological approach: Affinity maturation/reduction with binding kinetics assessment

  • Combinatorial targeting:

    • Design CAR-T cells requiring recognition of multiple B cell-specific antigens (AND logic gates)

    • Methodological approach: Dual-CAR systems or synthetic Notch receptors

  • Spatial control:

    • Incorporate inhibitory receptors recognizing brain-specific antigens

    • Methodological approach: NOT-gate CARs with inhibitory domains

  • Delivery optimization:

    • Develop delivery methods limiting CAR-T cell trafficking to the CNS

    • Methodological approach: Chemokine receptor engineering

  • Inducible safety switches:

    • Incorporate suicide genes or on/off switches to control CAR-T activity

    • Methodological approach: Integration of inducible caspase-9 or similar systems

Recent clinical trials are exploring fully human anti-CD19 CAR-T cells with a 4-1BB costimulatory domain, which may have improved safety profiles . These approaches require careful validation in models that accurately represent human CD19 expression patterns.

What methodological approaches can be used to study CD19 expression in rare cell populations?

Studying CD19 expression in rare cell populations requires specialized methodological approaches:

  • Single-cell RNA sequencing (scRNA-seq):

    • Enables unbiased transcriptome profiling of individual cells

    • Can identify rare populations based on clustering algorithms

    • In the discovery of CD19 in mural cells, researchers analyzed scRNA-seq data from human prefrontal cortex cells, identifying a small population (~1.5% of non-neuronal cells) expressing CD19

  • Meta-cell transcriptome analysis:

    • Aggregates transcriptomes of similar cells to increase signal

    • Researchers aggregated scRNA-seq data into "meta-cell" transcriptomes to efficiently identify populations of interest within large datasets

  • Enrichment strategies:

    • Cell sorting based on known markers (e.g., CD248, PDGFRB for mural cells)

    • Regional microdissection to focus on areas of interest

  • Spatial transcriptomics:

    • Preserves spatial context while providing transcriptomic data

    • Particularly valuable for vascular-associated cells

  • Multi-parameter immunohistochemistry:

    • Simultaneous detection of CD19 and mural cell markers

    • Perivascular CD19 staining was identified using a clinically validated anti-human CD19 antibody (clone BT51E)

  • Validation across multiple datasets:

    • Integration of data from independent sources

    • Researchers confirmed CD19 expression in mural cells across multiple scRNA-seq datasets from different brain regions and developmental timepoints

  • Correlation analysis with known markers:

    • Assessment of co-expression patterns with established markers

    • CD19-expressing cells co-expressed mural cell markers like CD248, RGS5, and PDGFRB, while being negative for B cell markers like CD79A

These approaches collectively enabled the discovery that CD19 is expressed in brain mural cells, highlighting the importance of using multiple complementary methods when studying rare cell populations.

How does CD19 isoform expression differ between brain mural cells and B cells?

Understanding CD19 isoform differences between brain mural cells and B cells is crucial for targeted therapeutics:

AspectB cellsBrain Mural CellsMethodological Approaches
Splice variantsMultiple variants documentedFull-length isoform containing target epitope RNA-seq, PCR for splice variants
Post-translational modificationsHeavily glycosylatedLess characterizedGlycoproteomics, Western blot
Surface densityHigh expressionLower expressionQuantitative flow cytometry
Co-receptor complexForms complex with CD21, CD81, CD225Expresses CD81 but other components unclear Co-immunoprecipitation
Subcellular localizationPrimarily surface membranePerivascular localization observed Confocal microscopy
Regulatory elementsB-cell specific enhancers (PAX5-dependent)Unknown alternative regulatory elementsChIP-seq, ATAC-seq

Research has confirmed that the CD19 isoform expressed in brain mural cells contains the epitope targeted by clinical CAR-T cells and BiTEs . The co-expression of CD81, which chaperones CD19 through secretory pathways and is required for surface expression in B cells, suggests that CD19 may be properly trafficked to the cell surface in mural cells as well .

To characterize these differences, researchers should employ:

  • Isoform-specific PCR and sequencing

  • Epitope mapping with various monoclonal antibodies

  • Mass spectrometry to identify post-translational modifications

  • Functional studies to determine if CD19 in mural cells participates in signaling

Understanding these differences could enable development of therapeutics that selectively target B cell-specific CD19 isoforms or conformations while sparing mural cells.

What are the current challenges and potential solutions in managing neurotoxicity in CD19-directed immunotherapies?

Managing neurotoxicity in CD19-directed immunotherapies presents several challenges with emerging solutions:

ChallengeDescriptionPotential Solutions
Balancing efficacy and safetyMaintaining efficacy against malignant B cells while sparing brain mural cellsControlled affinity CAR designs responding to high CD19 expression but not low expression
Limited predictive modelsMouse models show lower Cd19 expression in mural cells Development of humanized mouse models expressing human CD19 in mural cells
Risk stratificationIdentifying patients at higher risk for neurotoxicityMulti-omics profiling to identify genetic or protein biomarkers of severe neurotoxicity
Regional vulnerabilityCD19 expression varies across brain regions Comprehensive mapping of regional CD19 expression patterns
Therapeutic windowDetermining optimal dosing to maintain efficacy while minimizing toxicityCareful dose-escalation studies with comprehensive neurological monitoring
Monitoring approachesEarly detection of emerging neurotoxicityDevelopment of real-time monitoring systems and prophylactic neuroprotective strategies
Next-generation approachesDesigning inherently safer CD19-directed therapiesLogic-gated CARs requiring multiple target recognition, spatially-controlled activation

Recent clinical trials are exploring fully human anti-CD19 CAR-T cells with a 4-1BB costimulatory domain, which may have different safety profiles compared to earlier generations . Early data suggests potential for lower rates of cytokine release syndrome (CRS) and severe immune effector associated neurotoxicity syndrome (ICANS), but comprehensive safety data is still being collected through carefully designed clinical trials .

Product Science Overview

Structure and Function

CD19 is a 95 kDa transmembrane glycoprotein composed of several domains:

  • Extracellular Domain (ECD): This domain consists of 272 amino acids and contains two immunoglobulin-like domains .
  • Transmembrane Segment: A 22 amino acid segment that anchors the protein to the cell membrane .
  • Cytoplasmic Domain: Comprising 243 amino acids, this domain is involved in intracellular signaling .

The primary function of CD19 is to lower the threshold for B cell receptor (BCR) signaling, thereby enhancing the sensitivity of B cells to antigens. This modulation is critical for the activation and differentiation of B cells, which are essential components of the adaptive immune system .

Expression and Clinical Significance

CD19 is expressed on 100% of peripheral B cells, as defined by the expression of kappa or lambda light chains . It is also expressed on myeloid leukemia cells, particularly those of monocytic lineage . Due to its consistent expression on B cells, CD19 serves as a valuable biomarker for both normal and neoplastic B cells . It is also used in the diagnosis and treatment of B cell-related diseases, including various forms of lymphoma and leukemia .

Recombinant CD19

Recombinant CD19 proteins are produced using recombinant DNA technology, which involves inserting the CD19 gene into an expression system, such as bacteria, yeast, or mammalian cells. This allows for the large-scale production of CD19 proteins, which can be used for research, diagnostic, and therapeutic purposes.

For example, the recombinant human CD19 protein consists of 283 amino acids and has a predicted molecular mass of 31.6 kDa. However, due to glycosylation, it migrates as an approximately 47 kDa band in SDS-PAGE under reducing conditions .

Applications

Recombinant CD19 proteins have several applications:

  • Research: Used to study B cell development, activation, and signaling pathways.
  • Diagnostics: Employed in flow cytometry and immunohistochemistry to identify and quantify B cells in clinical samples.
  • Therapeutics: Utilized in the development of targeted therapies, such as chimeric antigen receptor (CAR) T-cell therapy, which involves engineering T cells to express a receptor that targets CD19 on B cells.

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.