CD38 is a 300-amino acid protein with a single transmembrane domain and a C-terminal catalytic site. Key structural and enzymatic features include:
CD38 metabolizes NAD+ to produce cyclic ADP-ribose (cADPR) and ADP-ribose (ADPR), which regulate intracellular calcium signaling. It also performs a base-exchange reaction to synthesize nicotinic acid adenine dinucleotide phosphate (NAADP) under specific conditions .
CD38 is ubiquitously expressed but most abundant in hematopoietic tissues. Its expression varies across immune cells:
CD38 is also detected in non-immune tissues, including the prostate, kidney, and brain .
Calcium Signaling: cADPR and ADPR mobilize Ca²⁺ from endoplasmic reticulum stores, modulating T cell activation, B cell receptor signaling, and neutrophil migration .
Cell Adhesion: Binds CD31 (PECAM-1) on endothelial cells, facilitating leukocyte transmigration during inflammation .
NAD+ Homeostasis: Depletes extracellular NAD+, impacting energy metabolism and immune cell function .
Multiple Myeloma (MM): Overexpressed on malignant plasma cells; targeted by monoclonal antibodies (e.g., daratumumab) .
Autoimmunity: Regulates inflammatory pathways in rheumatoid arthritis and lupus .
Asthma: Amplifies airway smooth muscle contractility via cADPR-mediated Ca²⁺ signaling .
CD38 is a validated therapeutic target, particularly in hematologic malignancies:
Recombinant human CD38 (e.g., BPS Bioscience #71277) is used to study enzymatic activity and drug screening:
Parameter | Specification |
---|---|
Construct | Amino acids 43–300 (C-terminal His-tag) |
Purity | ≥90% (low aggregation) |
Enzymatic Activity | Cyclase: 0.5–2.0 nmol/min/μg; Hydrolase: Inhibited by 78% at 10 μM flavonoid |
B Cell Receptor Regulation: CD38 stabilizes IgM:CD19 synapses, enhancing B cell activation .
Metabolic Impact: CD38 deficiency elevates NAD+ levels, suggesting therapeutic potential in age-related diseases .
Inflammation: CD38 knockout mice show impaired neutrophil recruitment during bacterial infections .
CD38 is a single-chain 45-kDa type II glycoprotein with a unique pattern of surface expression. It is present on early hematopoietic cells, lost during maturation, and re-expressed during cell activation . The human CD38 gene is an eight-exon complex located on chromosome 4p15, forming part of the eukaryotic nicotinamide adenine dinucleotide (NAD+) glycohydrolase/ADP-ribosyl cyclase gene family . CD38 functions both as an ectoenzyme and as a receptor, participating in cell adhesion and signal transduction pathways.
Key structural features include:
Type II transmembrane protein (N-terminus inside the cell)
Molecular weight of approximately 45 kDa
Significant glycosylation contributing to mature protein mass
Enzymatic domains for NAD+ hydrolysis and cyclic ADP-ribose synthesis
CD38 displays a variable expression pattern across different cell lineages and developmental stages:
Hematopoietic system: Present on early hematopoietic progenitors, downregulated during maturation, and re-expressed upon activation
Immune cells: Highly expressed on activated lymphocytes, mature plasma cells, and subsets of NK cells
Non-hematopoietic tissues: Found in brain cells, pancreatic islets, and smooth muscle cells
Pathological contexts: Extremely high expression in multiple myeloma plasma cells and certain other hematological malignancies
This differential expression pattern makes CD38 both an interesting target for basic research on cellular differentiation and a potential therapeutic target in various disease contexts.
CD38 serves multiple functions in human physiology:
Enzymatic activity: Functions as an NAD+ glycohydrolase and ADP-ribosyl cyclase, generating second messengers like cyclic ADP-ribose that regulate calcium signaling
Adhesion molecule: Acts as a counter-receptor for CD31 (PECAM-1), mediating leukocyte adhesion to endothelial cells and facilitating leukocyte migration
Immune regulation: Contributes to NK cell function by facilitating immune synapse formation with target cells
Signaling receptor: Triggers intracellular calcium fluxes and cytokine production upon engagement with its ligand CD31
These diverse functions highlight CD38's importance in multiple physiological processes, from immune surveillance to cell migration and communication.
CD38 plays a critical role in human NK cell immune synapse formation, which is essential for NK cell cytotoxicity and cytokine production. Research shows that CD38 expression marks a mature subset of human NK cells with high functional capacity .
CD38 localizes and accumulates at the immune synapse between NK cells and their target cells
Blockade of CD38 severely impairs NK cells' ability to form conjugates and immune synapses with target cells
CD38-mediated synapse formation is independent of its enzymatic activity
NK cells expressing high levels of CD38 display enhanced killing capacity
These CD38-high NK cells produce more IFN-γ when encountering influenza virus-infected and tumor cells
Blocking CD38 or CD31 abrogates NK cell killing and cytokine secretion
Methodologically, researchers investigating this aspect of CD38 function typically employ conjugate formation assays, confocal microscopy to visualize immune synapse formation, and cytotoxicity assays with and without CD38-blocking antibodies.
The CD38 gene shows polymorphism, with the rs3796863 SNP being particularly well-studied. Research indicates that variations in CD38 are associated with differences in social-emotional processing and sensitivity:
Carriers of the A allele (AA/AC genotypes) of CD38 rs3796863 display higher distress-related responses to emotional stimuli compared to individuals with the CC genotype
The distress response effect size was η² = 0.027, indicating a small but significant effect
This heightened emotional reactivity appears to be specific to personal distress rather than empathic concern
Sex differences interact with genotype effects, with females generally showing higher emotional responses regardless of genotype
Studies examining CD38 genetic variation typically use genotyping assays (e.g., PCR-based methods)
Emotional responses are measured using standardized stimuli and validated self-report measures
Controlling for sex differences is essential due to their significant impact on emotional responses
This research suggests that CD38 variation may contribute to individual differences in social sensitivity and distress responses, potentially through effects on oxytocin system function .
CD38 and CD31 cognate interactions play a crucial role in regulating leukocyte migration and inflammatory responses:
Adhesion cascade: CD38 on leukocytes interacts with CD31 on endothelial cells, contributing to the early stages of leukocyte rolling and tethering
Signal transduction: This receptor-ligand interaction triggers:
Functional outcomes:
This complex interplay represents an important step in the orchestrated process of leukocyte migration and homing, with implications for normal immune surveillance and pathological processes such as inflammation and cancer metastasis.
Researchers employ several complementary techniques to investigate CD38's function in immune synapse formation:
When designing experiments to study immune synapse formation, researchers should consider:
Using appropriate target cells that express CD31 (the CD38 ligand)
Including both enzymatic activity inhibitors and receptor-blocking antibodies to distinguish between these functions
Incorporating advanced imaging techniques like TIRF microscopy for detailed synapse visualization
Studying CD38 genetic variation and its phenotypic effects requires a multidisciplinary approach:
PCR-RFLP analysis (the CD38 gene has a bi-allelic polymorphism identifiable by the restriction endonuclease pvu)
SNP genotyping arrays for high-throughput analysis
Next-generation sequencing for comprehensive variant identification
Standardized emotional stimuli (e.g., videos eliciting empathic responses)
Physiological measures (e.g., oxytocin levels, autonomic responses)
Behavioral assays (e.g., prosocial behavior tasks)
Control for sex differences, as they significantly impact emotional responses
Consider potential gene-environment interactions
Use appropriate statistical models to account for covariates
Calculate and report effect sizes (e.g., η²) for proper interpretation of results
For robust studies of CD38 genetic variation, researchers should employ sample sizes adequate for genetic association studies (typically hundreds of participants) and consider replications across diverse populations.
Research on CD38 as a therapeutic target in multiple myeloma encompasses several methodological approaches:
Expression profiling:
Antibody development and characterization:
Generation of anti-CD38 monoclonal antibodies
Assessment of antibody binding affinity, specificity, and effector functions
Evaluation of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC)
Preclinical evaluation:
In vitro cytotoxicity assays using primary myeloma cells
Patient-derived xenograft models
Combination studies with established myeloma treatments
Translational research:
Biomarker studies to identify patients most likely to respond to anti-CD38 therapy
Resistance mechanism investigations
Pharmacokinetic/pharmacodynamic analyses
When designing studies on CD38-targeted therapies, researchers should consider the heterogeneity of CD38 expression within myeloma cells and potential mechanisms to upregulate CD38 expression, such as ATRA treatment, which acts through the retinoic acid α receptor (RARα) .
CD38 serves as an important biomarker in several hematological malignancies:
CD38 is extremely highly expressed on plasma cells and their malignant counterparts in MM
Expression levels can be used for diagnosis, monitoring disease progression, and assessing minimal residual disease
CD38 expression patterns may correlate with disease subtype and prognosis
Expression varies across different leukemia and lymphoma types
Can be used as part of immunophenotyping panels for classification
May serve as a prognostic indicator in certain malignancies
Flow cytometry remains the gold standard for CD38 detection and quantification
Immunohistochemistry provides spatial context in tissue samples
RNA sequencing or qPCR can assess CD38 gene expression levels
The value of CD38 as a biomarker is enhanced by its differential expression pattern: absent on early hematological progenitors but extremely high in plasma cells and myeloma . This expression profile minimizes potential toxicity to normal hematopoietic stem cells when targeting CD38.
Several approaches exist to modulate CD38 expression for therapeutic applications:
Retinoid-mediated upregulation:
Transcriptional regulation:
Various cytokines and inflammatory mediators can influence CD38 expression
Understanding the transcriptional control mechanisms allows targeted modulation
Epigenetic modifications:
DNA methylation and histone modifications influence CD38 expression
Epigenetic modifying drugs may alter CD38 levels
Post-translational modifications:
Regulation of protein trafficking and surface retention
Protection from proteolytic shedding or internalization
Research has shown that increased CD38 protein antigen levels were observed in normal CD34+ bone marrow exposed to ATRA, but not on normal circulating granulocytes, suggesting tissue-specific regulation mechanisms . This differential effect may be leveraged to enhance therapeutic targeting while minimizing unwanted effects.
CD38 plays a significant role in oxytocin-related social behaviors through several mechanisms:
Oxytocin pathway involvement:
Neurobiological mechanisms:
CD38 contributes to calcium signaling in the brain
Involved in oxytocin release from hypothalamic neurons
May modulate neural circuits involved in social cognition
Social-emotional sensitivity:
Research methodologies in this area typically combine genetic analysis, hormone measurements, psychological assessments, and neuroimaging approaches. Findings suggest that like other "sensitivity genes," CD38 variation may influence susceptibility to social-environmental influences, potentially leading to heightened emotional reactivity in threatening or distressing situations .
Several important contradictions and unresolved questions remain in CD38 research:
Dual roles in health and disease:
How can CD38 contribute to both normal immune function and pathological processes?
What determines whether CD38 activity is beneficial or detrimental in a given context?
Enzymatic vs. receptor functions:
Social behavior paradox:
Therapeutic targeting challenges:
How can therapeutic approaches target CD38 on malignant cells while sparing normal cells with important CD38 functions?
What mechanisms underlie resistance to CD38-targeted therapies?
These unresolved questions highlight the complexity of CD38 biology and the need for continued research using diverse methodological approaches and interdisciplinary perspectives.
Several cutting-edge technologies are transforming CD38 research:
Technology | Application to CD38 Research | Advantage |
---|---|---|
CRISPR/Cas9 gene editing | Precise modification of CD38 gene and regulatory elements | Allows causality testing and identification of critical domains |
Single-cell transcriptomics | Analysis of CD38 expression at single-cell resolution | Reveals heterogeneity within cell populations |
Advanced imaging techniques | Visualization of CD38 dynamics during immune synapse formation | Provides spatial and temporal resolution of CD38 function |
Structural biology tools | Determination of CD38 protein structure and interaction sites | Informs rational drug design for CD38 targeting |
Bispecific antibodies | Simultaneous targeting of CD38 and other molecules | Enhances therapeutic specificity and efficacy |
Systems biology approaches | Integration of CD38 into broader cellular networks | Provides context for CD38 function in complex systems |
These technologies are enabling researchers to address long-standing questions about CD38 function and to develop more effective therapeutic strategies targeting this molecule.
CD38 research is poised to expand into several promising directions:
Neurodegenerative diseases:
Exploring CD38's role in neuroinflammation and microglial function
Investigating connections between CD38-mediated calcium signaling and neuronal health
Potential therapeutic targeting in conditions like Alzheimer's and Parkinson's disease
Metabolic disorders:
CD38's involvement in NAD+ metabolism connects it to metabolic regulation
Potential roles in diabetes, obesity, and aging-related metabolic changes
Interaction with sirtuins and other NAD+-dependent enzymes
Autoimmune conditions:
Investigation of CD38's contribution to aberrant immune activation
Potential as a biomarker or therapeutic target in autoimmune diseases
Role in regulatory T cell function and immune tolerance
Precision medicine approaches:
Using CD38 genetic variation to predict treatment responses
Developing personalized therapeutic strategies based on CD38 expression patterns
Integrating CD38 into broader genetic and biomarker panels
As research tools become more sophisticated and our understanding of CD38 biology deepens, these emerging areas represent fertile ground for translational and clinical applications.
CD38 was first identified in 1980 by E. L. Reinherz, S. Schlossman, and colleagues during their pioneering analysis of the human lymphocyte surface using monoclonal antibodies . Initially, it served as a marker for the study of thymocytes and activated T cells. Over time, its expression was also observed on other cell types, including B cells and monocytes .
Structurally, CD38 is composed of a single polypeptide chain with an extracellular domain, a transmembrane region, and a short cytoplasmic tail. The extracellular domain contains the active site responsible for its enzymatic functions.
CD38 has several important functions:
CD38 has gained significant attention in the field of oncology, particularly in the treatment of multiple myeloma (MM). Multiple myeloma is a hematological cancer characterized by the proliferation of malignant plasma cells in the bone marrow . CD38 is highly and uniformly expressed on multiple myeloma cells, making it an attractive target for immunotherapeutic approaches .
Several CD38-targeting monoclonal antibodies have been developed, including daratumumab and isatuximab . These antibodies have shown promising results in clinical trials, demonstrating significant anti-tumor activity and manageable toxicity profiles. CD38-targeted therapies are now an integral part of the treatment regimen for multiple myeloma patients .
Recombinant CD38 refers to the artificially produced form of the protein, typically generated using recombinant DNA technology. This involves inserting the gene encoding CD38 into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. Recombinant CD38 is used in various research applications, including the study of its enzymatic activity, receptor functions, and potential therapeutic uses.