BST1 Human, also known as CD157, is a glycosylphosphatidylinositol (GPI)-anchored glycoprotein with a molecular mass of 30.5–31 kDa (depending on glycosylation status) . Key structural features include:
The protein comprises a single polypeptide chain with conserved cysteine residues critical for enzymatic activity . Recent studies identified two isoforms due to alternative splicing:
BST1 Human exhibits dual enzymatic capabilities:
Activity | Function | Comparison to CD38 |
---|---|---|
ADP-Ribosyl Cyclase | Converts NAD+ to cyclic ADP-ribose (cADPR) | Weaker catalytic efficiency |
Cyclic ADP-Ribose Hydrolase | Hydrolyzes cADPR to ADP-ribose | Absent in CD38 |
Cross-Linking: Induces tyrosine phosphorylation of a 130 kDa protein in myeloid cells (e.g., U937, THP-1), triggering signaling cascades .
Immune Regulation: Mediates neutrophil migration/adhesion via interaction with the CD11b/CD18 complex .
Cell Proliferation: Supports pre-B-cell growth in stromal cell lines .
Bst1−/− Mice: Protected against AKI but prone to impaired neutrophil migration .
Recombinant BST1 Human: Enhances monocyte and hematopoietic stem cell proliferation in vitro and in vivo .
BST1 (Bone Marrow Stromal Cell Antigen 1) is a cell surface protein belonging to the CD38 family and is anchored to the cell membrane by a GPI (glycosylphosphatidylinositol) anchor. Initially identified on bone marrow stromal cells, BST1 acts as an ectoenzyme, similar to ADP-ribosyl cyclase CD38. Both BST1 and CD38 possess DP-ribosyl cyclase and cyclic ADP ribose hydrolase activities. Increased expression of BST1 in rheumatoid arthritis (RA) suggests its involvement in the disease, particularly through its elevated presence in RA-derived bone marrow stromal cell lines. BST1 is also found on myeloid lineage cells and is thought to function as a receptor with signal transduction capabilities.
Produced in Sf9 insect cells using a baculovirus expression system, BST1 is a single-chain polypeptide containing 267 amino acids (residues 33-293). A 6-amino acid Histidine tag is present at the C-terminus to facilitate purification. The protein has a molecular weight of 30.5 kDa, but due to glycosylation, it appears as a band of approximately 40-57 kDa on SDS-PAGE. Purification is achieved using proprietary chromatographic techniques.
The product is a clear, colorless, and sterile-filtered solution.
BST1 is supplied as a 0.5 mg/mL solution in phosphate-buffered saline (pH 7.4) containing 10% glycerol.
The purity of BST1 is greater than 95%, as determined by SDS-PAGE analysis.
Bone Marrow Stromal Cell Antigen 1, ADP-Ribosyl Cyclase 2, Bone Marrow Stromal Antigen 1, Cyclic ADP-Ribose Hydrolase 2, NAD(+) Nucleosidase, CADPr Hydrolase 2, ADP-Ribosyl Cyclase/Cyclic ADP-Ribose Hydrolase 2, CD157 Antigen, EC 3.2.2.6, CD157, BST-1, ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 2, ADP-ribosyl cyclase 2, Bone marrow stromal antigen 1, Cyclic ADP-ribose hydrolase 2, cADPr hydrolase 2.
Sf9, Baculovirus cells.
RWRGEGTSAH LRDIFLGRCA EYRALLSPEQ RNKNCTAIWE AFKVALDKDP CSVLPSDYDL FINLSRHSIP RDKSLFWENS HLLVNSFADN TRRFMPLSDV LYGRVADFLS WCRQKNDSGL DYQSCPTSED CENNPVDSFW KRASIQYSKD SSGVIHVMLN GSEPTGAYPI KGFFADYEIP NLQKEKITRI EIWVMHEIGG PNVESCGEGS MKVLEKRLKD MGFQYSCIND YRPVKLLQCV DHSTHPDCAL KSAAAATQRK AHHHHHH.
BST1 (bone marrow stromal cell antigen 1), also known as CD157, is a stromal cell line-derived glycosylphosphatidylinositol-anchored molecule that serves important biological functions. Originally identified as a factor that facilitates pre-B-cell growth, BST1 exhibits approximately 33% amino acid sequence similarity with CD38, suggesting evolutionary and functional relationships between these molecules. The protein is encoded by the BST1 gene located on chromosome band 4p15.32 in humans. BST1 functions in multiple physiological processes including immune response regulation, cell adhesion, and migration. In neural tissues, BST1 appears to play roles in social behavior regulation and neuronal signaling pathways that may impact neurodevelopmental outcomes. Research methodologies for studying BST1's molecular functions typically include protein interaction studies, enzymatic activity assays, and gene expression analyses across different tissue types and developmental stages .
BST1 expression regulation involves complex mechanisms that vary across different human tissues and developmental stages. The gene demonstrates tissue-specific expression patterns, with notable presence in immune cells, bone marrow stromal cells, and certain neuronal populations. Transcriptional regulation of BST1 involves multiple transcription factors and epigenetic modifications that respond to various cellular stimuli. When designing experiments to study BST1 expression, researchers should consider implementing tissue-specific sampling protocols, developmental time-course analyses, and comparative expression studies between neurotypical individuals and those with conditions like autism spectrum disorder. Methodologically, quantitative PCR, RNA sequencing, and immunohistochemistry represent the gold-standard approaches for analyzing BST1 expression patterns. For epigenetic regulation studies, techniques such as bisulfite sequencing and chromatin immunoprecipitation provide insights into the methylation status and histone modifications of the BST1 promoter region. When interpreting expression data, researchers should account for potential confounding variables including tissue heterogeneity, age-related changes, and the influence of environmental factors on gene expression .
Selecting appropriate experimental models for BST1 research requires careful consideration of the specific research questions and translational goals. Animal models, particularly BST1-knockout mice, have proven valuable for investigating the behavioral and physiological consequences of BST1 deficiency. These mouse models demonstrate phenotypes including anxiety-related behaviors, depression-like symptoms, and social avoidance that can be partially rescued with pharmacological interventions such as oxytocin administration. When establishing BST1-knockout models, researchers should implement comprehensive behavioral phenotyping protocols that assess multiple domains including social interaction, anxiety, depression-like behaviors, and cognitive function. For cellular models, both primary cultures and cell lines expressing BST1 can be utilized to investigate molecular mechanisms. iPSC-derived neurons from individuals with BST1 variants offer particularly valuable insights into human-specific aspects of BST1 function. When designing experiments with these models, researchers should incorporate appropriate controls, consider developmental timing effects, and validate findings across multiple model systems to strengthen translational relevance .
The association between BST1 and autism spectrum disorders (ASD) is supported by both genetic and functional evidence. Significant genetic evidence comes from a case-control study in a Japanese population that identified two intronic single nucleotide polymorphisms (SNPs) in BST1 with substantially higher frequencies in individuals with ASD compared to controls. Specifically, rs4301112 showed an odds ratio of 6.4 (p=0.0007) and rs28532698 showed an odds ratio of 6.2 (p=0.0012), with both associations remaining significant after multiple testing correction. Further supporting this genetic link, a deletion involving both CD38 and BST1 resulting in a fusion transcript was identified in a patient with autism and asthma. To properly investigate BST1 variants in research settings, established protocols should include careful phenotyping, consideration of population stratification, and replication in diverse cohorts. Functional evidence comes from BST1-knockout mouse models that display behaviors relevant to ASD, including anxiety-related behaviors and social avoidance. Importantly, these behaviors can be rescued by oxytocin administration, suggesting potential therapeutic avenues. When interpreting these findings, researchers should recognize both the strengths and limitations of current evidence, including the need for larger genetic studies across diverse populations and more extensive functional characterization .
BST1's SFARI Gene score provides a standardized assessment of its relevance to autism spectrum disorders based on rigorous evaluation criteria. As of April 2022, BST1 holds a SFARI Gene Score of 2, classifying it as a "Strong Candidate" gene for autism. This scoring represents an evolution from previous evaluations – BST1 was scored as category 4 prior to April 2017, was elevated to category 3 in October 2019 under a new scoring scheme, and was subsequently upgraded to category 2 in April 2022. The current classification indicates that while BST1 has not achieved the highest level of evidence (which would require genome-wide statistical significance with independent replication), it meets slightly relaxed criteria that still represent strong evidence for association with autism. The scoring system methodology considers multiple lines of evidence including genetic associations, functional studies, and animal models. When interpreting SFARI scores for research priorities, investigators should recognize that these classifications are regularly updated as new evidence emerges and that current scores reflect cumulative evidence rather than individual study results. The systematic evaluation represented by the SFARI score provides researchers with a standardized framework for prioritizing genes for further investigation .
Time Period | SFARI Gene Score | Category Description |
---|---|---|
Before April 2017 | 4 | Minimal evidence |
April 2017 - September 2019 | 4 | Minimal evidence |
October 2019 - March 2022 | 3 | Suggestive evidence |
April 2022 - Present | 2 | Strong candidate |
Detecting BST1 variants in autism research requires sophisticated methodological approaches tailored to specific research questions. For comprehensive variant detection, whole-genome or whole-exome sequencing represents the gold standard, capable of identifying both common and rare variants throughout the gene. When targeting known variants like the significant intronic SNPs rs4301112 and rs28532698, custom genotyping arrays or targeted sequencing approaches offer cost-effective alternatives. Copy number variant detection, important given the reported case of a deletion involving BST1, should employ techniques such as chromosomal microarray analysis or read-depth analysis of sequencing data. For analyzing complex structural variants, long-read sequencing technologies provide advantages in resolving intricate genomic rearrangements. When implementing these approaches, researchers should adhere to rigorous quality control procedures including assessment of sequencing coverage, variant calling accuracy, and population stratification in case-control studies. Methodological limitations to consider include the challenges of detecting variants in low-complexity regions, the potential for technical artifacts, and the importance of functional validation for variants of uncertain significance. The integration of multiple techniques often provides the most comprehensive profile of BST1 variation in research settings .
Single-subject research designs offer valuable approaches for investigating BST1 function, particularly when examining interventions that target BST1-related pathways. When implementing such designs, researchers should adhere to established methodological principles including reliable measurement of the target behavior or biological outcome, operational definitions of all variables, sufficient measurements during each time frame to establish stability (minimum of 4 measurements), and comprehensive description of procedures. The A-B-A or A-B-A-B designs represent particularly robust approaches, where "A" represents baseline conditions and "B" represents treatment or intervention phases. For BST1-specific research, these designs can be effectively applied to test interventions such as oxytocin administration, which has shown promise in rescuing behavioral phenotypes in BST1-knockout mice. The multiple baseline approach, analyzing more than one subject, behavior, or setting, can strengthen external validity while maintaining the individualized focus of single-subject design. When analyzing results, researchers should evaluate both statistical and practical significance, with particular attention to the duration and stability of treatment effects. Key limitations to address include potential experimenter or observer effects and the need for control of confounding variables. Single-subject designs complement group-based approaches by providing detailed insights into individual variation in response to BST1-targeted interventions .
Designing appropriate control conditions for BST1 knockout studies is critical for valid interpretation of results. Complete knockout designs should incorporate multiple control groups including wild-type littermates, heterozygous animals to assess gene dosage effects, and when appropriate, conditional knockout models that restrict gene deletion to specific tissues or developmental windows. Beyond genotype controls, procedural controls should include sham procedures matching any manipulations performed on experimental animals, vehicle administration controls for pharmacological rescue experiments, and handling controls to account for stress effects. Environmental standardization across experimental and control groups is essential, with particular attention to factors known to affect BST1-related phenotypes such as social housing conditions and enrichment protocols. When implementing rescue experiments, such as oxytocin administration to BST1-knockout mice, researchers should include dose-response assessments and timing variations to establish optimal parameters. For behavioral studies, blinded assessment protocols prevent observer bias, while automated systems offer additional objectivity. The interpretation of results should consider potential developmental compensation mechanisms in constitutional knockout models and acknowledge that phenotypic differences may result from complex downstream effects rather than direct BST1 function. Statistical analysis plans should pre-specify primary and secondary outcomes to avoid post-hoc selection biases .
Integrating BST1 genomic data with functional outcomes requires sophisticated methodological approaches spanning multiple research domains. The research workflow should begin with comprehensive genetic variant identification through sequencing or genotyping, followed by bioinformatic prediction of variant effects using algorithms that assess conservation, protein structure impacts, and potential regulatory consequences. Functional validation should then proceed across multiple levels including cellular models examining protein function, animal models assessing behavioral and physiological phenotypes, and when possible, human studies correlating genotype with phenotype. Specific integration methodologies include expression quantitative trait loci (eQTL) analyses to connect variants with expression differences, pathway analyses to situate BST1 within broader biological networks, and longitudinal designs that capture developmental trajectories. When applying these approaches to autism research, investigators should consider implementing statistical methods that account for genetic background effects, environmental interactions, and phenotypic heterogeneity. Data integration platforms and collaborative research consortia can facilitate the combination of genomic, molecular, and clinical datasets from multiple sources. The interpretation of integrated data should acknowledge limitations including the often indirect relationship between genotype and phenotype, the challenges of translating between model systems and humans, and the potential for publication bias in available literature .
Analyzing conflicting data regarding BST1 variants across different populations requires a systematic approach that accounts for various sources of heterogeneity. The fundamental methodological framework should begin with critical evaluation of study designs, considering factors such as sample size, ascertainment criteria, diagnostic instruments, and genotyping or sequencing technologies. Meta-analytic approaches can integrate findings across studies while accounting for between-study heterogeneity, with random-effects models generally preferred for genetic association studies. When population-specific effects are suspected, as suggested by the strong association of specific BST1 SNPs in Japanese populations but potential differences in other groups, stratified analyses by ancestry should be conducted with appropriate correction for population structure. The investigation of gene-environment interactions and analysis of different inheritance models (additive, dominant, recessive) may help reconcile apparently discordant findings. For comprehensive assessment, researchers should implement pathway-based analyses that consider BST1 in the context of functionally related genes, potentially revealing consistent effects at the pathway level despite variant-level differences. When interpreting conflicting data, researchers should acknowledge limitations including publication bias favoring positive findings, inconsistent phenotyping approaches across studies, and technical artifacts. The development of standardized analytic protocols and data sharing initiatives can facilitate more robust cross-population comparisons .
Appropriate statistical approaches for analyzing the relationship between BST1 variants and autism phenotypes must address both the genetic complexity and phenotypic heterogeneity characteristic of autism spectrum disorders. Case-control association testing represents the foundational approach for identifying significant variants, as demonstrated in the identification of rs4301112 and rs28532698 as risk variants in Japanese populations. These analyses should implement rigorous multiple testing correction procedures, such as the Bonferroni method or false discovery rate control, to minimize false positives. Beyond basic association, quantitative trait analyses linking BST1 variants to specific dimensional phenotypes (social communication deficits, restricted/repetitive behaviors) can reveal more nuanced genotype-phenotype relationships. Advanced statistical methodologies include structural equation modeling to assess complex pathways linking genetic variation to clinical outcomes, machine learning approaches to identify patterns across multiple variables, and Bayesian methods that can incorporate prior biological knowledge. Longitudinal modeling can capture developmental trajectories and potential age-dependent effects of BST1 variants. When applying these methods, researchers should implement appropriate adjustments for covariates including sex, age, intellectual functioning, and comorbidities. Sample size considerations are critical, with power analyses conducted a priori to ensure sufficient statistical power to detect anticipated effect sizes. The interpretation of results should acknowledge statistical limitations including multiple comparison issues, the potential for spurious correlations, and challenges in distinguishing causation from association .
Quantifying the translational value of BST1 mouse models requires systematic evaluation of construct, face, and predictive validity using standardized metrics and comparative analyses. Construct validity assessment should examine the genetic and molecular similarity between mouse models and human conditions, including sequence homology analysis, comparative expression patterns across tissues, and evaluation of downstream molecular pathways. Currently available data indicate substantial conservation of BST1 function between mice and humans, supporting basic construct validity. Face validity evaluation requires systematic comparison of behavioral phenotypes between BST1-knockout mice and human autism presentations, implementing standardized behavioral assays that assess domains relevant to autism including social interaction, repetitive behaviors, and communication differences. The documented anxiety-related behaviors, depression-like symptoms, and social avoidance in BST1-knockout mice partially align with human autism phenotypes, though with important differences that must be acknowledged. Predictive validity can be quantified through systematic testing of intervention responses, such as the demonstrated efficacy of oxytocin in rescuing social deficits in BST1-knockout mice, followed by translational testing in human studies. Statistical approaches for quantifying translational alignment include correlation analyses between mouse and human phenotypic measures, regression models predicting human outcomes from mouse data, and systematic reviews with standardized effect size calculations across species. When interpreting these analyses, researchers should acknowledge the inherent limitations of animal models including species differences in neural circuitry, the inability to model subjective aspects of human conditions, and the simplified genetic background of laboratory mice compared to human genetic heterogeneity .
Based on current evidence, several promising therapeutic targets related to BST1 dysfunction in autism warrant further investigation through systematic research programs. Oxytocin pathway modulation represents perhaps the most immediate opportunity, building on evidence that oxytocin administration rescues social avoidance behavior in BST1-knockout mice. Research protocols should include dose-optimization studies, long-term administration paradigms, and investigation of oxytocin receptor expression patterns in BST1-deficient models. Beyond direct oxytocin administration, development of compounds that enhance endogenous oxytocin release or potentiate receptor signaling may provide alternative therapeutic approaches. Additional promising targets include downstream signaling pathways affected by BST1 dysfunction, particularly those involved in calcium signaling and cyclic ADP-ribose metabolism given BST1's enzymatic functions. Anti-psychiatric medications represent another avenue worth exploring, as these have shown efficacy in ameliorating anxiety-related and depression-like behaviors in BST1-knockout mice. For all potential therapeutics, research should progress through systematic preclinical validation before advancing to carefully designed clinical trials with clearly defined target populations and outcome measures. When designing such studies, researchers should implement stratification approaches that account for genetic heterogeneity, consider developmental timing of interventions, and incorporate biomarkers that can track molecular and physiological responses to treatment. Comprehensive safety monitoring is essential, particularly for interventions targeting developmentally sensitive pathways .
Multi-omics approaches offer powerful methodologies for comprehensively investigating BST1's role in human neurodevelopment through systematic integration of data across biological levels. The implementation of genomics, transcriptomics, proteomics, metabolomics, and epigenomics in coordinated research programs can reveal the complex networks through which BST1 influences neural development and function. Genomic approaches should extend beyond standard genotyping to include whole-genome sequencing for comprehensive variant detection, chromosome conformation capture technologies to identify long-range interactions affecting BST1 regulation, and CRISPR screening to systematically assess functional impacts of variants. Transcriptomic analyses should examine BST1 expression patterns across developmental trajectories, cell types, and in response to environmental stimuli, ideally at single-cell resolution to capture cellular heterogeneity. Proteomic investigations should map BST1 protein interaction networks, post-translational modifications, and changes in protein abundance across development. Metabolomic profiling can identify downstream metabolic consequences of BST1 dysfunction, while epigenomic analyses can reveal regulatory mechanisms controlling BST1 expression in neural tissues. When implementing multi-omics approaches, researchers should develop sophisticated data integration methods including network analyses, machine learning algorithms for pattern discovery, and systems biology models that capture dynamic relationships. Study designs should include carefully selected developmental timepoints, relevant tissue and cell types, and appropriate control samples. The interpretation of multi-omics data should acknowledge technical challenges including different noise levels across platforms, the complexity of integrating diverse data types, and the need for extensive validation of computational predictions .
The dual association of BST1 with both autism spectrum disorders and Parkinson's disease presents a complex picture requiring careful interpretative frameworks. Mechanistically, several hypotheses warrant systematic investigation: shared biological pathways affected by different BST1 variants, distinct functional domains of BST1 that impact different aspects of neural function, developmental timing differences where the same molecular pathways produce different outcomes depending on when disruption occurs, or pleiotropy where the same genetic variation produces different phenotypes depending on genetic background and environmental factors. Research methodologies to investigate these possibilities should include comparative genetic studies examining whether the same or different variants associate with each condition, functional characterization of variants in relevant cell types and developmental stages, and longitudinal studies tracking individuals with BST1 variants across the lifespan. The potential neurobiological intersection between these conditions may involve immune system regulation, calcium signaling pathways, or synaptic plasticity mechanisms. When designing studies to explore these connections, researchers should implement parallel phenotyping protocols in both conditions, investigate age-dependent effects, and consider shared endophenotypes that may bridge diagnostic categories. The clinical implications of this dual association include the potential for shared therapeutic approaches, the need for monitoring for symptoms across conditions in affected individuals, and opportunities for insights into fundamental neurobiological processes. Interpretative frameworks should acknowledge the complexity of genotype-phenotype relationships, recognize the limitations of current diagnostic categories, and consider evolutionary perspectives on why such variants persist in populations .
Ethical implementation of BST1 testing in both clinical and research contexts requires careful consideration of multiple dimensions through systematic frameworks and established guidelines. For clinical testing, key considerations include the uncertain predictive value of BST1 variants given the complex genetic architecture of associated conditions, appropriate pre- and post-test genetic counseling protocols, and procedures for handling incidental findings such as variants associated with Parkinson's disease risk when testing was conducted for autism-related purposes. Research testing raises additional considerations including informed consent procedures that clearly communicate the exploratory nature of BST1 research, data sharing policies that balance open science principles with privacy protections, and community engagement to ensure research priorities align with stakeholder needs. When designing testing protocols for vulnerable populations, particularly children and individuals with cognitive disabilities, researchers should implement additional safeguards including heightened scrutiny of risk-benefit ratios, assent procedures alongside guardian consent, and mechanisms to ensure findings are translated into tangible benefits for participants. Return of results policies should be carefully developed, with clear guidelines on which findings meet clinical validity and utility thresholds for disclosure. Specific methodological approaches to address these ethical considerations include the use of tiered consent models, establishment of community advisory boards, development of accessible educational materials about BST1 testing, and creation of governance structures for ongoing oversight of data use. The implementation of these approaches should acknowledge broader societal implications including potential stigmatization, insurance discrimination concerns, and the importance of diverse population representation in research .
Bone Marrow Stromal Cell Antigen 1 (BST1), also known as CD157, is a glycosylphosphatidylinositol (GPI)-anchored molecule that plays a significant role in the regulation of pre-B cell growth. It is a member of the ADP-ribosyl cyclase family of enzymes, which are involved in the metabolism of nicotinamide adenine dinucleotide (NAD+). BST1 is encoded by the BST1 gene located on chromosome 4 in humans .
BST1 is structurally similar to CD38, another member of the ADP-ribosyl cyclase family, with a 33% similarity in their amino acid sequences . The enzyme catalyzes the formation of cyclic ADP-ribose (cADPR) from NAD+, although it is a weaker catalyst compared to CD38 . cADPR is crucial for the regulation of calcium ion (Ca2+) signaling within cells .
BST1 is predominantly expressed in bone marrow stromal cells, particularly in those derived from patients with rheumatoid arthritis . The overexpression of BST1 in these cells is believed to contribute to the polyclonal B cell abnormalities observed in rheumatoid arthritis . Additionally, BST1 is expressed in various other tissues, including monocytes, granulocytes, and certain epithelial cells .
BST1 has several biological functions, including:
The expression of BST1 is enhanced in bone marrow stromal cell lines derived from patients with rheumatoid arthritis, suggesting a potential role in the pathogenesis of this autoimmune disease . Additionally, BST1 has been implicated in various other conditions, including certain cancers and inflammatory diseases .