Bone Morphogenetic Protein Receptor-1A Human Recombinant produced in CHO cells is a glycosylated homodimer chain containing 2x362 amino acids and having a total molecular mass of 80.8kDa.
BMPR1A is purified by proprietary chromatographic techniques.
The bone morphogenetic protein (BMP) receptors are transmembrane serine/threonine kinases. This family includes type I receptors BMPR1A and BMPR1B, and the type II receptor BMPR2. These receptors are closely related to ACVR1 and ACVR2 receptors. Ligands for these receptors belong to the TGF-beta superfamily. TGF-betas signal through heteromeric complexes formed with two types of serine (threonine) kinase receptors: type I receptors (approximately 50-55 kD) and type II receptors (approximately 70-80 kD). Type II receptors can bind ligands independently but require type I receptors for signaling. Conversely, type I receptors need their corresponding type II receptors for ligand binding.
Recombinant Human Bone Morphogenetic Protein Receptor-1A, produced in CHO cells, is a glycosylated homodimer. It consists of two chains, each containing 362 amino acids, resulting in a total molecular mass of 80.8kDa.
Purification of BMPR1A is achieved using proprietary chromatographic techniques.
The protein solution undergoes sterile filtration (0.2µm) and is then lyophilized in a buffer containing PBS.
For reconstitution, it is recommended to dissolve the lyophilized BMPR1A in sterile 18MΩ-cm H2O at a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Lyophilized BMPR1A remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated at a temperature below -18°C. After reconstitution, BMPR1A should be stored at 4°C for 2-7 days. For future use, store at -18°C.
Avoid repeated freeze-thaw cycles.
The purity is determined to be greater than 95.0% as assessed by SDS-PAGE.
The ED50, determined by measuring the inhibition of human BMP-4-induced alkaline phosphatase production in ATDC5 cells, is 120ng/ml. This corresponds to a specific activity of 8.3 x 10^3 units/mg.
BMPR-1A, BMP-R1A, BMPR1A, BMR1A, CD292, CD-292, Serine/threonine-protein kinase receptor R5, SKR5, ALK-3, ACVRLK3, EC 2.7.11.30, CD292 antigen.
CHO cells.
QNLDSMLHGT GMKSDSDQKK SENGVTLAPE DTLPFLKCYC SGHCPDDAIN NTCITNGHCF AIIEEDDQGE TTLASGCMKY EGSDFQCKDS PKAQLRRTIE CCRTNLCNQY LQPTLPPVVI GPFFDGSIRI EGRMDDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK.
BMPR1A is a cell surface receptor that functions as a type I receptor in the Bone Morphogenetic Protein (BMP) signaling pathway. This receptor plays crucial roles in multiple physiological processes including skeletal development, vascular homeostasis, and cellular differentiation. BMPR1A is essential for maintaining endothelial cell identity by modulating Tgfbr2 expression through an ID2/ZEB1-dependent mechanism. The receptor forms heterodimeric complexes with BMP type II receptors to activate downstream signaling cascades via Smad-dependent and Smad-independent pathways. In endothelial cells, BMPR1A activation leads to ID2 physically interacting with and sequestering ZEB1, which attenuates the transcription of Tgfbr2, thereby preventing excessive endothelial-mesenchymal transition (EndoMT) .
The transcriptional regulation of BMPR1A involves multiple transcription factor binding sites (TFBS) within its promoter region. Analysis of the BMPR1A promoter has identified Sp1 as a critical transcription factor regulating its expression. Research using deletion fragments of the BMPR1A promoter in luciferase reporter vectors has demonstrated that Sp1 binding sites at positions 160, 214, and 599 significantly contribute to promoter activity. Additionally, an ETSF site at position 603 and an NFKB site at position 188 have been shown to influence BMPR1A transcription. Site-directed mutagenesis of these binding sites results in substantial reduction of promoter activity, confirming their functional importance in regulating BMPR1A expression .
BMPR1A dysfunction has been implicated in several significant pathologies:
Pulmonary Arterial Hypertension (PAH): Decreased BMPR1A expression has been observed in the lungs of PAH patients. Deletion of endothelial Bmpr1a in mice leads to the development of PAH-like symptoms due to excessive EndoMT .
Juvenile Polyposis (JP): Mutations in BMPR1A predispose individuals to JP, which carries a >50% lifetime risk of colorectal cancer development. This suggests that defective BMPR1A production plays a central role in this oncogenic process .
Bone Disorders: BMPR1A is crucial for bone homeostasis, and its dysregulation contributes to bone-related disorders .
Tumor Formation: In certain contexts, BMPR1A appears to promote tumorigenesis. Studies in mouse models have shown that BMPR1A-knockout transformed astrocytes exhibit reduced tumor formation capacity and less aggressive growth compared to BMPR1A-intact cells .
To analyze BMPR1A promoter activity, researchers can employ the following methodological approach:
For optimal expression of human BMPR1A in CHO cells, researchers should implement the following protocol:
Vector Selection: Choose a mammalian expression vector with a strong promoter (CMV or EF1α) and appropriate selection markers (e.g., neomycin resistance).
Gene Optimization: Optimize the human BMPR1A coding sequence for CHO cell expression by adjusting codon usage and eliminating potential cryptic splice sites.
Transfection Method: Use either liposome-based transfection reagents (e.g., Lipofectamine) or electroporation to introduce the expression vector into CHO cells. For stable expression, consider using the Flp-In system or similar site-specific integration approaches.
Selection and Screening: Apply appropriate selection pressure (G418 for neomycin resistance) and isolate single-cell clones. Screen clones for BMPR1A expression using Western blotting or flow cytometry.
Culture Optimization: Determine optimal growth conditions including media composition, serum requirements, and culture duration to maximize protein expression. For recombinant protein production, consider adapting cells to suspension culture in serum-free media.
Protein Verification: Confirm proper folding and functionality of expressed BMPR1A using ligand binding assays with BMP2/4 or analyzing downstream signaling activation via phospho-Smad detection.
To evaluate BMPR1A-mediated signaling in cellular models, researchers can employ several complementary techniques:
Phospho-Smad Analysis: Western blotting to detect phosphorylation of Smad1/5/8, which indicates canonical BMP signaling activation. This should be performed with appropriate time courses following BMP ligand stimulation.
Transcriptional Reporter Assays: Utilize BMP-responsive element (BRE) luciferase reporters to quantify transcriptional activation downstream of BMPR1A signaling.
qRT-PCR Analysis: Measure expression changes in BMP target genes like ID1, ID2, and ID3 following receptor activation or inhibition.
Protein-Protein Interaction Studies: Employ co-immunoprecipitation or proximity ligation assays to detect interactions between BMPR1A and its binding partners, including BMPR2, ID2, and ZEB1.
Functional Assays: Assess biological outcomes of BMPR1A signaling using:
Endothelial barrier function assays
Cell migration and invasion assays
Proliferation assays
Differentiation markers
Pharmacological Manipulation: Use selective BMPR1A inhibitors (e.g., DMH1) to confirm receptor-specific effects .
The BMPR1A-ID2-ZEB1 signaling axis represents a sophisticated regulatory mechanism that maintains endothelial cell identity and prevents excessive endothelial-mesenchymal transition (EndoMT). This process operates through the following molecular mechanisms:
BMPR1A Activation and ID2 Induction: When BMPR1A is activated by BMP ligands, it triggers intracellular signaling that increases expression of ID2 (Inhibitor of DNA binding 2).
ID2-ZEB1 Physical Interaction: ID2 physically interacts with and sequesters ZEB1 (Zinc finger E-box-binding homeobox 1), a transcription factor that promotes mesenchymal transition.
Tgfbr2 Transcriptional Regulation: The ID2-ZEB1 interaction attenuates the transcription of Tgfbr2 (TGF-β receptor type II), which is crucial because:
Reduced Tgfbr2 expression lowers endothelial cell responsiveness to TGF-β stimulation
This reduced TGF-β sensitivity prevents excessive EndoMT
Pathological Consequences of Disruption: When BMPR1A is deleted or downregulated in endothelial cells, this protective mechanism fails:
ID2 levels decrease, allowing ZEB1 to become active
ZEB1 increases Tgfbr2 expression
Enhanced TGF-β signaling drives EndoMT
This leads to pathologies such as pulmonary arterial hypertension (PAH)
In mouse models, endothelial-specific deletion of Bmpr1a (Bmpr1a iECKO) leads to PAH-like symptoms due to excessive EndoMT. Notably, administering endothelial-targeting lipid nanoparticles containing siRNA against Tgfbr2 effectively ameliorates these symptoms, confirming the central role of this signaling axis .
BMPR1A exhibits context-dependent functional effects that sometimes appear contradictory across different tissue types and disease models:
These apparent contradictions can be explained by:
Differential Downstream Effector Activation: BMPR1A may preferentially activate different Smad proteins or non-Smad pathways depending on the cellular context and receptor complex composition.
Interaction with Tissue-Specific Transcription Factors: BMPR1A-initiated signaling interacts with tissue-specific transcription factors, resulting in context-dependent gene expression patterns.
Balance with Other Signaling Pathways: The net effect of BMPR1A signaling depends on its integration with other pathways like TGF-β, Wnt, and Notch, which vary across tissues.
Developmental Stage Effects: BMPR1A may have different roles during development versus tissue homeostasis in adult organisms.
Understanding these context-dependent effects is crucial for developing targeted therapeutic strategies that modulate BMPR1A signaling in specific tissues without unwanted effects in others .
Optimizing recombinant BMPR1A-Fc fusion proteins for experimental applications requires attention to several key factors:
Construct Design Considerations:
Include the extracellular domain of BMPR1A (amino acids 1-152) while excluding the transmembrane and intracellular domains
Link the BMPR1A domain to the Fc portion of human or mouse IgG1 via a flexible linker (e.g., (Gly4Ser)3)
Add purification tags (His, FLAG) that can be enzymatically removed if needed
Incorporate codon optimization for the expression system
Expression System Selection:
CHO cells typically yield properly folded and glycosylated BMPR1A-Fc with higher biological activity than prokaryotic systems
Establish stable cell lines rather than relying on transient expression for consistent production
Consider using GS-CHO or DHFR-CHO systems with gene amplification for higher yields
Implement serum-free suspension culture protocols to simplify purification
Purification Protocol Optimization:
Employ protein A/G affinity chromatography for initial capture
Follow with ion exchange and size exclusion chromatography for higher purity
Validate protein integrity via SDS-PAGE, Western blot, and mass spectrometry
Confirm proper disulfide bond formation and glycosylation pattern
Functional Validation Methods:
Assess BMP ligand binding using surface plasmon resonance (SPR)
Confirm the ability to inhibit BMP signaling in cell-based assays (phospho-Smad1/5/8 inhibition)
Evaluate dose-dependent effects on BMP-responsive gene expression (ID1, ID2)
Test stability under experimental conditions (temperature, pH, freeze-thaw cycles)
Storage and Handling:
Determine optimal buffer composition (typically PBS with 5-10% glycerol)
Establish appropriate aliquoting, storage temperature (-80°C), and freeze-thaw protocols
Validate long-term stability and activity retention
Studies have shown that soluble BMPR1A-Fc specifically binds BMP2/4 with high affinity and prevents downstream signaling. When administered systemically in mouse models, it increases bone mineral density, cortical thickness, and trabecular bone volume, demonstrating its potential as both an experimental tool and therapeutic agent .
Researchers conducting BMPR1A knockout studies frequently encounter several challenges that can impact experimental outcomes and interpretation:
Embryonic Lethality:
Compensatory Mechanisms:
Heterogeneous Recombination Efficiency:
Problem: Incomplete Cre-mediated recombination creates mosaic tissues with mixed BMPR1A-positive and negative cells.
Solution: Incorporate reporter genes (e.g., mTomato) to track recombination efficiency; use FACS to isolate successfully recombined cells; validate knockout at protein level by immunohistochemistry .
Context-Dependent Phenotypes:
Interpretation Challenges in Complex Tissues:
When working with BMPR1A expression in CHO systems, researchers can address common challenges through the following approaches:
Low Expression Levels:
Implement gene amplification strategies (e.g., MTX selection for DHFR system)
Screen multiple clones for high expressers using high-throughput methods
Optimize culture media components and feed strategies
Consider using stronger promoters or enhancer elements
Test different signal peptide sequences to improve secretion efficiency
Protein Misfolding and Aggregation:
Reduce expression temperature (30-32°C) during production phase
Add chemical chaperones to culture media (e.g., DMSO, glycerol)
Optimize oxidizing conditions for proper disulfide bond formation
Incorporate fusion partners that enhance solubility (e.g., SUMO, thioredoxin)
Evaluate different cell lysis buffers and conditions if producing intracellular protein
Proteolytic Degradation:
Add protease inhibitors during all purification steps
Reduce production time to minimize exposure to endogenous proteases
Identify and mutate susceptible protease cleavage sites without affecting function
Consider using protease-deficient CHO cell lines
Glycosylation Heterogeneity:
Characterize glycosylation profiles using mass spectrometry
Implement glycoengineered CHO cell lines for more homogeneous glycans
Consider enzymatic deglycosylation if glycans interfere with function
Use site-directed mutagenesis to remove non-essential glycosylation sites
Purification Challenges:
Develop multi-step purification strategies combining affinity, ion exchange, and size exclusion methods
Optimize buffer conditions (pH, salt concentration) for each purification step
Implement tangential flow filtration for efficient concentration without aggregation
Validate final product quality using multiple analytical techniques (SEC-MALS, CD spectroscopy, activity assays)
Reconciling contradictory findings about BMPR1A signaling requires systematic analysis of experimental variables:
Cell Type and Context Dependencies:
Analysis Method: Perform comparative studies using multiple cell types under identical experimental conditions.
Reconciliation Strategy: Map cell type-specific receptor complexes, co-receptors, and intracellular mediators that may explain differential responses.
Example Finding: BMPR1A deletion in osteoblasts increases bone mass , while in endothelial cells it leads to pathological EndoMT , illustrating tissue-specific outcomes of the same signaling pathway.
Ligand Specificity and Concentration Effects:
Analysis Method: Generate dose-response curves across a wide concentration range for multiple BMP ligands.
Reconciliation Strategy: Identify threshold effects and biphasic responses that may explain contradictory results obtained at single concentrations.
Key Consideration: Different BMP ligands (BMP2, BMP4, BMP7) may preferentially activate BMPR1A in combination with different type II receptors, leading to diverse signaling outcomes.
Acute versus Chronic Manipulation:
Analysis Method: Compare transient knockdown/inhibition with stable knockout/long-term inhibition.
Reconciliation Strategy: Document temporal changes in compensatory mechanisms and adaptive responses.
Observed Pattern: Short-term BMPR1A inhibition may yield different results than hereditary or long-term experimental BMPR1A deficiency due to compensatory upregulation of alternative pathways.
In Vitro versus In Vivo Discrepancies:
Analysis Method: Systematically compare cellular responses in isolated cells versus the same cells in their native tissue environment.
Reconciliation Strategy: Identify microenvironmental factors that modify BMPR1A signaling outcomes in vivo.
Example: Studies with transformed astrocytes showed that BMPR1A promotes cell proliferation and invasion in vitro, which correlates with more aggressive tumor formation in vivo .
Technical Variations in Experimental Approach:
Analysis Method: Directly compare genetic deletion, siRNA knockdown, and pharmacological inhibition approaches.
Reconciliation Strategy: Establish minimum criteria for concluding BMPR1A involvement (e.g., consistent results from at least two independent methods).
Practical Approach: Create standardized positive and negative controls that can be shared between laboratories to calibrate experimental systems.
Several cutting-edge technologies are poised to transform BMPR1A research and provide unprecedented insights:
CRISPR Base Editing and Prime Editing:
Beyond conventional knockout approaches, these technologies enable precise modification of BMPR1A regulatory elements or the introduction of specific disease-associated mutations
Applications include creating isogenic cell lines with patient-specific BMPR1A variants to study their functional consequences
Spatial Transcriptomics and Proteomics:
These techniques allow visualization of BMPR1A expression patterns and downstream signaling activation with spatial resolution in tissues
Can reveal microenvironmental influences on BMPR1A signaling and identify cell populations with unique receptor expression profiles
Cryo-EM and AlphaFold2 Structural Analysis:
High-resolution structural studies of BMPR1A-ligand complexes in different conformational states
Computational prediction of protein-protein interaction interfaces to guide drug design targeting specific BMPR1A interactions
Identification of allosteric regulatory sites for selective modulation
Organoid and Microphysiological Systems:
Development of tissue-specific organoids from primary cells with defined BMPR1A genotypes
Integration of multiple tissue types in organ-on-chip platforms to study cross-talk between BMPR1A signaling in different tissues
Potential for personalized drug screening using patient-derived cells
Single-Cell Multi-Omics:
Single-cell RNA-seq combined with ATAC-seq and proteomics to map cell state transitions controlled by BMPR1A
Identification of rare cell populations with unique BMPR1A-dependent phenotypes
Trajectory analysis to reveal how BMPR1A signaling influences cell fate decisions
The BMPR1A-ID2-ZEB1 signaling axis represents a promising therapeutic target with several potential intervention strategies:
Selective BMPR1A Agonists and Antagonists:
Context-specific modulation of BMPR1A activity could address pathological conditions:
Agonists for pulmonary arterial hypertension to prevent EndoMT
Antagonists for certain cancer types where BMPR1A promotes tumorigenesis
Small molecule compounds or engineered ligands with tissue-selective distribution profiles would help avoid unwanted effects in non-target tissues
ID2-ZEB1 Interaction Modulators:
Stabilizing the ID2-ZEB1 interaction could prevent excessive EndoMT in pulmonary hypertension
Peptide-based therapeutics mimicking the ID2-ZEB1 interface could provide highly specific intervention
High-throughput screening of small molecule libraries could identify compounds that enhance ID2-ZEB1 binding
RNA-Based Therapeutics:
Combination Approaches:
Targeting BMPR1A signaling in combination with other pathways (e.g., TGF-β, Notch, Wnt) may provide synergistic effects
This could allow for lower doses of individual agents, reducing side effects
Temporal sequencing of different agents could optimize efficacy
Translational Development Pathway:
Development Stage | Key Considerations | Potential Challenges |
---|---|---|
Target Validation | Confirm role of axis in human pathology | Discrepancies between mouse models and human disease |
Lead Identification | Screen for selective modulators | Achieving specificity among related BMP receptors |
Preclinical Testing | Evaluate efficacy in disease models | Context-dependent effects in different tissues |
Biomarker Development | Identify markers of target engagement | Need for accessible biomarkers in relevant tissues |
Clinical Translation | Initial focus on rare diseases with clear genetic basis | Patient stratification for optimal response |
Several methodological innovations could address critical knowledge gaps in BMPR1A biology:
Temporal and Spatial Precision in Signaling Analysis:
Development of optogenetic tools for precise spatiotemporal control of BMPR1A activation
FRET-based biosensors to monitor BMPR1A-mediated signaling in real-time within living cells
These approaches would help resolve conflicting data by providing dynamic information about signaling activation patterns
Comprehensive Interactome Mapping:
Proximity labeling techniques (BioID, APEX) to systematically identify BMPR1A-interacting proteins in different cellular contexts
Comparison of interactomes between normal and disease states to identify pathological alterations
Integration of interactome data with functional genomics to prioritize key interactions
Improved Animal Models:
Development of humanized BMPR1A mouse models that more accurately reflect human receptor variants
Conditional allelic series models enabling expression of varying BMPR1A levels to study dose-dependent effects
Models with fluorescent reporter knock-ins to facilitate cell isolation and lineage tracing
Systems Biology Approaches:
Mathematical modeling of BMPR1A signaling networks to predict system behaviors under various conditions
Integration of multi-omics data to identify emergent properties not evident from single-parameter studies
Network analysis to identify critical nodes and feedback loops regulating BMPR1A signaling
Translational Research Tools:
Development of standardized assays for BMPR1A pathway activation applicable to patient samples
Creation of patient-derived xenograft (PDX) models with defined BMPR1A status
Establishment of biobanks collecting tissues with characterized BMPR1A variants
High-throughput phenotypic screening platforms to identify context-specific modulators of BMPR1A signaling
Advanced Structural Biology:
Single-particle tracking to analyze BMPR1A receptor complex formation and trafficking in living cells
Hydrogen-deuterium exchange mass spectrometry to map conformational changes upon ligand binding
Cryo-electron tomography to visualize BMPR1A signaling complexes in their native cellular environment
These methodological innovations would help resolve contradictory findings, clarify context-dependent effects, and provide a more integrated understanding of BMPR1A biology across different physiological and pathological conditions.
Bone Morphogenetic Protein Receptor-1A (BMPR1A) is a protein encoded by the BMPR1A gene. This receptor is part of the transforming growth factor-beta (TGF-β) superfamily, which plays a crucial role in various cellular processes, including growth, differentiation, and apoptosis . BMPR1A is a type I receptor that binds to bone morphogenetic proteins (BMPs), which are involved in bone and cartilage development .
BMPR1A is a transmembrane serine/threonine kinase receptor. It forms a receptor complex with type II receptors upon ligand binding. This complex then phosphorylates intracellular SMAD proteins, which transduce the signal to the nucleus, leading to the regulation of target gene expression . The receptor is essential for the proper functioning of BMP signaling pathways, which are critical for skeletal development and homeostasis .
BMPR1A can be expressed in various systems, including Chinese Hamster Ovary (CHO) cells. The recombinant human BMPR1A produced in CHO cells is biologically active and is used in various research applications, such as kinase assays . The use of CHO cells for recombinant protein production is advantageous due to their ability to perform post-translational modifications similar to those in human cells .
Mutations in the BMPR1A gene are associated with several disorders, including juvenile polyposis syndrome and hereditary mixed polyposis syndrome . These conditions are characterized by the development of numerous polyps in the gastrointestinal tract, which can increase the risk of colorectal cancer .