BMP-7 Human, His (Bone Morphogenetic Protein-7 with a histidine tag) is a recombinant protein derived from the BMP7 gene, engineered for enhanced purification and stability. It belongs to the transforming growth factor-beta (TGF-β) superfamily and plays critical roles in osteogenesis, inflammation regulation, and organ development . The His-tag (usually six histidine residues) facilitates purification via metal affinity chromatography, making it a valuable tool in research and therapeutic applications .
BMP-7 Human, His is produced in E. coli as a monomeric, non-glycosylated protein. The His tag enables efficient purification using nickel or cobalt affinity columns. Post-purification, the protein retains bioactivity despite the absence of glycosylation, though its stability and receptor binding may differ from glycosylated forms .
BMP-7 Human, His induces osteoblast differentiation via SMAD1/5/8 and MAPK pathways, upregulating osteogenic markers like Runx2 and Osx . It promotes bone fracture repair and inhibits osteoporosis by stimulating mesenchymal stem cell differentiation .
Cardiovascular: Reduces plaque formation and monocyte infiltration in atherosclerosis .
Kidney: Antagonizes epithelial-mesenchymal transition (EMT) and fibrosis by inhibiting TGF-β signaling .
Organ System | Function | Source |
---|---|---|
Bone | Fracture healing, osteoblast differentiation | |
Kidney | Nephron development, fibrosis reversal | |
Pancreas | Induces exocrine-to-endocrine cell conversion |
BMP-7 Human, His is used in osteogenic implants for non-unions and spinal fusions. Clinical trials (e.g., Phase 1 for knee osteoarthritis) show promise in reducing pain and improving joint function, though efficacy varies by dose .
Gastric Cancer: Overexpression correlates with poor prognosis in undifferentiated tumors, serving as a biomarker for metastasis risk .
Pancreatic Cancer: Converts non-endocrine pancreatic tissue to insulin-producing cells, offering potential for diabetes therapy .
BMP-7 Human, His generates Langerhans cells (LCs) from progenitors, enabling studies on dendritic cell function. It induces LCs with Th1-polarizing capacity, contrasting with TGF-β1-generated LCs that favor Th2 responses .
Parameter | BMP-7 Human, His (Intra-Articular) | Placebo |
---|---|---|
Doses Tested | 0.03, 0.1, 0.3, 1.0 mg | Lactose |
Adverse Events | Mild injection site pain (1 mg dose) | Similar |
Efficacy | Trend toward improved pain/function at 0.1–0.3 mg | No effect |
Ectopic Bone | None observed | None |
Feature | BMP-7 Human, His (E. coli) | Native BMP-7 (Human) | BMP-7 Heterodimers |
---|---|---|---|
Structure | Monomer (non-glycosylated) | Homodimer (glycosylated) | Heterodimer (BMP-7/BMP-2/4) |
Bioactivity | Moderate | High | 20× higher than homodimers |
Receptor Binding | Type II (BMPR-II) | Type I/II | Enhanced Smad signaling |
Therapeutic Use | Research, bone implants | Bone repair (OP-1) | Preclinical studies |
BMP-7 Human His-tagged protein is a recombinant form of human Bone Morphogenetic Protein 7 engineered with a histidine tag to facilitate purification and experimental applications. The mature human BMP-7 protein consists of 139 amino acids derived from a larger 431 amino acid pro-protein form . When produced with a His-tag in E. coli expression systems, the protein typically contains 148 amino acids (spanning positions 293-431 of the native sequence) with a molecular weight of approximately 16.5kDa .
The production methodology typically involves:
Cloning the coding sequence for mature BMP-7 into a bacterial expression vector
Adding a 6x histidine tag (His-tag) sequence, usually at the C-terminus
Transforming the construct into E. coli
Inducing protein expression under optimized conditions
Purifying the protein through chelating chromatography that captures the His-tag
This recombinant form is non-glycosylated due to E. coli's inability to perform post-translational glycosylation, which distinguishes it from native BMP-7 and from versions produced in eukaryotic expression systems .
The expression system fundamentally alters the properties of recombinant BMP-7:
Feature | E. coli-Produced BMP-7-His | HEK Cell-Produced BMP-7 |
---|---|---|
Glycosylation | Non-glycosylated | Glycosylated |
Structure | Typically monomeric | Disulfide-linked homodimer |
Molecular Weight | 16.5 kDa | 30-38 kDa range |
Production Yield | Generally higher | Generally lower |
Endotoxin Risk | Higher (requires additional purification) | Lower |
Post-translational Modifications | Limited | Native-like |
The choice between these forms depends on your experimental needs. E. coli-produced His-tagged BMP-7 is advantageous for structural studies, high-yield applications, and experiments where glycosylation is not critical . Conversely, HEK cell-produced BMP-7 more closely resembles the native protein configuration and is preferred for in vivo studies and cellular assays where physiological activity is paramount .
Based on established protocols for recombinant BMP proteins, proper handling of BMP-7 His-tagged protein requires:
Storage at -80°C for long-term stability or -20°C for short-term use
Avoiding repeated freeze-thaw cycles (aliquot upon initial thawing)
Working with the protein in buffers containing stabilizing agents:
Typically phosphate or HEPES-based buffers at pH 7.4
Often containing 0.15M NaCl
May include low concentrations of carrier proteins (0.1-0.5% BSA)
Sometimes with 1-5% glycerol to prevent freeze damage
When conducting experiments, His-tagged BMP-7 is typically used at concentrations between 1-1000 nM depending on the assay type . Surface Plasmon Resonance (SPR) experiments with BMP-7 commonly employ HBS-EP buffer (0.01M HEPES, pH 7.4, 0.15M NaCl, 3mM EDTA, 0.005% surfactant P20) .
BMP-7 plays critical roles in the differentiation of mesenchymal cells into bone and cartilage through several key mechanisms:
Receptor Binding: BMP-7 binds to type I and type II serine/threonine kinase receptors on the cell surface
SMAD Pathway Activation: Upon binding, BMP-7 induces phosphorylation of SMAD1 and SMAD5 transcription factors
Gene Transcription: Phosphorylated SMADs translocate to the nucleus and activate transcription of numerous osteogenic genes
Differentiation Markers: BMP-7 treatment induces the expression of all genetic markers associated with osteoblast differentiation in multiple cell types
Cartilage Maintenance: BMP-7 helps maintain chondrocyte phenotype and counteracts pathological osteoarthritic changes in cartilage
For experimental verification of BMP-7 activity in osteogenesis, researchers commonly assess:
Alkaline phosphatase (ALP) activity
Expression of osteogenic markers including RUNX2, COL1A1, and osteocalcin
Mineral deposition using Alizarin Red staining
SMAD phosphorylation by Western blotting
For chondrogenesis, key assessment parameters include:
SOX9, COL2A1, and NKX3-2 expression (chondrocyte markers)
Suppression of COL10A1, RUNX2, and ALPL (hypertrophic markers)
Recent research has revealed that BMP-7 functions predominantly as a heterodimer with BMP-2 or BMP-4 during mammalian development, which significantly impacts its signaling properties:
Formation of Heterodimers: BMP-7 readily forms heterodimers with BMP-2 and BMP-4 in vivo, and these heterodimers appear to be the predominant active forms
Developmental Importance: Studies using a cleavage mutant mouse (Bmp7 R-GFlag) that prevents proteolytic activation of BMP-7 demonstrated that:
Signaling Potency: BMP heterodimers typically exhibit enhanced signaling potency compared to homodimers, with:
Increased binding affinity to receptors
More efficient induction of downstream SMAD phosphorylation
Different threshold concentrations for biological effects
To experimentally investigate BMP-7 heterodimers, researchers employ:
Co-immunoprecipitation assays to confirm the existence of endogenous BMP heterodimers
Recombinant production of defined heterodimers through co-expression systems
Cleavage-resistant mutant forms to disrupt heterodimer formation in vivo
Several methodological approaches have proven effective for investigating BMP-7 interactions with its receptors:
Surface Plasmon Resonance (SPR):
BMP-7 growth factor (GF) is covalently coupled to sensor chips (~500 resonance units)
Potential binding partners are flowed over in appropriate buffers
For receptor competition experiments, BMP-7 is immobilized and soluble BMPRII extracellular domain is injected at varying concentrations (0-500 nM)
Detection of bound growth factor can be enhanced using monoclonal anti-BMP-7 antibodies
Co-immunoprecipitation:
Cells expressing BMP receptors are treated with His-tagged BMP-7
Complexes are immunoprecipitated using anti-receptor or anti-His antibodies
Western blotting confirms the presence of receptor-ligand complexes
Reporter Gene Assays:
Cells transfected with BMP-responsive elements driving luciferase expression
Dose-dependent activation of reporter gene expression by BMP-7
Competition with soluble receptors or inhibitors quantifies binding affinities
SMAD Phosphorylation Assays:
Western blotting for phosphorylated SMAD1/5/8 following BMP-7 treatment
Time-course and dose-response analyses provide kinetic binding information
SPR experiments with BMP-7 have shown that pH significantly affects binding interactions, with studies conducted at both physiological pH 7.4 and acidic pH 4.5 revealing different binding characteristics .
Recent research has successfully identified bioactive peptides derived from BMP-7 that retain specific biological activities, particularly for applications in osteoarthritis treatment. The methodological approach includes:
Peptide Library Design:
Create overlapping sequential peptides covering the complete mature human BMP-7 sequence
For optimal screening, use 20-mer peptides with 2 amino acid intervals (18 amino acid overlap)
Substitute cysteine residues with serine to avoid uncontrolled oxidation of cysteine groups
This approach yielded 61 individual peptides spanning the complete mature BMP-7 sequence
Screening Methodology:
Validation of Candidate Peptides:
Confirm activity in disease-relevant environments (e.g., OA synovial fluid)
Analyze dose-response relationships
Compare with full-length BMP-7
Using this approach, researchers identified two specific regions within BMP-7 that yield bioactive peptides capable of attenuating the pathological osteoarthritic chondrocyte phenotype: peptides p[63-82] and p[113-132] . These peptides counteracted OA-associated changes in gene expression, including:
Increasing SOX9, COL2A1, and NKX3-2 expression
Decreasing RUNX2, COL10A1, ALPL, MMP13, ADAMTS5, COX-2, and IL6 expression
BMP-7 is recognized as a potent anti-inflammatory growth factor. To experimentally assess its anti-inflammatory properties:
Gene Expression Analysis:
qRT-PCR for inflammatory cytokines (IL-1β, IL-6, TNF-α)
Assessment of anti-inflammatory markers (IL-10, TGF-β)
Analysis of inflammatory enzymes (COX-2, iNOS)
Protein Secretion Measurements:
Signaling Pathway Analysis:
Western blotting for NFκB pathway components
Phosphorylation status of inflammatory signal transducers
Nuclear translocation of transcription factors
Functional Assays:
Migration and invasion assays of inflammatory cells
Inflammatory cell adhesion to endothelial cells
Macrophage polarization (M1/M2 ratio)
In cellular models of osteoarthritis, BMP-7 and its derived peptides have been shown to counteract inflammation by:
Reducing the expression of pro-inflammatory mediators (COX-2, IL-6)
Decreasing PGE2 secretion in chondrocytes exposed to osteoarthritic synovial fluid
Modulating the expression of matrix-degrading enzymes (MMP13, ADAMTS5)
BMP-7 plays critical roles in kidney development and protection from fibrosis, requiring specialized experimental approaches:
Developmental Studies:
Fibrosis Models:
BMP-7 inhibits epithelial-mesenchymal transition (EMT) which can lead to kidney fibrosis
Unilateral ureteral obstruction (UUO) models
Ischemia-reperfusion injury models
Monitoring of fibrotic markers (α-SMA, collagen deposition, fibronectin)
Histological assessment using Masson's Trichrome or Sirius Red staining
Dosing Considerations:
Recombinant BMP-7 is typically administered at 50-300 μg/kg in animal models
For in vitro studies with kidney cell lines (HK-2, MDCK), concentrations of 50-200 ng/ml are commonly used
Treatment duration varies from acute (24-72h) to chronic (1-4 weeks) depending on the model
Delivery Methods:
Systemic administration (intravenous or intraperitoneal injection)
Local administration (subcapsular injection)
Sustained release systems (critical for maintaining therapeutic levels)
BMP-7 expression is known to be attenuated when the kidney is under inflammatory or ischemic stress, which contributes to EMT and fibrosis development . Therefore, experimental timing is crucial when studying BMP-7's protective effects.
CRISPR/Cas9 technology offers powerful approaches for investigating BMP-7 function:
Gene Knockout Strategies:
Complete BMP-7 gene knockout to study null phenotypes
Conditional knockout using Cre-loxP systems for tissue-specific deletion
Targeting of specific domains to create truncated proteins
Knock-in Approaches:
Regulatory Element Modification:
Targeting enhancers or promoters to modulate expression levels
Creating reporter knock-ins to monitor BMP-7 expression patterns
Experimental Design Considerations:
A particularly informative approach used in recent research involved generating knock-in mice carrying a mutation (Bmp7 R-GFlag) that prevents proteolytic activation of the BMP-7 precursor protein, which revealed that BMP-7 functions predominantly as a heterodimer with BMP-2 or BMP-4 during mammalian development .
Rigorous quality control is essential to ensure experimental reproducibility with BMP-7 His-tagged protein:
SDS-PAGE Analysis:
Confirms proper molecular weight (16.5 kDa for E. coli-produced BMP-7-His)
Assess purity (typically >95% for research applications)
Under reducing conditions to evaluate monomer size
Under non-reducing conditions to assess potential dimer formation
Western Blotting:
Anti-BMP-7 antibodies confirm identity
Anti-His antibodies verify tag presence and integrity
Mass Spectrometry:
Precise molecular weight determination
Sequence coverage analysis
Post-translational modification assessment
Endotoxin Testing:
Critical for E. coli-produced proteins
Limulus Amebocyte Lysate (LAL) assay
Acceptable levels typically <1 EU/μg protein
Bioactivity Assays:
SMAD phosphorylation in responsive cell lines
ALP induction in pre-osteoblasts
Reporter gene assays with BMP-responsive elements
Structure Verification:
Circular dichroism to assess secondary structure
Dynamic light scattering for aggregation detection
A common issue with recombinant BMP-7 is the potential presence of misfolded or inactive protein. Functional assays are therefore essential to confirm biological activity before experimental use.
Bone Morphogenetic Protein-7 (BMP-7), also known as osteogenic protein-1, is a member of the Transforming Growth Factor-beta (TGF-β) superfamily. This family of proteins plays a crucial role in the transformation of mesenchymal cells into bone and cartilage . BMP-7 is particularly significant due to its ability to induce ectopic bone formation and promote fracture healing in human patients .
BMP-7 is a secreted ligand that binds to various TGF-β receptors, leading to the recruitment and activation of SMAD family transcription factors, which regulate gene expression . The protein is initially synthesized as a preproprotein and is proteolytically processed to generate each subunit of the disulfide-linked homodimer . This homodimer plays a vital role in bone, kidney, and brown adipose tissue development .
Recombinant BMP-7, particularly the human recombinant form with a His tag, is produced using recombinant DNA technology. This form is engineered to include a histidine tag (His tag) to facilitate purification and detection. The His tag allows for easy purification using nickel-affinity chromatography, which is a common method for isolating recombinant proteins.
Recombinant BMP-7 has several clinical applications. It is used to promote bone growth and healing, particularly in cases of bone fractures and spinal fusions . Additionally, BMP-7 has been studied for its potential in treating chronic kidney disease and other conditions involving tissue regeneration .