DC-STAMP is a seven-transmembrane protein essential for osteoclast (OC) precursor fusion and multinucleated osteoclast formation . It contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) that regulates signaling pathways during osteoclastogenesis . DC-STAMP interacts with SHP-1 phosphatase and CD16 (an ITAM-associated protein), balancing activation and inhibitory signals in myeloid cells .
Epitope: 447EVHLKLHGEKQGTQ460 (fourth extracellular domain, conserved in humans and mice) .
Applications:
Osteoclast Regulation:
Inflammatory Arthritis:
Cancer: High DC-STAMP expression in acute myeloid leukemia (AML) predicts poor survival and correlates with immune checkpoint genes (PDCD1, CTLA-4) .
Autoimmunity: Aged DC-STAMP<sup>-/-</sup> mice develop systemic autoimmunity, linking DC-STAMP to phagocytosis and self-tolerance .
1A2 Specificity: Detects DC-STAMP in RAW264.7 lysates transfected with PTHR-DC-STAMP fusion constructs .
Dimerization: Identifies ~106 kDa dimers under non-denaturing conditions .
Human Tissues: DC-STAMP is expressed in liver hepatocellular carcinoma cells (HepG2) and inflamed synovium .
Storage: Stable at -70°C for 12 months; reconstituted antibodies retain activity for 6 months .
Cross-Reactivity: Clone 1A2 recognizes both human and murine DC-STAMP , while R&D Systems’ MAB7824 is human-specific .
This rabbit polyclonal antibody targeting DCSTAMP is designed for use in ELISA, IHC, and IF research applications. Developed against a recombinant protein corresponding to amino acids 119-209 of human DCSTAMP, this antibody undergoes protein G purification, achieving a purity exceeding 95%. It exhibits reactivity with the human species.
The DCSTAMP protein plays a critical role in osteoclast differentiation and bone resorption. It has also been implicated in the regulation of immune responses, as well as cancer cell migration and invasion. Mutations in the DCSTAMP gene can lead to various pathological conditions, including osteoporosis, rheumatoid arthritis, and periodontitis.
DC-STAMP is a seven-pass-transmembrane receptor-like protein that contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain. It plays an essential role in:
Cell-to-cell fusion during osteoclastogenesis (bone resorption processes)
Regulating immune cell interactions and inflammatory responses
Modulating cytokine production in dendritic cells and macrophages
Research significance: DC-STAMP knockout mice exhibit mild osteopetrosis and impaired osteoclast fusion, indicating its importance in bone homeostasis . DC-STAMP also regulates inflammatory cytokine production, making it relevant to both bone and immunological research .
Validation should include multiple complementary approaches:
Genetic validation: Compare antibody binding between wild-type and DC-STAMP knockout samples. For example, Western blotting analysis with anti-DC-STAMP antibody 1A2 detected DC-STAMP in thymus and spleen lysates from wild-type mice but showed greatly reduced signals in Dcstamp-knockout mice .
Recombinant protein validation: Test antibody recognition of recombinant DC-STAMP. The 1A2 antibody showed specific binding to DC-STAMP in PTHR-DC-STAMP fusion construct-transfected RAW cells .
Immunoprecipitation-Western blot: Confirm antibody specificity by immunoprecipitating DC-STAMP followed by detection via Western blot. Monomeric (~53 kDa) and dimeric (~106 kDa) forms can be detected under denaturing and non-denaturing conditions respectively .
Cell type comparison: Compare staining patterns across different DC-STAMP-expressing cell populations (e.g., CD14+CD16- vs. CD14+CD16+ monocytes) .
Sample preparation: For surface staining, block Fc receptors with anti-CD16/CD32 for 15 minutes on ice before adding fluorophore-conjugated anti-DC-STAMP antibody (30 minutes, 4% FCS/PBS) .
Controls: Include 7-AAD for viability gating. For proper gating, use unstained, isotype control, and single-stained controls .
Antibody recommendations: 1A2 clone detects DC-STAMP with 100-fold higher sensitivity compared to KR104 in flow cytometry applications .
Intracellular staining: Use Cytofix/Cytoperm kit for permeabilization when assessing total (not just surface) DC-STAMP .
Membrane extraction: Use native membrane protein extraction methods for optimal results, especially when studying transmembrane proteins like DC-STAMP .
Non-denaturing vs. denaturing conditions: Under non-denaturing conditions, dimeric DC-STAMP (~106 kDa) is detected; under denaturing conditions, monomeric form (~53 kDa) is detected .
Antibody dilution: Start with 1:1000 dilution in 5% BSA/TBST for overnight incubation at 4°C .
Protocol: Use EZView Red Protein A Affinity Gel beads after incubating membrane proteins with anti-DC-STAMP antibody for 1 hour at 4°C .
Detection: Follow with SDS-PAGE and immunoblotting using the same or different anti-DC-STAMP antibody .
For accurate quantification:
Baseline comparisons: Compare mean fluorescence intensity (MFI) across experimental conditions. Note that CD14+CD16+ monocytes show higher DC-STAMP expression (MFI: 2748) compared to CD14+CD16- monocytes (MFI: 1747) .
Controls for accurate measurement:
Analysis considerations:
Gate on viable cells (using 7-AAD exclusion)
Consider cell size/complexity effects on fluorescence intensity
Report relative MFI compared to control populations
DC-STAMP antibodies provide valuable tools for investigating osteoclast development:
Functional blocking: The 1A2 antibody inhibits osteoclast precursor (OCP) fusion in vitro, making it useful for studying the mechanism of multinucleation during osteoclastogenesis .
Expression kinetics: DC-STAMP surface expression shows reciprocal regulation with CD16 during osteoclastogenesis. While CD16 expression increases over time, DC-STAMP levels decline .
Cellular localization: Use fluorescence microscopy with rhodamine phalloidin (for actin), FITC-conjugated anti-DC-STAMP 1A2, and DAPI (for nuclei) to visualize DC-STAMP distribution during cell fusion:
Expression analysis: Track DC-STAMP mRNA expression during osteoclast differentiation using qPCR with the following primer sets:
DC-STAMP bridges bone resorption and inflammation through several mechanisms:
Cytokine regulation: DC-STAMP knockout significantly alters cytokine production profiles. DC-STAMP knockdown mBMDCs secrete less IL-6, IL-12, TNF-α, and IL-10 while producing more IL-1 .
Transcriptional effects: Research shows DC-STAMP affects cytokine gene transcription:
Inflammatory model studies: In the Tg(hTNF) arthritis mouse model, DC-STAMP knockout shows:
Macrophage phenotype: DC-STAMP regulates macrophage polarization. DC-STAMP-deficient macrophages show impaired Tnf, Il1β, and Inos expression and reduced production of IL1β, TNF, IL6, and CCL2 .
The dimeric structure of DC-STAMP presents unique experimental challenges:
Sample preparation:
Antibody selection:
Detection strategy:
Controls and validation:
The immunoreceptor tyrosine-based inhibitory motif (ITIM) in DC-STAMP's cytoplasmic domain (S407FYPSV412) provides important signaling capabilities:
Signaling mechanism: The ITIM motif likely counteracts immunoreceptor tyrosine-based activation motif (ITAM) signaling from molecules like CD16. This suggests DC-STAMP may provide inhibitory counterbalance to activation signals .
Experimental approaches:
Phosphorylation studies: Use anti-phosphotyrosine mAb 4G10 to detect phosphorylated ITIM following immunoprecipitation with anti-DC-STAMP 1A2
Co-immunoprecipitation: Identify binding partners using anti-DC-STAMP 1A2 pulldown followed by mass spectrometry or Western blotting
Mutational analysis: Create ITIM-mutant DC-STAMP constructs to assess functional consequences
Technical considerations:
Several factors may contribute to contradictory findings:
Age-dependent effects: DC-STAMP knockout mice develop different phenotypes with age. Young mice show mild osteopetrosis, while aged mice develop systemic autoimmunity with lymphoproliferation, splenomegaly, and autoantibodies .
Model-specific differences: The suppression of inflammation in Dcstamp-/-;Tg(hTNF) model contrasts with autoimmunity in aged Dcstamp-/- mice, suggesting context-dependent functions .
Cell type variations: DC-STAMP knockdown in bone marrow dendritic cells impairs IL-6, IL-12, and TNF release while increasing IL-1β, promoting Th2 differentiation .
Technical variations: Different antibodies (1A2 vs. KR104) target distinct epitopes and show varying efficacy in different applications. For example, KR104 targets the 1st extracellular domain and fails to recognize PTHR-DC-STAMP fusion protein, while 1A2 targets the 4th extracellular domain .
Experimental interpretation: When findings conflict, consider:
Comparing antibody specificity and epitope locations
Validating with multiple detection methods
Confirming knockout/knockdown efficiency
Considering developmental stage and cell type
DC-STAMP stands at the intersection of bone homeostasis and inflammation, offering promising research avenues:
Biomarker development: DC-STAMP may serve as an activity biomarker in inflammatory arthritis, as knockout mice show decreased local and systemic inflammation in arthritis models .
Therapeutic targeting: Anti-DC-STAMP antibodies like 1A2 that block osteoclast formation could potentially be developed into therapeutic agents for conditions with excessive bone resorption .
Mechanistic studies: DC-STAMP antibodies can help elucidate:
Clinical correlations: Measuring DC-STAMP expression on peripheral blood monocytes using flow cytometry could potentially correlate with disease activity in conditions like rheumatoid arthritis or psoriatic arthritis .
Future methodological improvements could include:
Single-cell analysis: Combining DC-STAMP antibody labeling with single-cell RNA sequencing to correlate DC-STAMP expression with transcriptional profiles of osteoclast precursors or inflammatory cells.
Advanced imaging: Super-resolution microscopy with fluorescently-labeled DC-STAMP antibodies to visualize molecular distribution during cell fusion events.
Proximity labeling: Using DC-STAMP antibodies conjugated to proximity labeling enzymes (BioID, APEX2) to identify proximal interacting partners in living cells.
CRISPR screening: Combining DC-STAMP antibody-based phenotyping with CRISPR screens to identify genes that modulate DC-STAMP expression, dimerization, or signaling.
Structural studies: Using antibody fragments to facilitate crystallization and structural determination of DC-STAMP, particularly given the technical challenges in membrane protein structural studies highlighted in search result .