The SMPD3 antibody is available in multiple formats, including polyclonal and monoclonal variants, tailored for specific applications such as Western blot (WB), ELISA, and immunohistochemistry (IHC). Key characteristics include:
Polyclonal Antibodies (e.g., CAB10197, AF7184) offer broader epitope recognition, making them suitable for WB and IHC .
Monoclonal Antibodies (e.g., MAB7184, 68801-2-PBS) provide high specificity for targeted detection in ELISA and multiplex assays .
Cross-reactivity: Most antibodies target conserved regions across species, enabling studies in human, mouse, and rat models .
SMPD3 antibodies are pivotal in studying the enzyme’s role in cellular processes and diseases:
SMPD3 is predominantly localized to the Golgi complex, particularly in detergent-insoluble membrane domains (DIMs) enriched in sphingomyelin (SM) and cholesterol . Confocal microscopy and immunohistochemistry using SMPD3 antibodies confirm its colocalization with Golgi markers (e.g., K58, SMS1) in neurons, macrophages, and HEK293 cells .
Sphingolipid Metabolism: SMPD3 hydrolyzes SM into ceramide, a bioactive lipid that regulates apoptosis, inflammation, and cell growth . Antibodies enable tracking of ceramide production in response to SMPD3 activation or inhibition.
Neurodegenerative Diseases: SMPD3 deficiency in mice causes dwarfism and pituitary hormone deficiencies, linked to disrupted growth hormone-releasing hormone (GHRH) signaling. SMPD3 antibodies reveal its critical role in hypothalamic-pituitary axis regulation .
Cancer Research: In gliomas, SMPD3 knockdown via shRNA increases tumor growth, while its expression correlates with longer survival in IDH-mutant gliomas. Antibodies facilitate studies on its dual role in cell-autonomous and paracrine signaling .
Periodontal Disease: SMPD3 enhances cytodifferentiation in periodontal ligament (PDL) cells, promoting mineralization-related gene expression (e.g., ALPase, Runx2). A functional SNP (rs145616324) in SMPD3 is associated with aggressive periodontitis .
Biomarker Potential: SMPD3 is implicated in pulmonary emphysema and IDH-mutant gliomas, making it a candidate biomarker for targeted therapies .
SMPD3’s role in Golgi secretory pathways is well-documented:
Vesicle Trafficking: SMPD3 regulates SM/cholesterol dynamics in Golgi DIMs, facilitating COPI vesicle formation and secretion .
Ceramide Signaling: Ceramide generated by SMPD3 modulates apoptosis, inflammation, and proliferation, influencing tumor microenvironments .
Therapeutic Targeting: Inhibiting SMPD3 may suppress ceramide-driven apoptosis in neurodegenerative diseases, while enhancing its activity could limit tumor growth in gliomas .
Diagnostic Potential: SMPD3 expression levels correlate with survival in IDH-mutant gliomas, suggesting its utility as a prognostic marker .
Future Research:
Applications : Fluorescent Western Blotting
Sample type: cells
Review: SMPD3 expression did not show any tissue-specific features in either of the study groups.
SMPD3 (Sphingomyelin Phosphodiesterase 3) is an enzyme that catalyzes the hydrolysis of sphingomyelin to generate ceramide and phosphocholine. It plays crucial roles in bone and cartilage development, as evidenced by studies using fro/fro mice with defective SMPD3 expression . The importance of SMPD3 extends to both chondrocytes and osteoblasts, with distinct functions in each cell type. Research has shown that SMPD3 restoration in osteoblasts can correct bone mineralization defects, while its expression in chondrocytes is necessary for proper cartilage development . The enzyme is regulated by key transcription factors like SOX9 in chondrogenic cells, highlighting its integration in developmental pathways .
Multiple types of SMPD3 antibodies are available for research, varying in host species, clonality, and target regions:
Host species: Primarily rabbit and sheep polyclonal antibodies, with some mouse polyclonal options available
Clonality: Most commercially available antibodies are polyclonal, though specific applications may benefit from monoclonal antibodies for increased specificity
Target regions: Antibodies targeting different regions of SMPD3 are available, including:
These varieties allow researchers to select antibodies appropriate for their specific experimental needs, target availability in samples, and detection requirements.
When detecting SMPD3 via Western blotting, researchers should anticipate bands at specific molecular weights that may vary slightly depending on post-translational modifications and experimental conditions:
In specific experimental contexts, Western blots probed with anti-SMPD3 antibodies have detected bands at approximately 70-75 kDa in human cell lines including RPMI 8226 (multiple myeloma) and CEM (T-lymphoblastoid) . This slight discrepancy between calculated and observed molecular weights may be attributed to post-translational modifications such as glycosylation, which can increase the apparent molecular weight on SDS-PAGE gels. Researchers should validate the specific band pattern in their experimental system, as expression levels and post-translational modifications may vary between tissue types and cellular conditions.
Commercially available SMPD3 antibodies demonstrate varied cross-reactivity profiles, offering flexibility for researchers working with different model organisms:
When selecting an antibody for cross-species applications, researchers should consider sequence homology in the target epitope region. The high predicted reactivity percentages for the ABIN2783713 antibody suggest strong conservation of the middle region epitope (sequence: RPPEADDPVP GGQARNGAGG GPRGQTPNHN QQDGDSGSLG SPSASRESLV) across mammalian species . For critical cross-species applications, validation experiments should confirm reactivity in the specific species of interest.
Successful Western blot detection of SMPD3 requires careful optimization of several parameters:
Reducing conditions: SMPD3 detection is typically performed under reducing conditions
Buffer systems: Immunoblot Buffer Group 1 has been successfully used with SMPD3 antibodies
Secondary antibody selection: Match to the host species of the primary antibody (e.g., HRP-conjugated Anti-Sheep IgG for sheep primary antibodies)
Use appropriate positive controls such as cell lysates with known SMPD3 expression
RPMI 8226 (human multiple myeloma) and CEM (human T-lymphoblastoid) cell lines have been validated as positive controls
For novel sample types, optimize dilutions within the recommended range as suggested by manufacturers
If non-specific bands appear, increase antibody dilution or implement additional blocking steps
If signal is weak, consider longer exposure times or signal enhancement systems compatible with the detection method
Proper storage and handling of SMPD3 antibodies is critical for maintaining their specificity and sensitivity over time:
Aliquot antibodies upon receipt to avoid repeated freeze-thaw cycles
For working solutions, store according to manufacturer's recommendations (typically 4°C for short-term use)
Many SMPD3 antibodies are supplied in buffers containing:
Avoid repeated freeze-thaw cycles as they can lead to protein denaturation and reduced antibody performance
Monitor solution clarity; cloudiness may indicate protein aggregation
Follow manufacturer's expiration guidelines, typically 12-24 months when properly stored
Allow antibodies to equilibrate to room temperature before opening to prevent condensation
Use sterile technique when handling antibody solutions
Return to appropriate storage conditions promptly after use
Rigorous validation of SMPD3 antibody specificity is essential for generating reliable research data:
Use samples from SMPD3 knockout models (e.g., fro/fro mice) as negative controls
Compare conditional knockout models (e.g., Smpd3; flox/flox Osx-Cre or Smpd3; flox/flox Col2a1-Cre) with wild-type controls
Employ siRNA knockdown in cell culture systems to confirm specificity
Peptide competition assays using the immunizing peptide (e.g., synthetic peptide from the middle region of human SMPD3)
Multiple antibody approach - confirm findings using antibodies targeting different epitopes of SMPD3
Immunoprecipitation followed by mass spectrometry to confirm target identity
Use recombinant SMPD3-expressing systems as positive controls
Compare with endogenous expression in known SMPD3-expressing tissues/cells
Correlation of protein detection with mRNA expression levels across tissues
Include isotype controls matching the primary antibody host species and isotype
Evaluate secondary antibody non-specific binding by omitting primary antibody
Test reactivity across tissue panels to confirm expected expression patterns
While Western blotting is commonly used with SMPD3 antibodies, immunocytochemistry (ICC) provides valuable information about subcellular localization:
Fixation method affects epitope accessibility: 4% paraformaldehyde is commonly used for SMPD3 detection
Permeabilization is critical for accessing intracellular SMPD3, with 0.1-0.5% Triton X-100 typically used
Antigen retrieval methods may be necessary for formalin-fixed samples
Blocking with 5-10% normal serum corresponding to secondary antibody host
Primary antibody incubation typically overnight at 4°C for optimal signal-to-noise ratio
Longer washing steps help reduce background when using polyclonal antibodies
SMPD3 expression may be nearly undetectable in certain conditions, as seen in differentiated ATDC5 cells
Expression patterns vary by cell type; chondrocytes and osteoblasts show distinct SMPD3 localization patterns
Include positive controls with known SMPD3 expression
Compare patterns with published localization data
Verify specificity with peptide competition or genetic knockdown approaches
Research into SMPD3 regulation requires careful experimental design considering multiple factors:
SOX9 has been identified as a regulator of SMPD3 expression in chondrogenic cells
Promoter analysis and chromatin immunoprecipitation can identify transcription factor binding sites
Reporter assays using SMPD3 promoter constructs can validate regulatory elements
SMPD3 exhibits differential expression and function between chondrocytes and osteoblasts
Cell type-specific regulatory mechanisms should be considered when designing experiments
Comparison across multiple cell types can identify tissue-specific regulatory mechanisms
SMPD3 expression varies during development, particularly in the growth plate
Time-course experiments during differentiation can capture dynamic expression changes
Correlation with stage-specific markers helps contextualize expression patterns
qPCR for mRNA expression analysis
Western blotting for protein-level changes
Immunohistochemistry for spatial expression patterns in tissues
Promoter-reporter constructs for transcriptional regulation studies
SMPD3 plays distinct but critical roles in both bone and cartilage development, as revealed through genetic models:
SMPD3 deficiency in fro/fro mice leads to significant bone mineralization defects
Restoration of SMPD3 expression specifically in osteoblasts (fro/fro; Col1a1-Smpd3 mice) corrects bone mineralization defects, demonstrating its cell-autonomous function in osteoblasts
SMPD3 likely regulates matrix vesicle formation and function in mineralizing osteoblasts
SMPD3 deficiency results in an expanded zone of hypertrophic chondrocyte-like cells and poor mineralization of cartilage matrix
Cartilage abnormalities persist in fro/fro; Col1a1-Smpd3 mice despite correction of bone defects, indicating separate regulatory mechanisms
Transgenic restoration of SMPD3 in chondrocytes (fro/fro; Acan-Smpd3 mice) corrects cartilage abnormalities but not bone mineralization defects
SMPD3 is required for proper organization and mineralization of the growth plate
Its absence affects the hypertrophic chondrocyte zone while sparing chondrocyte proliferation
Expression of major chondrocyte differentiation markers remains largely intact in SMPD3-deficient growth plates
This dual role highlights the importance of studying SMPD3 function in a tissue-specific context and suggests that therapeutic approaches targeting SMPD3 may need to consider its distinct functions in different skeletal tissues.
The relationship between SMPD3 expression and chondrocyte differentiation reveals important insights into cartilage development:
SOX9, a master regulator of chondrogenesis, controls SMPD3 expression in chondrogenic cells
SMPD3 expression changes during chondrocyte differentiation stages
In some experimental systems, SMPD3 protein becomes nearly undetectable in fully differentiated ATDC5 chondrogenic cells
SMPD3 deficiency results in an expanded hypertrophic chondrocyte zone in growth plates
Despite altered morphology, proliferation of growth plate chondrocytes remains normal in fro/fro mice
The expression of major chondrocyte differentiation markers is not significantly affected by SMPD3 deficiency
Chondrocyte-specific expression of SMPD3 in fro/fro; Acan-Smpd3 mice corrects cartilage abnormalities but not bone defects
This demonstrates the cell-autonomous function of SMPD3 in chondrocytes
As a sphingomyelinase, SMPD3 influences local ceramide levels in the cartilage microenvironment
Ceramide and its metabolites likely influence chondrocyte maturation and function, particularly during the mineralization process
Recent research has begun to elucidate SMPD3's role in muscle tissue and its potential implications for metabolic health:
SMPD3 is expressed in human skeletal muscle, where it contributes to sphingolipid metabolism
Western blot analysis confirms SMPD3 protein presence in muscle tissue homogenates
Diacylglycerols and sphingolipids influence insulin sensitivity and mitochondrial function in skeletal muscle
SMPD3, through its enzymatic activity generating ceramide, may impact intracellular signaling pathways related to insulin response
Tissue homogenates from human skeletal muscle biopsies can be analyzed for SMPD3 expression
Western blotting with anti-SMPD3 antibodies provides information about expression levels and potential post-translational modifications
Alterations in sphingolipid metabolism, potentially involving SMPD3 activity, may contribute to insulin resistance mechanisms
This connection makes SMPD3 a potential target of interest in metabolic research
Research in this area is still developing, and further investigation will likely reveal more detailed mechanisms connecting SMPD3 function to muscle metabolism and systemic metabolic health.
Distinguishing between sphingomyelinase family members requires careful experimental design and specific methodological approaches:
Choose antibodies validated for specificity against individual SMPD family members
Confirm lack of cross-reactivity with other family members through western blot analysis of recombinant proteins
Consider using epitopes from regions with minimal sequence homology between family members
SMPD family members show distinct tissue and subcellular distribution patterns:
SMPD1 (acid sphingomyelinase): primarily lysosomal
SMPD2: primarily membrane-associated
SMPD3 (neutral sphingomyelinase 2): Golgi apparatus and plasma membrane
SMPD4: primarily endoplasmic reticulum
pH optima differ: SMPD1 (acidic pH), SMPD2/3/4 (neutral pH)
Selective inhibitors can distinguish between family members
Cell compartment-specific assays can help identify which enzyme is active in particular locations
Selective knockdown/knockout of individual family members:
Compensatory changes in other family members should be monitored in knockout models
SMPD3 deficiency causes specific skeletal abnormalities not seen with other family members
SMPD1 deficiency causes Niemann-Pick disease with distinct manifestations
These phenotypic differences help distinguish the non-redundant functions of each family member
Researchers frequently encounter specific challenges when working with SMPD3 antibodies that require systematic troubleshooting:
Possible causes: Insufficient protein expression, antibody degradation, suboptimal blocking
Solutions:
Possible causes: Cross-reactivity, degradation products, non-specific binding
Solutions:
Possible causes: Antibody batch variation, sample preparation differences
Solutions:
Possible causes: Variable expression levels, tissue-specific modifications
Solutions:
Enrich samples through immunoprecipitation
Use tissue-specific positive controls
Consider sample preparation methods optimized for specific tissues
Adapting SMPD3 research across different model systems requires specific methodological adjustments:
ATDC5 cells represent a useful model for studying SMPD3 in chondrogenic differentiation
RPMI 8226 and CEM cell lines serve as positive controls for human SMPD3 expression
Consider cell type-specific expression levels when planning experiments
fro/fro mice with SMPD3 deficiency serve as valuable negative controls
Transgenic models with tissue-specific expression/deletion provide mechanistic insights:
Consider tissue source variability and preservation methods
Account for potential genetic variation affecting antibody binding
Include appropriate controls for each tissue type
Antibody selection should account for species reactivity profiles
The ABIN2783713 antibody shows high predicted reactivity across multiple mammalian species
Sequence conservation in target epitopes influences cross-reactivity
Efficient SMPD3 protein isolation is critical for successful antibody-based detection:
Use buffers containing adequate detergents to solubilize membrane-associated SMPD3
RIPA buffer supplemented with protease inhibitors is commonly effective
For challenging samples, consider specialized sphingolipid-enriched membrane extraction protocols
SMPD3 localizes to specific cellular compartments including the Golgi apparatus
Enrichment through organelle isolation can improve detection sensitivity
Differential centrifugation protocols can separate membrane-associated and soluble fractions
If functional studies are planned, gentler extraction conditions may be necessary
Addition of phosphatase inhibitors preserves the phosphorylation state
Temperature control during extraction prevents degradation
Process tissues/cells rapidly to minimize protein degradation
Standardize protein quantification methods for consistent loading
Consider adding reducing agents to buffers to maintain protein stability
Aliquot samples to avoid repeated freeze-thaw cycles
Immunoprecipitation using validated SMPD3 antibodies can concentrate the target
For low abundance samples, consider concentration methods prior to analysis
Affinity purification using sphingomyelin-based substrates can enrich active enzyme
Multiplexed detection provides valuable contextual information about SMPD3 function and localization:
Select antibodies raised in different host species to avoid cross-reactivity
For same-species antibodies, consider directly conjugated primary antibodies
Sequential staining protocols can overcome antibody incompatibilities
Chondrocyte studies: Pair with SOX9 (regulator of SMPD3) and collagen type II
Osteoblast studies: Co-stain with osteocalcin or RUNX2
Subcellular localization: Combine with organelle markers (Golgi, plasma membrane)
Optimize antigen retrieval conditions compatible with all target epitopes
Test antibodies individually before combining to establish optimal conditions
Include appropriate controls for each antibody in the multiplex panel
Sequential chromogenic immunohistochemistry for challenging combinations
Tyramide signal amplification for detecting low-abundance targets
Proximity ligation assays for detecting protein-protein interactions involving SMPD3
Use sequential scanning for confocal microscopy to prevent channel bleed-through
Apply appropriate controls for autofluorescence, especially in bone/cartilage tissue
Quantitative co-localization analysis should include statistical validation
Correlating SMPD3 protein detection with enzymatic activity provides comprehensive functional insights:
Split samples for simultaneous protein detection and activity assays
Standardize extraction conditions to preserve both protein integrity and enzymatic activity
Compare protein levels via Western blotting with activity measurements
Fluorescent substrate assays: Using sphingomyelin analogs with fluorescent reporters
Radiometric assays: Using radiolabeled sphingomyelin substrates
Mass spectrometry: Direct measurement of ceramide production
SMPD3 shows optimal activity at neutral pH, distinguishing it from acid sphingomyelinase (SMPD1)
pH titration experiments can help attribute activity to specific family members
Selective inhibition of different sphingomyelinase family members
Correlation of activity inhibition with changes in cellular phenotypes
Comparison with genetic knockdown/knockout models