HMGCL Antibody, FITC conjugated refers to a fluorescently labeled antibody targeting the HMGCL protein (UniProt ID: P35914). FITC conjugation enables visualization through fluorescence-based techniques, making it valuable for cellular localization studies and quantitative assays. HMGCL catalyzes the cleavage of HMG-CoA into acetoacetate and acetyl-CoA, a rate-limiting step in ketone body synthesis .
Protocol: Fixed and permeabilized HeLa cells incubated with 1 µg/10^6 cells of FITC-conjugated HMGCL antibody for 45 minutes at 4°C. Signal specificity confirmed using rabbit IgG isotype controls .
Result: Distinct fluorescence peaks (FITC channel) observed in HMGCL-expressing cells, with >10,000 events analyzed .
Band Detection: A single band at ~35 kDa in human cell lysates (HeLa, HEK293, HepG2), consistent with HMGCL’s predicted molecular weight .
Secondary Antibody: FITC-conjugated goat anti-rabbit IgG (1:50,000 dilution) used for chemiluminescent detection .
Staining Protocol: Paraffin-embedded human prostate tissue sections treated with 1:100 diluted antibody. FITC signal amplified using HRP-polymer systems and DAB visualization .
The table below contrasts HMGCL antibodies from leading suppliers:
*FITC conjugation achieved via secondary antibodies (e.g., goat anti-rabbit IgG-FITC) .
Specificity Controls: Non-specific binding risks necessitate isotype controls and titration in preliminary experiments .
Batch Consistency: Recombinant monoclonal antibodies (e.g., Proteintech’s 84769 series) offer superior reproducibility compared to polyclonal variants .
Safety: Sodium azide preservative in some formulations requires careful handling .
Statins and Autoimmunity: Anti-HMGCR autoantibodies (distinct from HMGCL) are linked to statin-associated necrotizing myopathy, underscoring the need for precise epitope targeting .
Subcellular Localization: FITC-based imaging confirms HMGCL’s mitochondrial localization, while its isoform HMGCLL1 localizes to extramitochondrial compartments .
HMGCL (3-hydroxymethyl-3-methylglutaryl-Coenzyme A lyase) is a mitochondrial enzyme that catalyzes the final step of leucine degradation and plays a key role in ketogenesis. It performs a cation-dependent cleavage of (S)-3-hydroxy-3-methylglutaryl-CoA into acetyl-CoA and acetoacetate . This enzymatic activity is particularly important in understanding ketone body metabolism, which provides alternative energy sources during fasting or metabolic stress conditions. Ketone bodies (beta-hydroxybutyrate, acetoacetate, and acetone) serve as essential alternative energy sources to glucose, function as lipid precursors, and act as metabolic regulators . Research into HMGCL has significant implications for studying inborn errors of metabolism, particularly hydroxymethylglutaricaciduria, an autosomal recessive disorder that can lead to hypoglycemia and coma .
Human HMGCL is a 325 amino acid protein with a calculated molecular weight of approximately 34 kDa, which corresponds to its observed molecular weight in experimental settings . The protein is primarily localized to mitochondria, though recent research has identified an extramitochondrial homolog called HMGCLL1 . HMGCL contains specific domains required for its lyase activity and has been characterized through various structural and biochemical approaches. The protein's function is conserved across species, with high sequence homology observed between human, mouse, and rat HMGCL proteins, making it amenable to comparative studies across model organisms .
FITC-conjugated HMGCL antibodies offer direct fluorescent detection capability without requiring secondary antibodies, which provides several methodological advantages:
Reduced experimental steps and time in immunofluorescence and flow cytometry applications
Elimination of potential cross-reactivity issues associated with secondary antibodies
Direct visualization in microscopy applications with excitation/emission profiles of FITC (excitation ~495 nm, emission ~519 nm)
HMGCL antibodies have been validated for multiple applications with varying recommended dilutions:
Application | Unconjugated Dilution Range | FITC-Conjugated Dilution Range |
---|---|---|
Western Blot (WB) | 1:500-1:6000 | 1:300-5000 |
Immunohistochemistry (IHC) | 1:20-1:200 | 1:50-200 |
Immunoprecipitation (IP) | 0.5-4.0 μg for 1-3 mg lysate | Not typically used |
Immunofluorescence (IF) | 1:50-1:200 | 1:50-200 |
Flow Cytometry (FC) | 1:50-1:200 | 1:50-200 |
ELISA | Application-specific | Application-specific |
FITC-conjugated antibodies are particularly advantageous for IF and FC applications, where direct fluorescent detection is beneficial . While unconjugated antibodies can be used across all applications, FITC-conjugated versions are optimized for fluorescence-based techniques. For applications like Western blotting, unconjugated antibodies followed by enzyme-conjugated secondary antibodies (HRP or AP) are typically preferred over FITC-conjugated versions due to detection sensitivity considerations .
Based on validated flow cytometry protocols, the following methodology is recommended:
Cell Preparation:
Fix cells in 4% formaldehyde for 10-15 minutes at room temperature
Permeabilize with 0.2% Triton X-100 for 10 minutes
Block with 10% normal goat serum to reduce non-specific binding
Antibody Staining:
Apply FITC-conjugated HMGCL antibody at 1:100 dilution (1 μg per 10^6 cells)
Incubate for 45 minutes at 4°C in the dark to prevent photobleaching
Wash cells 3 times with PBS containing 1% BSA
Controls and Analysis:
Include an isotype control (e.g., FITC-conjugated rabbit IgG) at equivalent concentration
Acquire at least 10,000 events per sample for statistical robustness
Analyze using appropriate gating strategies based on negative controls
This protocol has been validated for detection of HMGCL in HeLa cells, with clear differentiation between positive staining and control samples . For optimal results, titration of antibody concentration may be necessary depending on cell type and expression level.
A comprehensive validation approach for HMGCL antibodies should include:
Western Blot Validation:
Verification of a single band at the expected molecular weight (~34 kDa)
Comparison between HMGCL-expressing and non-expressing cell lines
For definitive validation, comparison with HMGCL knockout/knockdown samples
Immunoreactivity Testing:
Examine cross-reactivity with recombinant HMGCL protein
Test reactivity against related proteins, particularly HMGCLL1
Evaluate species cross-reactivity if working with non-human samples
Application-Specific Controls:
For IHC/IF: Include peptide competition assays and isotype controls
For FC: Use isotype controls and known positive/negative cell populations
For IP: Perform reverse IP and mass spectrometry confirmation
Published validation data shows that high-quality HMGCL antibodies detect a specific 34 kDa band in Western blotting and show distinct mitochondrial localization patterns in immunofluorescence analyses . When using antibodies across species, verify reactivity, as some antibodies have been specifically tested with human, mouse, and rat samples .
Distinguishing between HMGCL and its homolog HMGCLL1 requires careful antibody selection and experimental design:
Antibody Selection:
Subcellular Fractionation:
Employ rigorous mitochondrial isolation protocols to separate mitochondrial and extramitochondrial fractions
Verify fraction purity using established markers (e.g., VDAC for mitochondria, GAPDH for cytosol)
Immunofluorescence Co-localization:
Perform dual staining with HMGCL antibody and mitochondrial markers
HMGCLL1 has been shown to associate with vesicular structures rather than mitochondria
Use confocal microscopy to determine precise subcellular localization patterns
Myristoylation Detection:
This differentiation is crucial as HMGCLL1 may have distinct physiological roles in extramitochondrial lipid metabolism or energy production in specific tissues and cancer cells .
For optimal results in immunofluorescence applications with FITC-conjugated HMGCL antibodies:
Fixation and Permeabilization:
For paraformaldehyde fixation (recommended): 4% PFA for 10-15 minutes at room temperature
For methanol fixation (alternative): 100% methanol at -20°C for 10 minutes
Permeabilization with 0.2-0.5% Triton X-100 for optimal mitochondrial access
Blocking Conditions:
Use 5-10% normal serum (from the species of secondary antibody if dual labeling)
Include 1% BSA to reduce non-specific binding
Consider adding 0.1% Tween-20 to blocking buffer
Antibody Concentration and Incubation:
Microscopy Settings:
Use appropriate filter sets for FITC (excitation ~495 nm, emission ~519 nm)
Minimize exposure time to prevent photobleaching
Consider using anti-fade mounting media containing DAPI for nuclear counterstaining
Controls:
Include isotype control antibodies conjugated to FITC
Consider a peptide competition assay to verify specificity
Use known positive and negative tissues/cells as biological controls
Successful immunofluorescence staining reveals a characteristic mitochondrial distribution pattern for HMGCL, as demonstrated in validated studies with HeLa cells .
Several robust quantitative methods are available:
Quantitative Western Blotting:
Use recombinant HMGCL protein standards for absolute quantification
Employ fluorescent secondary antibodies for wider linear dynamic range
Normalize to appropriate housekeeping proteins based on tissue type
Analyze using densitometry software with appropriate background correction
Quantitative Immunohistochemistry/Immunofluorescence:
Establish standardized staining protocols with consistent antibody concentrations
Include calibration standards on each slide
Acquire images using identical exposure settings
Analyze using specialized software that quantifies staining intensity and distribution
Consider automated tissue analysis platforms for higher throughput
Flow Cytometry:
Use calibration beads to standardize fluorescence intensity
Report results as molecules of equivalent soluble fluorochrome (MESF)
Analyze median fluorescence intensity (MFI) with appropriate background subtraction
Consider using mean fluorescence intensity ratio (MFIR) by dividing sample MFI by isotype control MFI
ELISA-Based Methods:
Develop sandwich ELISA using capture and detection antibodies against different HMGCL epitopes
Use purified recombinant HMGCL for standard curve generation
Optimize sample preparation to ensure complete protein extraction
When comparing expression across different samples or experimental conditions, standardize all preparation and analysis steps to minimize technical variability .
Several challenges are frequently encountered when using FITC-conjugated HMGCL antibodies:
Photobleaching:
Problem: FITC is relatively susceptible to photobleaching during extended imaging
Solution: Use anti-fade mounting media, minimize exposure during imaging, consider alternative more photostable fluorophores like Alexa Fluor 488 for critical applications
High Background Signal:
Problem: Non-specific binding or autofluorescence in the FITC channel
Solution: Optimize blocking (10% normal serum, 1% BSA), include 0.1-0.3% Triton X-100 in wash buffers, consider tissue autofluorescence quenching agents like Sudan Black B (0.1-0.3%)
Weak Signal Intensity:
Problem: Insufficient antibody concentration or epitope accessibility
Solution: Titrate antibody concentration (try 1:50 instead of 1:200), optimize antigen retrieval methods for fixed tissues, extend incubation time (overnight at 4°C)
Inconsistent Staining Pattern:
Problem: Variability in fixation or processing
Solution: Standardize fixation protocols, establish consistent time intervals between sample collection and fixation, consider using freshly prepared fixatives
Cross-Reactivity:
To improve signal-to-noise ratio in IHC applications:
Antigen Retrieval Optimization:
Antibody Concentration Optimization:
Blocking Enhancements:
For high background, implement dual blocking:
Block endogenous peroxidase activity with 3% H₂O₂ before primary antibody
Use avidin/biotin blocking kit if using biotin-based detection systems
Include 0.3% Triton X-100 in blocking buffer to reduce non-specific binding
Detection System Selection:
For weak signals, employ amplification systems:
Tyramide signal amplification (TSA)
Polymer-based detection systems instead of ABC method
Consider sequential multilayer detection methods
Sample-Specific Considerations:
For tissues with high endogenous biotin (liver, kidney), use non-biotin detection systems
For tissues with high autofluorescence, consider chromogenic rather than fluorescent detection
Modify fixation time for highly vascularized tissues to improve antibody penetration
Data from validated studies shows successful HMGCL detection in human liver, ovary, spleen, and testis tissues using appropriate retrieval methods and dilutions .
A comprehensive control strategy should include:
Specificity Controls:
Peptide Competition/Neutralization: Pre-incubate antibody with excess immunizing peptide to confirm specific binding
Genetic Controls: Include HMGCL knockout/knockdown samples whenever possible
Isotype Controls: Use matched isotype IgG at equivalent concentration to rule out non-specific binding
Technical Controls:
No Primary Antibody: Assess secondary antibody background
Concentration Gradient: Establish optimal antibody dilution through titration experiments
Processing Controls: Process all experimental samples identically and simultaneously
Biological Controls:
Application-Specific Controls:
For Flow Cytometry: Include fluorescence-minus-one (FMO) controls
For IP: Include IgG control IP and reverse IP verification
For WB: Include molecular weight markers and loading controls
Cross-Reactivity Assessment:
Test against closely related proteins, particularly HMGCLL1
Verify species cross-reactivity if working with non-human samples
Implementing this control strategy ensures reliable and reproducible results and facilitates troubleshooting if unexpected results occur .
HMGCL antibodies provide powerful tools for investigating metabolic disorders:
Clinical Sample Analysis:
Evaluate HMGCL protein expression in patient-derived fibroblasts, lymphoblasts, or liver biopsies
Compare protein levels and subcellular localization between patient and control samples
Correlate protein expression with enzymatic activity and clinical phenotypes
Functional Analysis in Disease Models:
Employ tissue-specific knockdown/knockout models to assess HMGCL's role in different organs
Use antibodies to verify knockout/knockdown efficiency
Correlate changes in HMGCL expression with metabolic parameters (ketone body levels, leucine catabolism)
Therapeutic Development:
Monitor HMGCL protein restoration in gene therapy or enzyme replacement approaches
Evaluate subcellular localization of therapeutically delivered HMGCL
Track changes in enzyme expression in response to pharmacological interventions
Biomarker Development:
Investigate correlation between HMGCL protein levels and disease severity
Develop quantitative assays for HMGCL in accessible samples (blood, urine)
Explore tissue-specific HMGCL expression patterns in metabolic disease states
Understanding HMGCL deficiency (hydroxymethylglutaricaciduria) requires detailed characterization of protein expression patterns and functional consequences of mutations, which can be facilitated by specific antibodies .
Investigating the distinct functions of these related proteins requires specialized approaches:
Subcellular Localization Studies:
Use differentially labeled antibodies to simultaneously visualize HMGCL and HMGCLL1
Combine with organelle-specific markers to confirm mitochondrial vs. extramitochondrial localization
Employ super-resolution microscopy to precisely define spatial distribution
Selective Protein Depletion:
Design siRNA/shRNA targeting unique regions of each transcript
Use CRISPR-Cas9 with isoform-specific guide RNAs
Validate knockdown/knockout specificity using antibodies with confirmed selectivity
Metabolic Flux Analysis:
Measure ketone body production and leucine catabolism in cells with selective depletion
Use stable isotope tracing to track metabolic pathways affected by each protein
Correlate metabolic changes with protein expression levels
Protein-Protein Interaction Studies:
Perform co-immunoprecipitation using specific antibodies against each protein
Identify differential binding partners through mass spectrometry
Validate interactions through proximity ligation assays or FRET microscopy
Tissue and Cell Type Distribution:
Map expression patterns of both proteins across tissues and cell types
Correlate expression with metabolic properties of different tissues
Investigate developmental regulation of expression patterns
This approach has revealed that HMGCLL1 is myristoylated and associates with vesicles, suggesting functions distinct from mitochondrial HMGCL in ketogenesis .
Emerging approaches include:
Metabolic Phenotyping of Cancer Cells:
Quantify HMGCL expression across cancer cell lines and tumor samples
Correlate expression with metabolic dependencies and growth characteristics
Investigate association between HMGCL levels and response to metabolic therapies
Spatial Metabolomics Integration:
Combine HMGCL immunohistochemistry with spatial metabolomics techniques
Map regional distribution of ketone body metabolism within heterogeneous tumors
Correlate enzyme expression with local metabolite concentrations
Single-Cell Analysis:
Apply antibodies in single-cell protein analysis platforms (e.g., CyTOF)
Integrate with single-cell RNA-seq to correlate protein and transcript levels
Identify distinct cell populations with unique metabolic profiles
Therapeutic Target Validation:
Use antibodies to validate HMGCL as a potential therapeutic target
Monitor protein expression changes in response to metabolic inhibitors
Develop companion diagnostics for metabolism-targeting therapies
Extramitochondrial HMGCL Function:
Investigate the role of HMGCLL1, particularly in cancer cells that may utilize ketone bodies for lipid synthesis
Explore connections between extramitochondrial HMG-CoA lyase activity and cancer cell growth
Assess potential as a novel biomarker or therapeutic target
Recent research indicates that extramitochondrial HMG-CoA lyase may be crucial to lipid biosynthesis or energy metabolism in certain tissues and cancer cells, opening new avenues for metabolic targeting strategies .