HMBS Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. For specific delivery details, please consult your local distributors.
Synonyms
HEM3_HUMAN antibody; HMBS antibody; Hydroxymethylbilane synthase antibody; PBG D antibody; PBG-D antibody; PBGD antibody; PORC antibody; Porphobilinogen deaminase antibody; porphyria, acute, Chester type antibody; Pre uroporphyrinogen synthase antibody; Pre-uroporphyrinogen synthase antibody; UPS antibody; Uroporphyrinogen I synthase antibody; Uroporphyrinogen I synthetase antibody
Target Names
HMBS
Uniprot No.

Target Background

Function
Hydroxymethylbilane synthase (HMBS) is an enzyme involved in the heme biosynthetic pathway. It catalyzes the sequential polymerization of four molecules of porphobilinogen to form hydroxymethylbilane, also known as preuroporphyrinogen. This process begins with the assembly of the dipyrromethane cofactor by the apoenzyme from two molecules of porphobilinogen or from preuroporphyrinogen. The covalently linked cofactor serves as a primer, around which the tetrapyrrole product is assembled. Finally, the product, preuroporphyrinogen, is released, leaving the cofactor bound to the holodeaminase intact.
Gene References Into Functions
  1. A heterozygous mutation in the HMBS gene was identified in a Chinese female patient and 7 other family members exhibiting classic Acute Intermittent Porphyria (AIP) symptoms. Genetic sequencing revealed a 55 basepair deletion (C.1078_1132delGCCCATTAACTGGTTTGTGGGGCACAGATGCCTGGGTTGCTGCTGTCCAGTGCCT), including the stop codon position, leading to a frameshift mutation. PMID: 30212967
  2. This study investigated the molecular dynamics of human hydroxymethylbilane synthase, focusing on the pyrrole chain elongation process. Specific residues that cause acute intermittent porphyria were identified. PMID: 29632172
  3. A patient underwent extensive diagnostic testing before elevated porphobilinogen levels were detected in their urine. Genetic analysis confirmed the diagnosis of AIP, revealing a previously unreported consensus splice-site mutation IVS4-1G>A in the HMBS gene. PMID: 27769855
  4. This research characterized HMBS mutants in a black South African population, providing insights into the loss of catalytic activity. These findings may inspire future investigations into personalized therapy based on identified molecular lesions. PMID: 27849156
  5. The study observed an approximately 3-fold elevation in ALAS1 mRNA and activity, and a 50% decrease in HMB synthase activity. PMID: 26062020
  6. A novel mutation in intron 2 (IVS2-2Ag-->G) was identified in a Chinese family affected by acute intermittent porphyria. PMID: 26228342
  7. Hepatic cancer tissue from two acute porphyria patients exhibited somatic second-hit mutations, resulting in near-complete inactivation of PPOX and HMBS. PMID: 25445397
  8. This study examined hydroxymethylbilane synthase mutations and polymorphisms in Brazilian families with acute intermittent porphyria. PMID: 25703257
  9. The R173W mutation in the HMBS gene was associated with rhabdomyolysis in patients with variant acute intermittent porphyria. PMID: 25389600
  10. This report describes a novel PBGD missense mutation. PMID: 25870942
  11. Novel porphobilinogen deaminase gene mutations were identified in Polish patients with non-erythroid acute intermittent porphyria. PMID: 25923088
  12. This study investigated the conformational stability and activity of hydroxymethylbilane synthase (HMSB) and the acute intermittent porphyria K132N and V215E HMSB mutations. PMID: 23815679
  13. A remote founder phenomenon was strongly suggested for Venezuelan acute intermittent porphyria families carrying an unreported but frequently occurring HMBS mutation due to dense geographic aggregation with one identical haplotype. PMID: 20978940
  14. This study explored the reliability of using TATA-binding protein (TBP) alone or in combination with hydroxymethylbilane synthase (HMBS) as endogenous controls for normalizing qRT-PCR data in hepatoma cell lines treated with TNF-alpha. PMID: 23811755
  15. The novel mutations identified in the HMBS gene included three missense (R26L, R173G, and D178H), two frameshift (c.749_765dup and c.874insC), and one intronic deletion (IVS12+3_+11delAGGGCCTGT). PMID: 19656452
  16. Informative SNPs of the HMBS gene revealed a distinctive haplotype associated with the R116W mutation found in Dutch AIP families (-64T, 1345 G, 2479 G, 3581 G, 6479 T, 7064 C, and 8578 A). PMID: 19656453
  17. A small deletion and six nucleotide substitutions within the 5'UTR and housekeeping promoter of the HMBS gene were identified as responsible for the non-erythroid form of acute intermittent porphyria. PMID: 22748422
  18. Two novel porphobilinogen deaminase mutations were identified in acute intermittent porphyria patients with accompanying anemia in mainland China. PMID: 21669542
  19. A monoallelic missense mutation p.Arg201Gly (c.601CNG) in the HMBS gene was identified in a patient exhibiting Lichen sclerosus et atrophicus-like skin lesions. PMID: 20580577
  20. This study explored the molecular genetics of acute intermittent porphyria in Russia, conducting mutation analysis and functional polymorphism searches within the porphobilinogen deaminase gene. PMID: 20536026
  21. Novel missense mutations were discovered in the human hydroxymethylbilane synthase gene in patients with acute intermittent porphyria. PMID: 11399210
  22. Molecular analysis of the HMBS gene in acute intermittent porphyria patients and family members of Italian ancestry revealed 13 distinct mutations among 14 patients, with 7 being newly identified. PMID: 11831862
  23. This study highlighted the heterogeneity of molecular defects causing AIP in Polish patients, with 40% of identified mutations being novel. PMID: 11857754
  24. A novel mutation in the PBGD gene was identified in a patient with acute intermittent porphyria exhibiting severe and bilateral axonal radial motor neuropathy. PMID: 11940707
  25. This study describes a mutation resulting in an inactive holo-protein, existing as a complex with two substrate molecules covalently bound to the dipyrromethane cofactor, arising from the reaction between the apo-protein and pre-uroporphyrinogen. PMID: 12773194
  26. The modulation of penetrance by the wild-type allele in dominantly inherited erythrohepatic and acute hepatic porphyrias was investigated using HMBS. PMID: 14669009
  27. DNA screening of 20 unrelated individuals identified 20 different mutations, 11 of which were previously unreported. These novel mutations affected intron 1, exon 5, intron 6, intron 7, intron 9, intron 13, and exon 15. PMID: 15003823
  28. Recurrent mutations G111R and R173Q occurring at CpG motifs in the porphobilinogen deaminase gene were observed in acute intermittent porphyria patients. PMID: 15669678
  29. Three novel mutations within the HMBS gene were associated with acute intermittent porphyria. PMID: 16211556
  30. Sequencing of the hydroxymethylbilane synthase and uroporphyrinogen decarboxylase genes confirmed the relatively uncommon diagnosis of dual porphyria and revealed a novel uroporphyrinogen decarboxylase mutation. PMID: 16390615
  31. The R173W mutation may play a role in acute intermittent porphyria. PMID: 16817012
  32. This study demonstrates that the PBGD cellular pool is regulated by proteasome activity, which is downregulated by hemin or upregulated by Pb-ALAD. PMID: 16935474
  33. A comprehensive search for mutations in Slavic acute intermittent porphyria patients identified 65 mutations, indicating no distinct prevalence of specific mutations among Slavs. PMID: 17298216
  34. A nonsense mutation in the porphobilinogen deaminase gene on chromosome 11q23.3, associated with acute intermittent porphyria, was identified as the underlying genetic defect in Chester porphyria. PMID: 17298217
  35. A patient exhibiting motor neuropathy showed gradual improvement after hematin treatment. Molecular analysis of the porphobilinogen deaminase gene revealed a short segment deletion. PMID: 17459418
  36. Molecular analyses of the PBGD gene identified a novel mutation in exon 15, the 973insG. PMID: 18070416
  37. A mother and two children presented with acute intermittent porphyria due to a C insertion in exon 14. PMID: 18405488
  38. Recombinant T59I and V215M exhibited residual activity of 80.6% and 19.4%, respectively, compared to the wild-type enzyme. PMID: 18406650
  39. A 12-bp deletion mutation leading to a 4-amino acid deletion (AA positions 337 to 340) was identified in a patient and two family members. PMID: 18554962
  40. Molecular analysis of 24 unrelated Chinese acute intermittent porphyria patients from Taiwan identified 25 HMBS mutations: 10 missense (40%), 4 nonsense (16%), 5 frameshift (20%), and 6 splice site (24%) mutations. PMID: 18627369
  41. The determined structure provides insights into the mechanism of action of PBGD at the molecular level, potentially aiding in the development of drugs for upregulating PBGD activity in acute intermittent porphyria. PMID: 18936296
  42. Under hypoxic conditions, the decrease in ubiquitous HMBS and UROS mRNA expression is associated with the accumulation of hypoxia-inducible factor 1alpha protein. PMID: 19021769
  43. Six mutations in exons common to housekeeping and erythroid-specific isoforms were evaluated at the structural level based on the 3-D structure of the E. coli enzyme. The novel missense c.95G>C(p.R32P) is the first de novo mutation identified in the Israeli AIP population. PMID: 19138865
  44. The structure of the human uPBGD (ubiquitous PBGD) mutant, R167Q, was determined by X-ray crystallography and refined to 2.8 Å resolution. PMID: 19207107
  45. Molecular analyses of the hydroxymethylbilane synthase gene revealed seven mutations in Acute intermittent porphyria. PMID: 19292878

Show More

Hide All

Database Links

HGNC: 4982

OMIM: 176000

KEGG: hsa:3145

STRING: 9606.ENSP00000278715

UniGene: Hs.82609

Involvement In Disease
Acute intermittent porphyria (AIP)
Protein Families
HMBS family
Subcellular Location
Cytoplasm.
Tissue Specificity
[Isoform 1]: Is ubiquitously expressed.; [Isoform 2]: Is found only in erythroid cells.

Q&A

What is HMBS and why is it significant in biomedical research?

HMBS (Hydroxymethylbilane Synthase) is a protein-coding gene that encodes the third enzyme in the heme biosynthetic pathway. The enzyme catalyzes the sequential polymerization of four porphobilinogen molecules to form hydroxymethylbilane, also known as preuroporphyrinogen . This process begins with the assembly of the dipyrromethane cofactor from two molecules of porphobilinogen, which then acts as a primer around which the tetrapyrrole product is assembled .

The significance of HMBS in research stems from its critical role in heme metabolism and its association with acute intermittent porphyria, an autosomal dominant disease characterized by neurological dysfunctions, hypertension, abdominal pain, and excessive amounts of aminolevulinic acid and porphobilinogen in the urine . Studying HMBS using antibody-based techniques enables researchers to:

  • Investigate heme biosynthesis pathway regulation

  • Explore mechanisms underlying porphyria-related disorders

  • Understand metabolic adaptations in various physiological and pathological conditions

  • Develop potential therapeutic strategies for porphyria

What types of HMBS antibodies are available for research applications?

Researchers can choose from several types of HMBS antibodies optimized for different experimental applications:

Polyclonal antibodies:

  • Recognize multiple epitopes on the HMBS protein

  • Examples include Thermo Fisher Scientific's PA5-62230 and Aviva Systems Biology's OACD04224

  • Generally offer higher sensitivity but may have increased background

Monoclonal antibodies:

  • Recognize specific epitopes with high specificity

  • Include products like Proteintech's 14797-1-AP

  • Provide consistent results across experiments with lower batch-to-batch variation

Available HMBS antibodies have been validated for multiple applications including:

ApplicationExample DilutionsValidated Cell Lines
Western Blot1:500-1:2000 (Proteintech)HeLa, A375, HEK-293, Raji
Immunofluorescence/ICC1:20-1:200 (Proteintech)MCF-7, HeLa, HepG2
Immunohistochemistry1:10-100 (paraffin sections)Multiple tissue types
Immunoprecipitation0.5-4.0 μg for 1-3 mg lysateHEK-293
ELISA1:100-1:5,000Various sample types

When selecting an HMBS antibody, researchers should consider the specific application requirements, species reactivity, and the region of HMBS targeted by the antibody .

What is the molecular information for HMBS and how does it help in antibody selection?

Understanding the molecular characteristics of HMBS is crucial for appropriate antibody selection and experimental design:

Key molecular characteristics:

  • Full name: Hydroxymethylbilane synthase

  • Calculated molecular weight: 39 kDa

  • Observed molecular weight on gels: 39-42 kDa

  • Gene ID (NCBI): 3145

  • UniProtKB/Swiss-Prot: P08397

Protein aliases to be aware of when searching literature:

  • Porphobilinogen deaminase (PBGD)

  • PBG-D

  • UPS

  • PORC

  • Pre-uroporphyrinogen synthase

Previous HGNC symbols for HMBS gene:

  • PBGD

  • UPS

  • PORC

This information helps researchers:

  • Verify antibody specificity by confirming the observed molecular weight matches expectations

  • Cross-reference between databases when searching for information

  • Understand the various nomenclature used in scientific literature

  • Identify the specific isoforms or regions targeted by different antibodies

What are the optimal protocols for Western blotting using HMBS antibodies?

For successful Western blot detection of HMBS, consider the following methodological approach:

Sample preparation:

  • Extract proteins using standard lysis buffers (RIPA or NP-40 based buffers)

  • Include protease inhibitors to prevent degradation

  • Determine protein concentration using BCA or Bradford assay

  • Load 10-30 μg total protein per lane

Electrophoresis and transfer:

  • Use 10-12% SDS-PAGE gels (HMBS is ~39-42 kDa)

  • Transfer to PVDF or nitrocellulose membrane

  • Verify transfer efficiency with reversible staining (Ponceau S)

Antibody incubation and detection:

  • Block with 5% non-fat milk or BSA in TBST

  • Incubate with anti-HMBS antibody:

    • Proteintech 14797-1-AP: 1:500-1:2000 dilution

    • Aviva OACD04224: 1:50-400 dilution

  • Wash thoroughly with TBST (3-5 times, 5-10 minutes each)

  • Incubate with appropriate HRP-conjugated secondary antibody

  • Develop using chemiluminescence detection system

Validated positive controls:

  • HeLa cells, A375 cells, HEK-293 cells, Raji cells

Troubleshooting:

  • If multiple bands appear: Increase antibody dilution or try different blocking reagent

  • If no signal: Decrease antibody dilution, increase protein loading, or verify sample preparation

  • For high background: Extend washing steps and increase detergent concentration

Optimization of antibody concentration is critical - start with the manufacturer's recommended dilution range and adjust based on signal-to-noise ratio.

How should immunofluorescence experiments with HMBS antibodies be designed?

Immunofluorescence (IF) and immunocytochemistry (ICC) with HMBS antibodies require careful optimization:

Cell preparation:

  • Culture cells on glass coverslips or chamber slides

  • Fix with 4% paraformaldehyde (10-15 minutes at room temperature)

  • Permeabilize with 0.1-0.5% Triton X-100 in PBS (5-10 minutes)

  • Block with 5-10% normal serum (from secondary antibody species) for 1 hour

Antibody staining:

  • Incubate with primary HMBS antibody:

    • Proteintech 14797-1-AP: 1:20-1:200 dilution

    • Aviva OACD04224: 1:50-500 dilution for formalin-fixed cells

  • Incubate overnight at 4°C or 1-2 hours at room temperature

  • Wash 3-5 times with PBS

  • Incubate with fluorophore-conjugated secondary antibody for 1 hour at room temperature

  • Counterstain nuclei with DAPI

  • Mount with anti-fade mounting medium

Validated cell lines:

  • MCF-7 cells, HeLa cells, HepG2 cells

Expected localization:
HMBS is primarily cytoplasmic, so expect diffuse cytoplasmic staining pattern.

Controls to include:

  • Primary antibody omission control

  • Isotype control

  • Positive control using cells known to express HMBS

  • Negative control using cells with HMBS knockdown (if available)

Imaging considerations:

  • Capture images using similar exposure settings across samples

  • Include scale bars in all images

  • Consider Z-stack acquisition for thick samples

  • Use appropriate filter sets to minimize bleed-through

What are the best practices for immunoprecipitation with HMBS antibodies?

Immunoprecipitation (IP) allows isolation of HMBS protein complexes for downstream analysis:

Sample preparation:

  • Prepare cell lysates in non-denaturing lysis buffer (typically containing 1% NP-40 or Triton X-100)

  • Include protease inhibitors and phosphatase inhibitors if studying phosphorylation

  • Clear lysates by centrifugation (14,000 × g, 10 minutes, 4°C)

  • Optional: Pre-clear with Protein A/G beads to reduce non-specific binding

Immunoprecipitation protocol:

  • Add HMBS antibody to cleared lysate:

    • Proteintech 14797-1-AP: 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate

    • Aviva OACD04224: Follow manufacturer's recommendation

  • Incubate overnight at 4°C with gentle rotation

  • Add protein A/G beads and incubate 2-4 hours at 4°C

  • Wash beads 4-5 times with lysis buffer

  • Elute bound proteins with SDS sample buffer or specific elution buffer

  • Analyze by Western blotting or mass spectrometry

Validated samples:

  • HEK-293 cells have been validated for IP with Proteintech antibody

Critical controls:

  • IgG control - use the same amount of isotype-matched IgG

  • Input control - load 5-10% of pre-IP lysate

  • Supernatant control - analyze unbound fraction to assess IP efficiency

Troubleshooting:

  • Low IP efficiency: Increase antibody amount, extend incubation time, optimize lysis conditions

  • High background: Increase washing stringency, use more selective lysis buffer

  • Non-specific bands: Use crosslinking to reduce antibody chain detection

How can HMBS antibodies be optimized for immunohistochemistry in different tissue types?

Immunohistochemistry (IHC) with HMBS antibodies requires tissue-specific optimization:

Tissue preparation:

  • For FFPE tissues:

    • Fix in 10% neutral buffered formalin for 24-48 hours

    • Process and embed in paraffin

    • Cut 4-5 μm sections

  • For frozen tissues:

    • Snap freeze in OCT compound

    • Cut 5-10 μm cryosections

    • Fix in cold acetone or 4% PFA

Antigen retrieval optimization:

  • Heat-induced epitope retrieval:

    • Citrate buffer (pH 6.0) - Start with this standard approach

    • EDTA buffer (pH 9.0) - Alternative for some epitopes

    • Optimize heating time (10-30 minutes)

  • Enzymatic retrieval (alternative):

    • Proteinase K (5-15 minutes)

    • Trypsin (5-15 minutes)

Staining protocol:

  • Block endogenous peroxidase with 3% H₂O₂

  • Block non-specific binding with serum

  • Incubate with HMBS antibody:

    • Aviva OACD04224: 1:10-100 for paraffin sections, 1:50-500 for frozen sections

  • Apply detection system (ABC, polymer-based)

  • Develop with DAB or other chromogen

  • Counterstain with hematoxylin

Tissue-specific considerations:

  • Liver: Often shows higher HMBS expression; may require higher antibody dilution

  • Brain: May need extended antigen retrieval

  • Highly pigmented tissues: Consider using alternative chromogens to distinguish from endogenous pigment

Quantification approaches:

  • H-score method (combines intensity and percentage of positive cells)

  • Digital image analysis for more objective assessment

  • Multiplex IHC for contextual information

How can researchers verify HMBS antibody specificity in their experimental system?

Verifying antibody specificity is critical for reliable research findings. Implement these methodological approaches:

Genetic validation:

  • CRISPR knockout validation:

    • Generate HMBS knockout cells

    • Compare antibody reactivity between wild-type and knockout

    • Complete absence of signal in knockout cells confirms specificity

  • siRNA/shRNA knockdown:

    • Reduce HMBS expression with targeted siRNA

    • Confirm knockdown by qPCR

    • Observe corresponding reduction in antibody signal

Biochemical validation:

  • Peptide competition:

    • Pre-incubate antibody with immunizing peptide

    • Use this mixture in parallel with regular antibody

    • Specific signal should be blocked by peptide competition

  • Recombinant protein controls:

    • Test antibody against purified recombinant HMBS

    • Verify detection at expected molecular weight

Orthogonal validation:

  • Multiple antibodies approach:

    • Use antibodies targeting different HMBS epitopes

    • Consistent results across antibodies suggest specificity

  • Correlation with mRNA:

    • Compare protein expression (antibody) with mRNA levels (qPCR)

    • Similar patterns support antibody specificity

Application-specific controls:

  • For Western blot:

    • Verify single band at 39-42 kDa

    • Check positive control samples (HeLa, HEK-293 cells)

  • For IF/IHC:

    • Confirm expected subcellular localization

    • Include primary antibody omission controls

What approaches can be used to study HMBS in relation to acute intermittent porphyria?

Acute intermittent porphyria (AIP) is directly linked to HMBS mutations, making HMBS antibodies valuable tools for investigating this disorder:

Expression analysis in patient samples:

  • Western blot quantification:

    • Compare HMBS protein levels between AIP patients and controls

    • Correlate protein levels with clinical manifestations

    • Analyze expression in different tissues using tissue lysates

  • Immunohistochemical assessment:

    • Examine HMBS distribution in liver biopsies

    • Compare cellular and subcellular localization between patients and controls

    • Quantify expression differences using digital pathology

Mutation impact studies:

  • In vitro expression systems:

    • Express wild-type and mutant HMBS in cell models

    • Use antibodies to assess protein stability and localization

    • Correlate with enzymatic activity measurements

  • Structural consequence assessment:

    • Immunoprecipitate wild-type and mutant HMBS

    • Analyze structural differences using limited proteolysis

    • Examine potential protein-protein interaction changes

Therapeutic development applications:

  • Compound screening:

    • Use antibodies to monitor HMBS stabilization by small molecules

    • Develop high-throughput screening assays

    • Validate hits in patient-derived cells

  • Gene therapy assessment:

    • Evaluate restoration of HMBS expression after gene delivery

    • Compare exogenous vs. endogenous protein levels

    • Monitor tissue-specific expression

Methodological considerations:

  • Include both wild-type and mutant HMBS controls

  • Consider using antibodies that specifically detect common HMBS mutations

  • Combine with enzymatic activity assays for functional correlation

  • Account for tissue-specific expression patterns

How can mass spectrometry be combined with HMBS antibodies for advanced protein analysis?

Integrating antibody-based enrichment with mass spectrometry provides powerful analytical capabilities:

Immuno-mass spectrometry workflow:

  • Sample preparation:

    • Digest protein samples with trypsin

    • Add stable isotope-labeled HMBS peptide standards

  • Antibody enrichment:

    • Couple anti-HMBS antibodies to magnetic beads

    • Incubate digested samples with antibody-beads

    • Wash to remove non-specific binders

    • Elute bound peptides

  • MS analysis:

    • Develop multiple reaction monitoring (MRM) assays for HMBS peptides

    • Monitor multiple transitions per peptide

    • Quantify based on endogenous/labeled peptide ratios

Applications in HMBS research:

  • Absolute quantification:

    • Measure precise HMBS concentrations in biological samples

    • Compare levels across different tissues or conditions

    • Establish reference ranges for diagnostic applications

  • Post-translational modification analysis:

    • Identify phosphorylation, acetylation, or other modifications

    • Compare modification patterns between normal and disease states

    • Correlate modifications with enzymatic activity

  • Protein interaction studies:

    • Immunoprecipitate HMBS complexes

    • Identify interacting partners by MS

    • Validate interactions using orthogonal methods

Technical considerations:

  • Select antibodies that don't preferentially bind modified peptides

  • Include appropriate controls (e.g., immunoprecipitation from HMBS knockout samples)

  • Optimize digestion conditions for complete proteolysis

  • Consider immunoprecipitation before or after protein digestion

This approach combines antibody specificity with the analytical power of mass spectrometry, enabling detailed characterization of HMBS protein in complex samples .

What are common technical challenges when working with HMBS antibodies and how can they be addressed?

Researchers may encounter several challenges when working with HMBS antibodies. Here are methodological solutions for common issues:

Western blotting challenges:

IssuePotential CausesSolutions
Multiple bandsNon-specific binding, degradation, isoformsIncrease antibody dilution (1:1000-1:2000), use fresh samples with protease inhibitors, optimize blocking conditions
Weak signalLow protein expression, insufficient antibody, poor transferIncrease protein loading, decrease antibody dilution, optimize transfer conditions, use enhanced chemiluminescence
High backgroundInsufficient blocking, inadequate washing, high antibody concentrationExtend blocking time, increase wash duration and number, use alternative blocking agents (BSA vs. milk)

Immunostaining challenges:

IssuePotential CausesSolutions
Non-specific stainingInadequate blocking, high antibody concentrationOptimize blocking (try 5% BSA, normal serum, or commercial blockers), increase antibody dilution to 1:100-1:200
Weak or no signalInsufficient antigen retrieval, low expressionOptimize antigen retrieval (try longer heating times or different buffers), use signal amplification systems
Inconsistent resultsProtocol variations, antibody instabilityStandardize protocol steps, avoid freeze-thaw cycles of antibody, prepare fresh working solutions

Immunoprecipitation challenges:

IssuePotential CausesSolutions
Poor pull-down efficiencyInsufficient antibody, inadequate bindingIncrease antibody amount (2-4 μg), extend incubation time (overnight at 4°C), optimize lysis buffer
Co-IP contaminationNon-specific bindingPre-clear lysates more thoroughly, use more stringent washing conditions, add mild detergents to wash buffer
Heavy/light chain interferenceAntibody chains in Western blotUse HRP-conjugated protein A/G, use antibodies from different species, use clean blot detection system

General optimization approaches:

  • Titrate antibody concentrations to determine optimal working dilution

  • Include positive controls (HeLa, HEK-293 cells) in each experiment

  • Run parallel negative controls (primary antibody omission, isotype control)

  • Prepare all buffers fresh and maintain consistent incubation times/temperatures

How can researchers design experiments to study HMBS in the context of the heme biosynthesis pathway?

Investigating HMBS within the broader heme biosynthesis pathway requires careful experimental design:

Pathway analysis approaches:

  • Multi-protein detection:

    • Design antibody panels targeting multiple heme pathway enzymes

    • Perform Western blots or multiplex immunofluorescence

    • Analyze coordinated expression changes across the pathway

  • Enzyme activity correlation:

    • Measure HMBS protein levels using antibody-based methods

    • Perform parallel enzyme activity assays

    • Correlate protein expression with enzymatic function

  • Regulatory mechanism investigation:

    • Use HMBS antibodies to identify transcription factor binding via ChIP

    • Study post-translational modifications affecting enzyme activity

    • Examine protein-protein interactions through co-immunoprecipitation

Experimental models:

  • Cell culture systems:

    • Erythroid cell lines (K562, MEL) with high heme synthesis

    • Hepatocytes with regulated HMBS expression

    • Genetic manipulation models (CRISPR, RNAi)

  • Tissue analysis:

    • Liver and bone marrow (major heme synthesis sites)

    • Compare expression across tissues with immunohistochemistry

    • Analyze pathway coordination in disease states

Methodological considerations:

  • Pathway perturbation strategies:

    • Heme precursor supplementation or depletion

    • Hypoxia or oxidative stress induction

    • Drug treatments affecting heme metabolism

  • Temporal dynamics:

    • Time-course experiments after pathway stimulation

    • Pulse-chase studies to assess protein turnover

    • Circadian rhythm analysis of pathway components

  • Control selections:

    • Use housekeeping proteins unaffected by heme metabolism

    • Include both positive regulation and negative feedback controls

    • Consider tissue-specific expression patterns

These approaches enable comprehensive analysis of HMBS within its biological context, providing insights into pathway regulation and disease mechanisms.

How should HMBS antibodies be validated for use in multiplexed immunoassays?

Multiplexed immunoassays require rigorous validation to ensure reliable detection of multiple targets simultaneously:

Pre-multiplexing validation:

  • Single-target optimization:

    • Validate each antibody individually before multiplexing

    • Determine optimal working concentrations

    • Confirm specificity using appropriate controls

  • Cross-reactivity assessment:

    • Test each antibody against all targets in the panel

    • Perform sequential staining to identify potential interference

    • Conduct antibody omission controls for each component

  • Spectral compatibility verification:

    • For fluorescence-based assays, assess fluorophore spectral overlap

    • Create single-color controls for each fluorophore

    • Test various fluorophore combinations to minimize bleed-through

Multiplexing strategies for HMBS studies:

ApplicationMethodological ApproachValidation Requirements
Multiplex IHC/IFSequential staining with careful antibody stripping between roundsComplete removal of previous antibodies must be verified; tissue integrity must be maintained
Multiplex flow cytometrySimultaneous staining with spectrally distinct fluorophoresCompensation controls; Fluorescence Minus One (FMO) controls; isotype controls
Multiplex Western blottingDifferent size targets on same blot or reprobing with strippingComplete stripping must be confirmed; different species antibodies preferred
Multiplex bead assaysSpatially or spectrally encoded beads with specific antibodiesCross-reactivity testing; standard curves for each analyte; spike-recovery tests

Advanced validation approaches:

  • Orthogonal confirmation:

    • Verify multiplexed results with single-plex assays

    • Compare results from different methodological platforms

    • Correlate with gene expression or other orthogonal measures

  • Reference standards:

    • Include calibrated reference samples in each run

    • Use samples with known target concentrations

    • Establish internal control metrics for assay performance

  • Statistical validation:

    • Assess intra-assay and inter-assay variability

    • Determine limits of detection for each target

    • Validate dynamic range in multiplexed format

These validation approaches ensure that multiplexed assays involving HMBS antibodies provide reliable and reproducible results across experiments .

How can nanobody technology be applied to HMBS research?

Nanobodies (single-domain antibodies) represent an emerging technology with significant potential for HMBS research:

Advantages of nanobodies over conventional antibodies:

  • Small size (~15 kDa vs. ~150 kDa for IgG)

  • High stability and solubility

  • Efficient tissue penetration

  • Access to cryptic epitopes

  • Cost-effective production

  • Simple engineering into multivalent formats

Applications in HMBS research:

  • Live-cell imaging:

    • Generate fluorescently tagged anti-HMBS nanobodies

    • Track HMBS dynamics in living cells

    • Study real-time changes in localization and interactions

  • Super-resolution microscopy:

    • Use nanobodies for improved resolution due to reduced linkage error

    • Achieve precise localization of HMBS in subcellular compartments

    • Perform multi-color super-resolution with orthogonal nanobodies

  • Intracellular targeting:

    • Express intrabodies targeting HMBS in living cells

    • Disrupt specific interactions or functions

    • Create targeted protein degradation systems

  • Therapeutic exploration:

    • Develop nanobodies targeting mutant HMBS forms

    • Create delivery systems for enzyme replacement

    • Design diagnostics for porphyria-related disorders

Methodological approaches:

  • Nanobody development:

    • Immunize camelids with purified HMBS protein

    • Construct phage display libraries from B cells

    • Select specific binders through panning

    • Characterize binding properties and epitopes

  • Engineering strategies:

    • Create bispecific constructs targeting HMBS and interacting proteins

    • Develop nanobody-based biosensors for HMBS activity

    • Fuse with cell-penetrating peptides for intracellular delivery

The evolutionary innovation of heavy-chain antibodies in camelids has enabled this technology, which offers significant advantages for certain research and therapeutic applications .

What are emerging methods for developing and validating antibodies for rare proteins like HMBS?

Advanced technologies are transforming antibody development and validation processes:

Next-generation antibody development approaches:

  • Recombinant antibody technologies:

    • Phage display libraries for antibody selection

    • Yeast display for affinity maturation

    • Mammalian display for functional screening

    • Selection under defined conditions for application-specific antibodies

  • In silico design and screening:

    • Computational prediction of antibody structures

    • Virtual screening of antibody-antigen interactions

    • Knowledge-based approaches like AbPredict algorithm

    • Machine learning for epitope prediction

  • Synthetic antibody libraries:

    • Rationally designed frameworks with diverse CDRs

    • Scaffold-based binding proteins

    • Minimalist antibody designs

High-throughput validation methodologies:

  • Automated screening platforms:

    • Parallel testing of multiple antibody candidates

    • Robotics-based sample handling and analysis

    • High-content imaging for cellular assays

  • Developability assessment:

    • Early-stage evaluation of antibody properties

    • Predicting stability, solubility, and manufacturability

    • High-throughput biophysical characterization

  • Integrated validation workflows:

    • Combining multiple testing platforms

    • Standardized protocols across applications

    • Comprehensive data management systems

Example integrated workflow from discovery to validation:

StageTechnologiesMetrics
Antigen designIn silico epitope prediction, protein structure analysisEpitope accessibility, conservation, uniqueness
Antibody generationPhage display, recombinant expression, hybridomaBinding affinity, epitope coverage
Initial screeningELISA, flow cytometry, Western blotBinding specificity, sensitivity
Advanced validationGenetic knockout controls, orthogonal methodsTarget specificity, application performance
Developability assessmentThermal stability, aggregation propensityShelf-life, batch consistency

These advances in antibody technology enable the development of higher quality reagents for challenging targets like HMBS, supporting more reliable and reproducible research outcomes .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.