ALB Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production Methodology

Recombinant ALB monoclonal antibodies are generated through:

  • Gene Cloning: Isolation of antibody-coding DNA sequences from immunized host animals (e.g., rabbits or mice) .

  • In Vitro Expression: Transfection of plasmids into mammalian cells (e.g., HEK293) for antibody production, bypassing hybridoma instability .

  • Screening: Selection of high-affinity clones via ELISA or flow cytometry .

This method ensures minimal batch-to-batch variability compared to traditional hybridoma techniques .

Key Advantages

FeatureBenefit
Batch ConsistencyDefined genetic sequences eliminate variability from genetic drift .
High SpecificityTargets ALB with minimal cross-reactivity (e.g., AE00131 shows S-scores >2.5 against 19,000 human proteins) .
ScalabilityRapid production in bioreactors supports large-scale studies .
EngineerabilityModifiable Fc regions reduce non-specific binding in assays .

Diagnostic and Experimental Use

  • Immunohistochemistry (IHC): Stains extracellular albumin in hepatocellular carcinoma at 1–3 µg/ml .

  • Western Blotting: Detects albumin in human, mouse, and rat samples (e.g., JF32-10) .

  • Flow Cytometry: Utilizes Fc-engineered variants to minimize background noise .

Clinical Relevance

While primarily used in research, ALB antibodies have potential therapeutic roles in:

  • Liver Disease Monitoring: Tracking albumin leakage in cirrhosis .

  • Drug Delivery: Albumin-binding antibodies could enhance pharmacokinetics of conjugated therapeutics .

Specificity Testing (AE00131 Clone)

ParameterResult
Host SpeciesMouse
IsotypeIgG1, κ
ReactivityHuman
Cross-ReactivityNone detected (Z-score: 43 for ALB vs. 14 for next-highest protein)

Performance in IHC

AntibodyConcentrationEpitope RetrievalStaining Result
AE001311–2 µg/mlpH6 boiling (10–20 min)Clear extracellular signal in hepatocellular carcinoma
JF32-100.5–2 µg/mlNot specifiedConsistent detection across human, mouse, and rat samples

Research Limitations and Future Directions

  • Epitope Mapping: Most ALB antibodies lack published epitope details, complicating mechanistic studies .

  • Therapeutic Development: Engineering bispecific ALB antibodies for drug delivery remains exploratory .

Recent advancements in single-cell ASC cloning (e.g., CD138-ferrofluid technology) may accelerate ALB antibody discovery with functional screening .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The ALB recombinant monoclonal antibody production commences with the extraction of ALB antibody genes. These genes are isolated from B cells derived from immunoreactive rabbits, subsequently amplified, and cloned into phage vectors. These vectors are then introduced into mammalian cell lines to facilitate the generation of functional antibodies. The resulting ALB recombinant monoclonal antibody is purified from the culture supernatant of the transfected cell lines using affinity chromatography. This antibody can be effectively employed for recognizing the human ALB protein in ELISA and FC applications.

Serum albumin (ALB) constitutes the most abundant protein within human blood plasma and plays a pivotal role in maintaining the physiological stability and homeostasis of the bloodstream. Its multifaceted capabilities, encompassing the transport, buffering, and protection of various molecules, as well as its influence on blood volume regulation, underscore its critical importance to human health. Dysregulation of albumin levels or function can have significant clinical implications.

Form
Liquid
Lead Time
Typically, we can dispatch the products within 1-3 working days after receiving your orders. Delivery times may vary based on the specific purchasing method or location. Please consult your local distributors for precise delivery times.
Synonyms
Serum albumin, ALB
Target Names
ALB
Uniprot No.

Target Background

Function

Serum albumin exhibits a wide range of binding capabilities, including water, Ca(2+), Na(+), K(+), fatty acids, hormones, bilirubin, and drugs. Its primary function is believed to be the regulation of blood's colloidal osmotic pressure. Notably, it serves as the major zinc transporter in plasma, typically binding approximately 80% of all plasma zinc. Furthermore, it acts as a major calcium and magnesium transporter in plasma, binding roughly 45% of circulating calcium and magnesium. It potentially possesses more than two calcium-binding sites and might additionally bind calcium in a non-specific manner. The shared binding site between zinc and calcium at residue Asp-273 suggests a potential interplay between zinc and calcium transport in the blood. The rank order of affinity is zinc > calcium > magnesium.

Serum albumin binds to the bacterial siderophore enterobactin, inhibiting enterobactin-mediated iron uptake by E.coli from ferric transferrin. This interaction may limit the utilization of iron and subsequently restrict the growth of enteric bacteria such as E.coli. However, it does not prevent iron uptake mediated by the bacterial siderophore aerobactin.

Gene References Into Functions
  1. Relatively low serum concentrations of albumin may constitute an independent risk factor for Mild cognitive impairment in the elderly. PMID: 30249938
  2. The albumin-globulin ratio, in combination with CEA, provides a more precise indicator of poor prognosis and early recurrence in rectal carcinoma patients undergoing preoperative chemoradiotherapy. PMID: 29996133
  3. Patients exhibiting steroid-refractory GI Acute graft versus host disease have demonstrated higher fecal calprotectin levels and lower albumin levels compared to patients with steroid-responsive disease. PMID: 29397331
  4. Lower serum albumin concentrations in patients diagnosed with pulmonary arterial hypertension are associated with higher mortality rates and can serve as a marker of disease severity within this patient population. PMID: 29604093
  5. This meta-analysis suggests that the C-reactive protein/albumin ratio (CAR) may represent a potential prognostic marker in solid cancers. PMID: 28128229
  6. Thiol/Disuldife homeostasis balance and ischemia-modified albumin levels may be utilized as biomarkers to monitor tissue ischemia in diabetic mellitus and to discriminate the different stages of diabetic retinopathy. PMID: 29298534
  7. Mitochondrial dysfunction activates the PINK1/Parkin signaling and mitophagy in renal tubular epithelial cells under albumin overload conditions. PMID: 29494565
  8. Serum ischemia-modified albumin (IMA) levels, along with the prevalence of small for gestational age (SGA), were significantly increased in the pre-eclampsia group. Therefore, cord blood IMA might be a predictive biomarker for SGA in pre-eclampsia pregnancies. PMID: 29570915
  9. By employing a fluorescence activatable nanosensor as the structural indicator, the stepwise conformational transitions of HSA during denaturing and the partial refolding of subdomain IIA of HSA were readily visualized. PMID: 27232658
  10. Serum albumin adduct features were both positively and negatively associated with smoking. This indicates that some adducts arise from reactions between Cys34 and constituents of cigarette smoke (e.g., ethylene oxide and acrylonitrile), while others (Cys34 oxidation products and disulfides) seem to reflect alterations in the serum redox state that resulted in reduced adduct levels in smokers. PMID: 27684351
  11. The increased lipophilicity of sulfur-containing compounds might contribute to their enhanced binding affinity to HSA. PMID: 29128746
  12. The results obtained provide a comprehensive understanding of the interaction between the acid azo dye AO10 and serum albumins. PMID: 29126006
  13. A higher preoperative serum ALB level is associated with improved long-term survival in intrahepatic cholangiocarcinoma patients. PMID: 29672305
  14. The presence of hydroxyl groups is capable of modulating the interaction between riparins and HSA. PMID: 28915468
  15. Spectroscopic and molecular docking studies have been conducted to investigate the interaction of human serum albumin with copper(II) complexes. PMID: 27792986
  16. Aberrant HSA may play a role in the initiation or progression of type 1 diabetes mellitus. PMID: 29051099
  17. Results indicate that albumin from chronic hemodialyzed (CH) patients is highly modified in vivo and is less susceptible to oxidation compared to normal albumin. PMID: 29554100
  18. These findings suggest that low serum albumin levels correlate with increased parameter measures of hepatocellular carcinoma (HCC) aggressiveness, in addition to their role as a monitor of systemic inflammation. Decreased serum albumin might play a role in HCC aggressiveness. PMID: 28862714
  19. A hypothesis has been proposed suggesting that human serum albumin acts as a modulator and transporter of cannabinoid drugs. PMID: 28976704
  20. The CRP/albumin ratio is a novel, promising, easy-to-measure, repeatable, and non-invasive inflammation-based prognostic score for acute pancreatitis. PMID: 28823374
  21. Homodimerization represents a newly described structural alteration of the HA molecule in decompensated cirrhosis. This contributes to the progressive reduction of the monomeric native HA, the only isoform possessing structural and functional integrity. PMID: 27782157
  22. Lower serum albumin concentrations are associated with a poorer prognosis for patients with stable coronary heart disease. PMID: 28413113
  23. Human serum albumin binds to Cu(I) at pH 7.4 with an apparent conditional affinity of KCu(I):serum albumin = 10(14.0), utilizing digonal coordination in a structure similar to the bis-His coordination modes characterized for amyloid beta (Abeta) and the prion protein. PMID: 29166002
  24. The C-reactive protein/albumin ratio (CAR), an inflammation-based prognostic score, is a potential independent predictor for disease progression and death in patients with operable non-small cell lung cancer. PMID: 27823974
  25. These peptides are identified as fragments of albumin. The isolated fragments activate MRGPRX2 and degranulate MRGPRX2 expressing LAD 2 cells in a dose-dependent manner. The isolated basic peptides generated from human albumin are capable of degranulating mast cells via the MRGPRX2. PMID: 28844982
  26. The use of EPI-X4, a fragment of serum albumin, as a urinary biomarker for late acute GvHD was found to be not meaningful in case control studies. PMID: 27042833
  27. Serum ALB levels may be more useful than the degree of fibrosis for predicting hepatocellular carcinoma after sustained virological responses in chronic hepatitis C. PMID: 29040987
  28. Elevated Ischemia-modified albumin levels and Carotid intima-media thickness may be linked to increased oxidative stress and an elevated risk of atherosclerosis in individuals with Obstructive sleep apnea. PMID: 27029471
  29. Peak unbound bilirubin was primarily associated with a decrease in bilirubin albumin binding affinity in infants under 30 weeks gestational age with hyperbilirubinemia. PMID: 29132818
  30. This meta-analysis demonstrates increased ischaemia-modified albumin (IMA) levels in both hypothyroidism and hyperthyroidism patients. This association with thyroid profile suggests that serum IMA could serve as a simple measure of increased oxidative stress in thyroid dysfunction. PMID: 27920278
  31. The optimal cut-off values for serum albumin (ALB), serum globulin (GLB), and albumin/globulin ratio (AGR) in terms of survival were identified to be 45.6 g/L, 26.9 g/L, and 1.30, respectively. PMID: 27105522
  32. Physiological serum copper concentrations commonly observed in malignancies induce unfolding-induced aggregation of human serum albumin in vitro. PMID: 29122590
  33. Despite significant structural homology between HSA and ESA, their crystal complexes with Myr-2S-cPA demonstrate distinctly different modes of binding of this LPL analogue. HSA binds three molecules of Myr-2S-cPA, whereas ESA only binds one. Moreover, none of the identified Myr-2S-cPA binding sites overlap in both albumins. PMID: 27129297
  34. Ghrelin binds to serum albumin, and this interaction influences the hormone's biological activity. PMID: 27431015
  35. Higher levels of serum albumin are associated with a lower mortality rate in patients undergoing maintenance hemodialysis. PMID: 27542730
  36. Results indicate that, in addition to its role as a monitor of systemic inflammation, albumin might play a direct role in inhibiting hepatocellular carcinoma growth. This could occur through the modulation of alpha-fetoprotein or through its actions on growth-controlling kinases. PMID: 28486226
  37. These results provide an unprecedented view of how albumin interacts with Abeta and illustrate the potential of dark-state exchange saturation transfer NMR in mapping the interactions between amyloid-inhibitory proteins and amyloidogenic peptides. PMID: 28798235
  38. The C-reactive protein-to-albumin ratio is a promising predictor of survival and recurrence in patients with rectal cancer treated with chemoradiotherapy followed by surgery. PMID: 28982904
  39. Lower serum albumin levels are associated with venous thromboembolic events in nephrotic syndrome. PMID: 28391310
  40. Data suggest that increased levels of anti-Amadori-albumin autoantibodies may be used as a biomarker for disease diagnosis and its progression. PMID: 28192530
  41. Data indicate that repeated measurements after five hours showed a statistically significant difference in the fractional synthesis rate of fibrinogen but not for albumin. PMID: 28350862
  42. Data suggest that, unlike albumin with low FcRn-binding affinity, albumin with high FcRn-binding affinity (due to genetic variation/genetic engineering) is directed less to lysosomes and more to endosomes. This suggests FcRn-directed albumin salvage from lysosomal degradation. (FcRn = neonatal Fc receptor) PMID: 28637874
  43. The mean albumin level for patients who developed heparin-induced thrombocytopenia-thrombosis (HITT) was significantly lower than that for patients who developed heparin-induced thrombocytopenia (HIT). PMID: 28289828
  44. Data suggest that posttranslational modifications of human serum albumin (HSA) typical of hyperhomocysteinemia (N and Shomocysteinylation) reduce the binding affinity of catechins to HAS. The catechins used in these studies were dietary flavonoids, epicatechin (EC), epigallocatechin (EGC), epicatechin gallate (ECG), and epigallocatechin gallate (EGCG). PMID: 26658763
  45. The studies suggest that impaired endocytosis of megalin/cubilin ligands, hemoglobin, and albumin, rather than heme toxicity, may be the underlying cause of tubular proteinuria in sickle cell disease patients. PMID: 28356267
  46. Decreased serum albumin upon admission is significantly associated with long-term all-cause mortality in hospital survivors of acute myocardial infarction. PMID: 27257851
  47. Systemic inflammation intensity was assessed at all time points using C-reactive protein and albumin as prognostic covariates for overall survival in an extended Cox regression model. Time-dependent effects are a prominent feature of systemic inflammation intensity, serving as a powerful prognostic biomarker for metastatic renal cell carcinoma. PMID: 28618965
  48. The presence of serum albumin in human eye tissues suggests its involvement in metabolism, tissue volume maintenance, and antioxidant reactions. PMID: 28361409
  49. A study was conducted to determine ischemia-modified albumin (IMA) levels in inflammatory bowel diseases (IBD) and IBD subgroups, and to examine their relationship with the disease activity index. IMA levels in IBD were found to be higher compared to the control group. IMA in IBD might be linked to the pathogenesis of the disease and correlated with its severity. PMID: 26642816
  50. Serum ischemia-modified albumin levels were higher in type 2 DM patients than in control subjects and were higher in diabetic patients with diabetic peripheral neuropathy compared to those without neuropathy. PMID: 27965983
Database Links

HGNC: 399

OMIM: 103600

KEGG: hsa:213

STRING: 9606.ENSP00000295897

UniGene: Hs.418167

Involvement In Disease
Hyperthyroxinemia, familial dysalbuminemic (FDAH); Analbuminemia (ANALBA)
Protein Families
ALB/AFP/VDB family
Subcellular Location
Secreted.
Tissue Specificity
Plasma.

Q&A

What is an ALB Recombinant Monoclonal Antibody?

An ALB (Albumin) recombinant monoclonal antibody is a laboratory-engineered protein designed to specifically recognize and bind to albumin, a soluble monomeric protein that comprises approximately half of the blood serum protein. Unlike traditional monoclonal antibodies produced from hybridomas, recombinant monoclonal antibodies are generated in vitro from synthetic genes by cloning antibody genes into expression vectors without using hybridoma technology . This process involves identifying and isolating the genetic sequences coding for the desired antibody, followed by recombinant expression in suitable host systems. The resulting antibodies maintain the specificity of monoclonal antibodies while offering greater consistency and engineering potential .

How are ALB Recombinant Monoclonal Antibodies produced?

The production of ALB recombinant monoclonal antibodies follows a multi-step process that begins with obtaining the genetic sequence of the desired antibody. For rabbit-derived recombinant monoclonal antibodies, the process typically involves cloning specific antibody DNA sequences from immunoreactive rabbits . These sequences are then inserted into expression vectors and introduced into suitable host expression systems such as mammalian cells, bacteria, yeast, or insect cells. Following expression, the antibodies undergo purification and quality control testing to ensure specificity and functionality.

Modern rapid protocols enable the identification and expression of recombinant antigen-specific monoclonal antibodies in less than 10 days using RT-PCR to generate linear Ig heavy and light chain gene expression cassettes, called "minigenes," for rapid expression without traditional cloning procedures . This approach not only saves time but also allows for screening individual antigen-specific antibody-secreting cells (ASCs) for effector function prior to recombinant antibody cloning .

What distinguishes recombinant monoclonal antibodies from traditional antibodies?

Recombinant monoclonal antibodies offer several distinct advantages over traditional polyclonal and hybridoma-derived monoclonal antibodies, as outlined in this comparative analysis:

CharacteristicPolyclonal (pAb)Hybridoma-derived Monoclonal (mAb)Recombinant (rAb)
SpecificityModerate (recognizes multiple epitopes)High (recognizes single epitope)High (recognizes specific epitope)
ReproducibilityLimited (batch-to-batch variation)Virtually reproducibleFully reproducible
Production timeShort (3–4 months)Long (up to a year)Short to moderately long
StabilityHighModerateDepends on the format
Engineering potentialNot possibleOnly after converting to rAbHighly engineerable
Animal dependenceHighHighLow/None (animal-free process)

The key advantages of recombinant antibodies include increased reproducibility, control, and consistency in performance across experiments; faster production time compared to hybridoma technology; easier format switching to create specialized variants; and reduced or eliminated animal use in production . Additionally, recombinant antibodies allow for comprehensive analysis of variable region repertoires in combination with functional assays to evaluate specificity and function .

What are the main research applications for ALB Recombinant Monoclonal Antibodies?

ALB recombinant monoclonal antibodies have diverse applications in biomedical and toxicological research. Given that albumin functions primarily as a carrier protein for steroids, fatty acids, and thyroid hormones and plays a crucial role in stabilizing extracellular fluid volume , these antibodies are valuable tools in multiple research contexts:

  • Protein detection and quantification: Used in Western blotting, ELISA, and immunohistochemistry to detect albumin levels in biological samples.

  • Biomarker research: Employed in studies investigating albumin as a biomarker for liver function, nutritional status, or disease progression.

  • Drug delivery research: Utilized in studies exploring albumin's role as a carrier protein for therapeutic compounds.

  • Structural and functional studies: Applied in research examining albumin's interactions with other biomolecules and its role in maintaining oncotic pressure.

  • Therapeutic development: Used in the development of albumin-targeting therapies or albumin-based drug conjugates.

The high specificity and reproducibility of recombinant antibodies make them particularly valuable for these applications, as they provide consistent results across experiments and can be engineered for specific research needs .

How do ALB Recombinant Monoclonal Antibodies function in experimental settings?

ALB recombinant monoclonal antibodies function by recognizing and binding to specific epitopes on the albumin protein with high affinity and specificity . In experimental settings, these antibodies serve as highly specific detection tools that can identify and quantify albumin in complex biological samples.

The binding mechanism follows the standard antibody-antigen interaction, where the variable regions of the antibody, particularly the complementarity-determining regions (CDRs), form specific contacts with epitopes on the albumin molecule. This binding can be utilized in various experimental techniques:

  • Immunoassays: In ELISA or other immunoassay formats, the antibody captures or detects albumin in solution, allowing for quantification.

  • Immunohistochemistry/Immunofluorescence: The antibody binds to albumin in tissue sections or cells, enabling visualization of albumin distribution.

  • Immunoprecipitation: The antibody can pull down albumin and associated proteins from complex mixtures for further analysis.

  • Flow cytometry: When coupled with fluorophores, these antibodies can detect cell-associated albumin.

The recombinant nature of these antibodies ensures consistent binding properties across experiments, addressing the batch-to-batch variability often encountered with traditional antibodies .

What are the optimal expression systems for ALB Recombinant Monoclonal Antibody production?

The selection of an expression system for ALB recombinant monoclonal antibody production depends on several factors including required yield, post-translational modifications, downstream applications, and resource constraints. Each system offers distinct advantages and limitations:

Mammalian Cell Expression Systems:

  • Preferred for complete recombinant antibodies requiring proper folding and glycosylation

  • Chinese Hamster Ovary (CHO) cells and Human Embryonic Kidney (HEK293) cells are commonly used

  • Advantages: Proper post-translational modifications, high similarity to native antibodies

  • Limitations: Higher cost, longer production time, potential for viral contamination

Bacterial Expression Systems:

  • Escherichia coli is commonly used for smaller antibody fragments (Fab, scFv)

  • Advantages: Rapid growth, high yields, cost-effectiveness, well-established protocols

  • Limitations: Limited post-translational modifications, potential for inclusion body formation requiring refolding

Yeast Expression Systems:

  • Pichia pastoris and Saccharomyces cerevisiae are commonly used

  • Advantages: Higher yields than mammalian cells, some post-translational modifications, secretion capacity

  • Limitations: Different glycosylation patterns than mammalian cells

Insect Cell Expression Systems:

  • Baculovirus expression vectors in insect cells (Sf9, Sf21, High Five)

  • Advantages: Higher yields than mammalian cells, more complex post-translational modifications than bacteria or yeast

  • Limitations: Different glycosylation patterns than mammalian cells

For full-length ALB recombinant monoclonal antibodies requiring all functional properties, mammalian expression systems typically provide the best results despite higher costs . For applications where only the antigen-binding property is required, bacterial expression of antibody fragments may be sufficient and more cost-effective.

How can researchers troubleshoot specificity issues with ALB Recombinant Monoclonal Antibodies?

Specificity issues with ALB recombinant monoclonal antibodies can significantly impact experimental outcomes. Troubleshooting these issues requires a systematic approach:

Validation using multiple techniques:

  • Compare results across different methodologies (ELISA, Western blot, immunohistochemistry)

  • Discrepancies between techniques may indicate context-dependent specificity issues

Cross-reactivity assessment:

  • Test antibody against related proteins or albumin from different species

  • Perform competition assays with purified albumin to confirm specific binding

  • For ALB antibodies, test specifically against closely related serum proteins

Epitope characterization:

  • Identify the specific epitope recognized by the antibody through epitope mapping

  • Verify that epitope accessibility is not compromised by sample preparation methods

  • Consider using antibodies recognizing different epitopes on albumin for confirmation

Sample-specific considerations:

  • Evaluate potential interfering substances in your specific sample type

  • For recombinant ALB antibodies, cross-reactivity to variants of albumin, such as Abeta [1-37], Abeta [1-38], Abeta [1-40], or Abeta [1-43], should be assessed at various antibody concentrations

Antibody engineering solutions:

  • If specificity issues persist, consider antibody engineering to improve specificity

  • Mutation of specific residues in the CDR regions can enhance specificity

  • Alternative antibody formats (e.g., different fragments) may resolve some specificity issues

Careful negative controls:

  • Use samples known to lack albumin or where albumin has been depleted

  • Include isotype controls to identify non-specific binding

By systematically addressing these aspects, researchers can identify and resolve specificity issues with ALB recombinant monoclonal antibodies, ensuring reliable experimental results .

What technical considerations are important when designing experiments with ALB Recombinant Monoclonal Antibodies?

Designing robust experiments with ALB recombinant monoclonal antibodies requires attention to several technical factors:

Antibody characterization:

  • Thoroughly characterize the antibody's specificity, affinity, and cross-reactivity profile

  • Understand the specific epitope recognized and its conservation across species if relevant

  • Consider the antibody format (full-length, Fab, scFv) and how it might impact experimental design

Optimal concentration determination:

  • Titrate antibody concentrations to identify the optimal working concentration

  • For ALB antibodies, consider that cross-reactivity may be concentration-dependent; cross-reactivity to variants might not be observed at low concentrations (up to 30 ng/mL)

Buffer compatibility:

  • Verify compatibility with experimental buffers and conditions

  • Consider albumin's biochemical properties (65,000 Da, globular unglycosylated protein) when designing extraction or preparation methods

Sample preparation considerations:

  • Ensure that sample preparation methods preserve the epitope structure

  • For native albumin detection, avoid denaturing conditions that might alter epitope accessibility

  • Consider that albumin undergoes processing from preproalbumin to proalbumin to the secreted form, which may affect antibody recognition

Appropriate controls:

  • Include positive controls with known albumin concentrations

  • Use negative controls to assess background and non-specific binding

  • Consider using multiple anti-albumin antibodies recognizing different epitopes for confirmation

Detection system optimization:

  • Select detection systems appropriate for the expected albumin abundance

  • Consider signal amplification methods for low-abundance detection

  • Ensure detection system does not interfere with antibody-antigen interaction

Interference mitigation:

  • Be aware that endogenous albumin in biological samples might interfere with assays

  • Consider methods to block or account for endogenous albumin when necessary

By carefully addressing these technical considerations, researchers can design more robust and reproducible experiments using ALB recombinant monoclonal antibodies .

How can ALB Recombinant Monoclonal Antibodies be engineered for enhanced functionality?

Engineering ALB recombinant monoclonal antibodies offers opportunities to enhance their functionality for specific research applications:

Affinity maturation:

  • Directed evolution or rational design approaches can improve binding affinity

  • Site-directed mutagenesis of CDR regions can enhance specificity and reduce cross-reactivity

  • Computational modeling can guide mutation selection for optimized binding

Format modifications:

  • Conversion to various antibody fragments (Fab, F(ab')2, scFv, nanobodies) for altered tissue penetration, clearance, or multivalent binding

  • Creation of bispecific formats to simultaneously target albumin and another protein of interest

  • Development of antibody-drug conjugates for targeted delivery studies

Protein fusion strategies:

  • Fusion with reporter proteins (fluorescent proteins, enzymes) for direct detection

  • Creation of albumin-targeting fusion proteins for drug delivery research

  • Development of chimeric antigen receptors (CARs) incorporating anti-albumin binding domains

Stability engineering:

  • Introduction of stabilizing mutations to improve thermal and pH stability

  • Optimization of frameworks to enhance expression and reduce aggregation

  • Addition of protective modifications to extend half-life in experimental systems

Post-translational modification control:

  • Engineering of glycosylation sites to modulate effector functions

  • Removal of potential deamidation or oxidation sites to improve stability

  • Introduction of site-specific conjugation sites for controlled labeling

Expression optimization:

  • Codon optimization for the selected expression system

  • Signal sequence optimization for improved secretion

  • Removal of problematic sequence motifs that might impair expression

The recombinant nature of these antibodies makes them particularly amenable to engineering approaches, providing researchers with customizable tools for specific experimental needs . The ability to generate these engineered variants without returning to animal immunization represents a significant advantage over traditional antibody production methods.

What are the latest methodological advances in ALB Recombinant Monoclonal Antibody development?

Recent methodological advances in ALB recombinant monoclonal antibody development have significantly enhanced production efficiency, quality, and applicability:

Rapid isolation and expression technologies:

  • Development of ferrofluid-based technologies for isolating single antigen-specific antibody-secreting cells

  • Implementation of RT-PCR techniques to generate linear Ig heavy and light chain gene expression cassettes ("minigenes")

  • Streamlined workflows enabling recombinant antibody generation in less than 10 days

High-throughput screening approaches:

  • Advanced single-cell sorting and analysis platforms for more efficient identification of antibody-producing cells

  • Development of microfluidic systems for rapid antibody screening and characterization

  • Implementation of functional screening assays prior to full antibody production

Advanced bioinformatics tools:

  • Computational approaches for antibody design and optimization

  • Prediction tools for antibody stability, immunogenicity, and developability

  • Database resources integrating antibody sequence, structure, and functional data

Novel expression system developments:

  • Cell-free expression systems for rapid antibody production

  • Engineered host cell lines with humanized glycosylation patterns

  • Development of transient expression systems with improved yields

Animal-free production methods:

  • Fully in vitro antibody discovery platforms using synthetic libraries

  • Display technologies (phage, yeast, mammalian) for antibody selection without animal immunization

  • Computational approaches to design antibodies from scratch

Functional characterization innovations:

  • Advanced epitope mapping technologies including hydrogen-deuterium exchange mass spectrometry

  • Higher-resolution binding kinetics analysis using surface plasmon resonance and bio-layer interferometry

  • Comprehensive assessment of cross-reactivity using proteome arrays

These methodological advances are particularly significant for albumin antibodies given albumin's importance as a carrier protein and its potential role in various physiological and pathological processes . By implementing these technologies, researchers can develop ALB recombinant monoclonal antibodies with improved specificity, reduced development time, and enhanced functionality.

What validation techniques should be employed for ALB Recombinant Monoclonal Antibodies?

Comprehensive validation of ALB recombinant monoclonal antibodies is essential to ensure reliable experimental results. A multi-faceted validation approach should include:

Specificity assessment:

  • Western blot analysis with purified albumin, serum samples, and tissue lysates

  • Immunoprecipitation followed by mass spectrometry to confirm target identity

  • Immunohistochemistry with appropriate positive and negative control tissues

  • Competitive binding assays with purified albumin to demonstrate specific displacement

  • Testing against structurally related proteins to assess cross-reactivity

Sensitivity evaluation:

  • Determining limit of detection using purified albumin standards

  • Comparison with established reference antibodies or detection methods

  • Assessing performance across a range of albumin concentrations

Reproducibility testing:

  • Inter-lot consistency assessment to verify manufacturing reproducibility

  • Multiple-user testing to ensure method robustness

  • Cross-platform performance evaluation (e.g., different ELISA formats, flow cytometry, IHC)

Functionality verification:

  • Application-specific validation in the experimental context of intended use

  • Appropriate positive and negative controls for each application

  • Evaluation in different buffer conditions relevant to intended applications

Epitope characterization:

  • Epitope mapping to identify the specific binding region on albumin

  • Assessment of epitope accessibility in different sample preparation conditions

  • Evaluation of epitope conservation across species if cross-reactivity is desired

Sequence validation:

  • Verification of the recombinant antibody sequence

  • Confirmation of proper folding and post-translational modifications

  • Assessment of batch-to-batch consistency through sequencing and functional assays

Application-specific controls:

  • For sandwich ELISA formats, testing for cross-reactivity to various albumin forms, such as Abeta [1-37], Abeta [1-38], Abeta [1-40], or Abeta [1-43]

  • For immunohistochemistry, inclusion of absorption controls with purified albumin

By implementing this comprehensive validation approach, researchers can ensure that ALB recombinant monoclonal antibodies perform reliably in their specific experimental contexts .

How can researchers assess cross-reactivity of ALB Recombinant Monoclonal Antibodies?

Thorough assessment of cross-reactivity is crucial for ensuring the specificity of ALB recombinant monoclonal antibodies. A systematic approach includes:

Sequence-based prediction:

  • Computational analysis to identify proteins with sequence similarity to albumin

  • Particular attention to other serum proteins and albumin family members

  • Consideration of species-specific variants if working across multiple species

Protein panel testing:

  • Testing against a panel of purified proteins including:

    • Albumin variants (preproalbumin, proalbumin, and processed albumin)

    • Related serum proteins

    • Albumin from different species if relevant

  • Concentration-dependent testing, as cross-reactivity may only appear at higher antibody concentrations

Immunoassay-based cross-reactivity assessment:

  • ELISA testing against potential cross-reactive proteins

  • Western blot analysis of complex biological samples

  • Sandwich ELISA formats to evaluate capture and detection antibody specificity

  • Specific testing for cross-reactivity with variants like Abeta [1-37], Abeta [1-38], Abeta [1-40], or Abeta [1-43]

Tissue cross-reactivity studies:

  • Immunohistochemistry on tissues with varying albumin expression

  • Inclusion of tissues known to express potential cross-reactive proteins

  • Parallel staining with different anti-albumin antibodies for comparison

Competition assays:

  • Pre-incubation of antibody with purified albumin to demonstrate specific inhibition

  • Lack of inhibition by non-target proteins indicates specificity

  • Partial inhibition by related proteins can quantify cross-reactivity

Mass spectrometry verification:

  • Immunoprecipitation followed by mass spectrometry to identify all captured proteins

  • Provides unbiased assessment of potential cross-reactivity

Surface plasmon resonance (SPR) analysis:

  • Quantitative binding analysis against albumin and potential cross-reactive proteins

  • Determination of binding kinetics and affinity constants

  • Comparison of affinity profiles between target and non-target proteins

By implementing these approaches, researchers can thoroughly characterize the cross-reactivity profile of ALB recombinant monoclonal antibodies, ensuring appropriate interpretation of experimental results .

What are recommended storage and handling protocols for maintaining ALB Recombinant Monoclonal Antibody integrity?

Proper storage and handling are essential for maintaining the integrity and functionality of ALB recombinant monoclonal antibodies. Following these protocols can minimize degradation and maximize shelf life:

Storage temperature considerations:

  • Long-term storage: Typically at -20°C to -80°C in small aliquots to minimize freeze-thaw cycles

  • Working stocks: 2-8°C for short-term use (1-2 weeks)

  • Avoid repeated freeze-thaw cycles which can lead to protein denaturation and aggregation

  • Monitor storage unit temperature regularly to ensure consistency

Buffer composition optimization:

  • Stabilizing buffers typically contain:

    • Buffering agent (e.g., phosphate, Tris) at appropriate pH (usually 7.2-7.6)

    • Stabilizing proteins (e.g., BSA, gelatin) at 0.1-1%

    • Cryoprotectants (e.g., glycerol) at 30-50% for frozen storage

    • Preservatives (e.g., sodium azide) at 0.02-0.05% for solutions stored at 2-8°C

  • Avoid buffer components that may interfere with downstream applications

Aliquoting strategy:

  • Prepare small single-use aliquots upon receipt

  • Calculate volume based on typical experimental needs

  • Use sterile techniques and containers

  • Label comprehensively with antibody details, concentration, date, and storage conditions

Thawing and handling procedures:

  • Thaw frozen aliquots quickly at room temperature or 37°C

  • Mix gently by inversion or gentle pipetting to ensure homogeneity

  • Avoid vortexing which can cause protein denaturation

  • Centrifuge briefly to collect contents at the bottom of the tube

Contamination prevention:

  • Use sterile technique when handling antibody solutions

  • Use sterile pipette tips and containers

  • Include preservatives for solutions stored at 2-8°C

  • Avoid repeated entry into stock solutions

Transportation guidelines:

  • Transport on ice or with cold packs for short periods

  • Use dry ice for longer transportation times

  • Validate protein stability after transportation with functional assays

Quality control monitoring:

  • Periodically test antibody function after storage

  • Check for visible signs of degradation (precipitation, cloudiness)

  • Consider implementing stability testing programs for critical antibodies

By following these storage and handling protocols, researchers can maintain the integrity and performance of ALB recombinant monoclonal antibodies throughout their experimental timeline .

How can researchers optimize immunoassays using ALB Recombinant Monoclonal Antibodies?

Optimizing immunoassays with ALB recombinant monoclonal antibodies requires systematic refinement of multiple parameters to achieve maximum sensitivity, specificity, and reproducibility:

Antibody concentration optimization:

  • Perform titration experiments to determine optimal working concentration

  • Balance signal-to-noise ratio with reagent conservation

  • For ALB antibodies, consider testing various concentrations as cross-reactivity may be concentration-dependent

Blocking strategy refinement:

  • Select blockers that don't contain albumin when working with anti-albumin antibodies

  • Test multiple blocking agents (e.g., casein, non-fat milk, synthetic blockers)

  • Optimize blocking time and temperature for background reduction

Sample preparation considerations:

  • Evaluate different sample dilution buffers for optimal signal

  • Consider pre-treatment steps to enhance epitope accessibility

  • For albumin detection, be aware of endogenous albumin in biological samples that might interfere

Assay format selection:

  • Compare direct, indirect, sandwich, and competitive formats

  • For albumin quantification, sandwich ELISAs often provide the best sensitivity and specificity

  • Consider using capture and detection antibodies recognizing different epitopes

Incubation parameters optimization:

  • Test various incubation times and temperatures

  • Evaluate static versus shaking incubation

  • Determine optimal washing procedures (buffer composition, number of washes)

Signal development refinement:

  • Compare different detection systems (colorimetric, fluorescent, chemiluminescent)

  • Optimize substrate concentration and development time

  • Consider signal amplification methods for detecting low-abundance albumin

Validation with reference methods:

  • Compare results with established albumin quantification methods

  • Include standard reference materials when available

  • Perform spike-recovery experiments to assess accuracy

Multiplex considerations:

  • When developing multiplex assays, test for antibody cross-reactivity and interference

  • Optimize signal balance across all analytes

  • Validate each analyte individually before combining

Data analysis optimization:

  • Select appropriate standard curve models

  • Determine assay working range, limit of detection, and limit of quantification

  • Implement quality control measures for assay monitoring

By systematically optimizing these parameters, researchers can develop robust immunoassays for albumin detection and quantification using ALB recombinant monoclonal antibodies .

What approaches can resolve contradictory results when using ALB Recombinant Monoclonal Antibodies?

When researchers encounter contradictory results using ALB recombinant monoclonal antibodies, a structured troubleshooting approach can help identify and resolve the underlying issues:

Antibody characterization verification:

  • Re-validate antibody specificity using orthogonal methods

  • Confirm that the antibody recognizes the intended epitope

  • Verify antibody functionality with positive controls

  • Check for lot-to-lot variability if using different antibody batches

Methodological comparison:

  • Apply multiple detection methods (e.g., ELISA, Western blot, immunohistochemistry)

  • Compare results across different assay formats

  • Consider that each method may access different epitopes due to varying protein conformations

  • For ALB antibodies, be aware that detection sensitivity might vary across applications

Sample preparation analysis:

  • Evaluate how different sample preparation methods affect results

  • Consider that albumin processing (from preproalbumin to secreted albumin) may impact detection

  • Assess whether sample components might interfere with antibody binding

  • Test alternative extraction or preparation protocols

Experimental condition standardization:

  • Standardize key experimental variables across experiments

  • Control for temperature, pH, buffer composition, and incubation times

  • Implement detailed standard operating procedures

  • Use the same reagent lots when possible for comparative studies

Biological variability assessment:

  • Consider inherent biological variability in albumin expression

  • Evaluate physiological factors that might affect results

  • Increase sample sizes to account for biological variation

  • Stratify samples based on relevant biological parameters

Technical validation with complementary approaches:

  • Use mass spectrometry to confirm protein identity and quantity

  • Apply genetic approaches (e.g., qPCR, RNA-seq) to correlate protein and transcript levels

  • Consider genetic knockdown/knockout validation where feasible

  • Implement isotope-labeled internal standards for absolute quantification

Collaborative cross-validation:

  • Engage with other laboratories to independently verify results

  • Share detailed protocols to identify potential methodological differences

  • Consider inter-laboratory studies with standardized samples

  • Compare results with published literature on albumin detection

Epitope accessibility investigation:

  • Examine whether contradictory results might stem from differential epitope accessibility

  • Test multiple anti-albumin antibodies recognizing different epitopes

  • Consider native versus denatured detection methods

  • Evaluate potential post-translational modifications affecting epitope recognition

By systematically applying these approaches, researchers can identify sources of contradictory results and develop more robust experimental protocols for albumin detection and quantification .

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 2024 Thebiotek. All Rights Reserved.