GSN Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Biology

GSN Recombinant Monoclonal Antibody is a genetically engineered antibody designed to detect and bind human, mouse, and rat gelsolin (GSN), a 80–95 kDa protein involved in cytoskeletal dynamics, cell motility, and apoptosis . GSN severs actin filaments and caps barbed ends, playing roles in inflammation, cancer metastasis, and amyloidosis .

Production Methodology

The antibody is generated through a multi-step recombinant process:

StepDetails
Gene CloningHeavy/light chain genes cloned into expression vectors (e.g., plasmids) .
Host Cell TransfectionVectors transfected into HEK293 human embryonic kidney cells .
Expression & PurificationAntibody expressed in vitro and purified via affinity chromatography .

Key advantages over traditional methods include elimination of hybridoma cell-line drift and batch-to-batch variability .

3.1. Primary Applications

  • Western Blot (WB): Detects GSN at 0.5 µg/mL in human SK-Mel-28, mouse MEF, and rat NR8383 lysates .

  • Immunohistochemistry (IHC): Localizes GSN in human kidney glomeruli and distal tubules at 8–25 µg/mL .

  • Flow Cytometry (FC): Effective at 1:50–1:200 dilution .

3.2. Validation Data

Validation MethodResults
Knockout SpecificityNo signal in GSN-knockout U2OS cells; band observed at ~80 kDa in wild-type lysates .
Species Cross-ReactivityConfirmed for human, mouse, and rat tissues .
ELISA SpecificityBinds recombinant human GSN (UniProt ID: P06396) .

4.1. Cancer Research

  • Detects GSN overexpression in renal cell carcinoma and melanoma cell lines (SK-Mel-28) .

  • Used in seqIF™ staining to analyze tumor microenvironments .

4.3. Functional Studies

  • Validated for actin-binding studies in MEF cells .

  • Neutralization assays confirm no cross-reactivity with unrelated cytoskeletal proteins .

Advantages Over Conventional Antibodies

  • Reproducibility: Defined genetic sequence eliminates lot-to-lot variability .

  • Scalability: High-yield production in HEK293 cells .

  • Ethical Compliance: Reduces animal use compared to hybridoma-based methods .

Product Specs

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

The production of the GSN recombinant monoclonal antibody involves a meticulous multi-step process. It begins with in vitro cloning, where the genes encoding both the heavy and light chains of the GSN antibody are integrated into expression vectors. These vectors are then transfected into host cells, enabling the expression of the recombinant antibody within a cell culture environment. Following expression, the GSN recombinant monoclonal antibody is rigorously purified from the supernatant of transfected host cell lines using an affinity-chromatography purification method. This antibody exhibits specific reactivity with the human GSN protein and demonstrates versatility in three key applications: ELISA, IF, and FC.

Gelsolin (GSN) is a multifaceted protein that plays a pivotal role in regulating the dynamics of the actin cytoskeleton. Its interactions with actin filaments are essential for controlling cell shape, motility, and a variety of cellular processes, impacting both normal physiology and pathological conditions.

Form
Liquid
Lead Time
Typically, we are able to dispatch products within 1-3 business days after receiving your orders. Delivery time may vary depending on the chosen purchasing method or location. For precise delivery information, please contact your local distributors.
Synonyms
Gelsolin (AGEL) (Actin-depolymerizing factor) (ADF) (Brevin), GSN
Target Names
GSN
Uniprot No.

Target Background

Function

Gelsolin is a calcium-regulated, actin-modulating protein that binds to the plus (or barbed) ends of actin monomers or filaments, preventing monomer exchange (end-blocking or capping). It can facilitate the assembly of monomers into filaments (nucleation) as well as sever already formed filaments. Gelsolin also plays a role in ciliogenesis.

Gene References Into Functions
  1. The gelsolin N184K variant undergoes the same aberrant proteolytic events that lead to the formation of amyloidogenic fragments in previously characterized mutants. PMID: 27633054
  2. Induction of cell stress alters the actin cytoskeleton in intestinal epithelial cells through changes in the actin-binding proteins villin-1 and gelsolin. PMID: 29274870
  3. Upregulation of gelsolin promotes radioresistance in non-small cell lung cancer cells, at least partially, through activation of phosphoinositide 3-kinase/Akt signaling. PMID: 27121073
  4. Research identifies GSN as a target of miR-200a, which inhibits the release of microvesicles in hepatocellular carcinoma cells by functionally regulating GSN. PMID: 28440466
  5. Gelsolin enhances the invasive capacity of colon cancer cells by elevating intracellular superoxide (O2.-) levels through interaction with Cu/ZnSOD. Moreover, gelsolin gene expression positively correlates with urokinase plasminogen activator (uPA), a crucial matrix-degrading protease involved in cancer invasion. PMID: 27391159
  6. These data highlight the significance of the physical interaction between HPV16 E7 and gelsolin in the acquisition of the metastatic phenotype by cervical cancer cells. PMID: 27072581
  7. This study underscores gelsolin as a crucial pro-disseminative factor contributing to the aggressive phenotype of diffuse gastric cancer. PMID: 27058427
  8. The study strongly supports the contribution of the genes ITGA2B, GSN, and RHOA, and the two pathways “regulation of actin cytoskeleton” and “leukocyte transendothelial migration” to osteoporosis risk. PMID: 27153759
  9. SSH1 binds to gelsolin via actin filaments in the cytosolic fraction. Gelsolin promotes solubilization of actin filaments and SSH1 in cell-free assays and in cultured cells. PMID: 25451266
  10. Free actin likely contributes to impaired host defense by blocking scavenger receptor binding of bacteria. PMID: 28385809
  11. This analysis elucidates how YopO disrupts normal gelsolin function to alter host actin dynamics, thereby hindering phagocytosis. PMID: 28280241
  12. The present study suggests that the synergistic effect of pGSN and pIgA induces glomerular fibrosis through the TGF-beta1/Smads signal transduction pathway. This might be a potential mechanism underlying the glomerular fibrosis observed in IgAN patients. PMID: 28208683
  13. GSN expression levels were observed to be reduced in colon carcinoma (CC) cells, and the decreased expression level of GSN is often associated with a poorer metastasis-free survival rate in patients with CC (P=0.04). Additionally, overexpression of GSN inhibited the invasion of CC cells in vitro. PMID: 27573444
  14. In chronic hemodialysis patients, lower pGSN levels were not associated with hospitalization, all-cause, and cardiovascular mortality, even though pGSN was inversely correlated with age, CRP, and IL-6, suggesting that inflammation may influence pGSN. PMID: 28114138
  15. PODXL enhances motility and invasiveness through an increase in gelsolin-actin interactions in cell protrusions. PMID: 27461278
  16. These findings suggest that gelsolin may play a role in the disease process in atopic dermatitis patients. PMID: 26318415
  17. Plasma GSN may play a significant role in the development of immunoglobulin A (IgA) nephropathy. PMID: 27997897
  18. Beta-catenin appears to be involved in the regulation of gelsolin expression, which in turn affects the migratory ability of colonic cancer cells. PMID: 27798885
  19. Hypoxia or GSN overexpression induces HIF-1alpha expression and reduces the expression of survival markers p-Akt and Bcl-2 in H9c2 cardiomyoblast cells. PMID: 27193608
  20. Increasing amounts of amyloid are associated with the severity of clinical features in hereditary gelsolin amyloidosis. PMID: 27879149
  21. The rs1078305 and rs10818524 SNPs of GSN were associated with an increased risk for oral squamous cell carcinoma development in a Chinese Han population. PMID: 26848502
  22. Lower blood levels of gelsolin were associated with progressive aortic arch calcification. PMID: 26941566
  23. Gelsolin protein, human, yielded a receiver operating characteristics value of 89% for node-positive OSCC. PMID: 25223295
  24. GSN overexpression suppresses apoptosis, while downregulated GSN promotes apoptosis in hepatocellular carcinoma. PMID: 26823700
  25. Fragmented gelsolins may be associated with the pathogenesis of fibrosis in RA-ILD. PMID: 26666486
  26. Gelsolin interacts with specific components of the three cytoskeleton systems and is a constituent of midbodies. PMID: 26598132
  27. Novel roles for actin-depolymerizing factor and cofilin-1 in regulating the remodeling and permeability of epithelial junctions. PMID: 26878213
  28. The overexpression of GSN may inhibit the proliferation, adhesion ability, and invasion of the 786-0 clear cell renal cell carcinoma cell line. PMID: 26398833
  29. Serum-expressed apolipoprotein B-100 protein, C9 Complement, and gelsolin can be used for the differential diagnosis of Barrett’s esophagus and adenocarcinoma of the esophagus. PMID: 26404905
  30. TGF-beta1 induced epigenetic modification of GSN could alter the EMT process in breast cancer cells. PMID: 26482896
  31. GSNs might play a regulatory role in the suppression of tissue damage induced by acute radiation exposure. PMID: 25164111
  32. We designed and synthesized siRNA against various exons in the gelsolin gene (GSN). Our findings reveal that siRNA, derived from a target gene exon, can form a complex with H3 histone to be involved in the regulation of gene expression. PMID: 25600697
  33. Results indicate that high gelsolin levels are associated with a better prognosis in ER+HER2- breast cancer and a reduction in tumor cell migration. PMID: 26408687
  34. Gelsolin expression promoted tumor-associated phenotypes by facilitating proliferative and invasive capacities of hepatocellular carcinoma (HCC) cells, which might serve as a potential therapeutic target for HCC treatment. PMID: 26149653
  35. Based on the obtained results, we propose that gelsolin is a crucial cellular target for cotinine, through which this compound influences the basic processes involved in neoplastic transformation and metastasis, such as migration and apoptosis. PMID: 25544037
  36. Group B Streptococcus-beta-haemolysin is solely responsible for the increase in gelsolin, causing, through membrane permeability defects, calcium influx, and calpain activation. PMID: 25130983
  37. Plasma gelsolin levels decrease in the blood of type II diabetics. Recombinant gelsolin helped improve glycemic control in diabetic mice. PMID: 25478578
  38. Data suggest that gelsolin plays a vital role in the regulation of gynecological cell fate, as reflected in dysregulation in chemosensitivity. PMID: 25246592
  39. Data indicate that gelsolin contributes to the invasive potential of LNCaP prostate adenocarcinoma cells. PMID: 25581609
  40. Gelsolin levels were a valuable tool for predicting functional outcome and mortality after aneurysmal subarachnoid hemorrhage. PMID: 23880145
  41. Increased plasma levels after allergen-specific immunotherapy. PMID: 24980225
  42. Patients developing cardiopulmonary bypass acute lung injury had a lower plasma gelsolin reservoir and a much more substantial and rapid consumption of plasma gelsolin early after operation. PMID: 25126004
  43. This study enhances the understanding of the genetic features of Mexican patients with corneal stromal dystrophies by identifying mutations in the TGFBI, CHST6, and GSN genes. PMID: 24801599
  44. GSN is crucial for chemoresistance in head-and-neck cancer. PMID: 24771612
  45. We predict that gene silencing of GSN and/or the downstream blocking of GSN along the p38 pathway could be applied to ameliorate pathological cardiac hypertrophy in the future. PMID: 24505034
  46. Ceruloplasmin and gelsolin are closely interacted with the oncogene NF-kappab. PMID: 23925487
  47. Plasma gelsolin is lower in normal pregnancy than in non-pregnant women. It is also significantly lower in pre-eclampsia than in normal pregnancy. PMID: 24239294
  48. This study found significantly lower plasma gelsolin levels in patients with systemic lupus erythematosus and rheumatoid arthritis compared with healthy controls. PMID: 24122723
  49. Expression of a mutant form of human gelsolin in mice under the control of a muscle-specific promoter induced myopathic changes reminiscent of human inclusion body myositis. PMID: 24047347
  50. Gelsolin can constitute a barrier that restricts HIV-1 infection of CD4+ lymphocytes in a pre-fusion step. PMID: 23575248

Show More

Hide All

Database Links

HGNC: 4620

OMIM: 105120

KEGG: hsa:2934

STRING: 9606.ENSP00000362924

UniGene: Hs.522373

Involvement In Disease
Amyloidosis 5 (AMYL5)
Protein Families
Villin/gelsolin family
Subcellular Location
[Isoform 2]: Cytoplasm, cytoskeleton.; [Isoform 1]: Secreted.
Tissue Specificity
Phagocytic cells, platelets, fibroblasts, nonmuscle cells, smooth and skeletal muscle cells.

Q&A

What are recombinant monoclonal antibodies and how do they differ from traditional monoclonal antibodies?

Recombinant monoclonal antibodies (rMAbs) are engineered antibodies generated in vitro using synthetic genes typically expressed from plasmids or other expression vectors, without requiring hybridoma cell lines. Unlike traditional monoclonal antibodies produced via hybridoma technology, recombinant antibodies are created through genetic engineering techniques that involve cloning antibody genes into expression vectors . This fundamental difference enables greater control over antibody properties, improved reproducibility, and reduced dependence on animal immunization. Recombinant antibodies maintain the same specificity characteristics as traditional monoclonal antibodies but offer enhanced customization options for research applications .

What are the key advantages of using recombinant monoclonal antibodies in research?

Recombinant monoclonal antibodies offer several significant advantages for research applications:

  • Improved reproducibility: Since rMAbs are produced from defined genetic sequences, they provide batch-to-batch consistency that addresses standardization issues common with traditional antibodies .

  • Ethical considerations: The reduced reliance on animals for antibody production addresses ethical concerns regarding large-scale animal use in traditional antibody generation .

  • Sequence transparency: Having access to the complete genetic sequence enables further engineering and modification for specialized applications .

  • Customization potential: Researchers can readily modify antibody characteristics including species specificity, format (full-length vs. fragments), and functional properties .

  • Compatibility with in vitro selection methods: rMAbs can be selected against targets that may not work well in animals due to host protein similarities .

These advantages make rMAbs increasingly preferred for applications requiring highly reproducible and customizable antibody reagents.

What are the main approaches for producing recombinant monoclonal antibodies?

The production of recombinant monoclonal antibodies can follow several methodological paths:

From known sequences:

  • Design DNA geneblocks optimized for expression in the intended host cells using codon optimization tools .

  • Clone heavy and light chain variable domain sequences into appropriate expression vectors using methods like Gibson assembly .

  • Co-express heavy and light chain plasmids in suitable expression systems (commonly HEK293 suspension culture cells) .

From existing hybridomas:

  • Extract and sequence the DNA from hybridoma cell lines to determine antibody chain sequences .

  • Clone these sequences into expression vectors for recombinant production .

From protein samples:

  • Use mass spectrometry to determine amino acid sequences from purified antibodies (such as from patient blood) .

  • Synthesize genes encoding these amino acid sequences and test resulting antibodies for antigen binding .

De novo selection:

  • Perform in vitro selection from antibody libraries using display technologies (phage, yeast, ribosome, or mammalian display) .

  • Clone selected antibody genes into expression vectors for production .

Each approach has distinct advantages depending on the starting material and specific research requirements.

What is the transcriptionally active PCR (TAP) method for rapid antibody generation, and what are its advantages?

The transcriptionally active PCR (TAP) method is a rapid approach for generating functional recombinant antibodies without traditional cloning procedures. This methodology involves:

  • Isolating single antigen-specific antibody secreting cells (ASCs) from peripheral blood

  • Using RT-PCR to generate linear Ig heavy and light chain gene expression cassettes (called "minigenes")

  • Directly expressing these minigenes as recombinant antibodies through transient transfection

Key advantages of this approach include:

  • Time efficiency: Enables identification and expression of antigen-specific monoclonal antibodies in less than 10 days .

  • Resource conservation: Eliminates the need for time-consuming in vitro differentiation of memory B cells .

  • Functional screening: Allows researchers to screen individual ASCs for effector function prior to recombinant antibody cloning, enabling selection of antibodies with desired characteristics .

  • Comprehensive analysis: Enables analysis of variable region repertoires in combination with functional assays to evaluate specificity and function .

This approach has been successfully demonstrated with COVID-19 convalescent patients, where it enabled rapid identification and production of neutralizing antibodies against SARS-CoV-2 variants .

How can species specificity of recombinant monoclonal antibodies be customized for research applications?

Species specificity customization of recombinant monoclonal antibodies involves precise genetic engineering techniques:

  • Variable region preservation: Design and synthesize geneblocks corresponding only to the variable regions of the heavy and light chains, preserving the antigen recognition portions .

  • Constant region substitution: Generate PCR fragments corresponding to the constant regions from the target species for both heavy and light chains .

  • Recombinant assembly: Combine the variable region geneblocks with the species-specific constant region PCR fragments using Gibson assembly or similar methods .

  • Expression vector construction: Clone the assembled fragments into appropriate expression vectors (such as the rMAbParent plasmid) .

  • Co-expression: Transfect the heavy and light chain constructs into an expression system like HEK293 cells for antibody production .

This approach enables researchers to generate antibodies with the same antigen specificity but with constant regions from different species (human, mouse, rabbit, etc.), which is particularly valuable for applications where the host species of the experiment influences antibody performance or when avoiding cross-reactivity with endogenous antibodies is necessary .

What strategies exist for converting antibody formats between full-length, scFv, and other fragment variants?

Several methodological strategies can be employed to convert between different antibody formats:

From full-length to scFv (single-chain variable fragment):

  • PCR-amplify the variable regions of both heavy and light chains

  • Connect these regions using a flexible linker sequence (typically (Gly₄Ser)₃)

  • Clone the assembled construct into an expression vector

From scFv to full-length bivalent antibody:

  • Extract the variable regions from the scFv construct

  • Clone these regions into separate heavy and light chain expression vectors containing the appropriate constant regions

  • Co-express both vectors in a suitable mammalian expression system

Creating scFv-Fc fusion (scFvC):

  • Construct a fusion protein containing:

    • Heavy chain variable region

    • Flexible linker

    • Light chain variable region

    • Selected constant regions (typically CH2 and CH3 domains)

  • Clone into a single expression vector for production

These conversion strategies provide researchers with flexible options for different experimental needs, balancing factors such as tissue penetration, avidity, stability, and effector functions. For example, smaller fragments like scFvs offer better tissue penetration, while full-length antibodies provide increased stability and effector functions .

Antibody FormatApproximate SizeKey AdvantagesTypical Applications
Full-length IgG150 kDaStability, effector functions, bivalent bindingMost research applications, therapeutics
scFv25-30 kDaSmall size, tissue penetration, genetic encodingImaging, intracellular targeting
scFv-Fc (scFvC)55-60 kDaCombines scFv targeting with Fc functionsApplications requiring both targeting and effector functions
Fab50 kDaMonovalent binding, reduced immunogenicityWhen Fc functions are undesirable

How can recombinant antibody technology accelerate response to emerging infectious diseases?

Recombinant antibody technology offers a powerful methodology for rapidly responding to emerging infectious diseases through multiple mechanisms:

  • Rapid isolation and characterization pipeline:

    • Direct isolation of antibody-secreting cells (ASCs) from convalescent patients

    • Functional screening of antibody specificity and neutralizing capacity

    • Generation of recombinant antibodies in under 10 days using transcriptionally active PCR (TAP) methods

  • Parallel analysis of antibody repertoires:

    • Sequencing of variable region repertoires from infected/recovered individuals

    • Correlation of sequence characteristics with neutralizing capabilities

    • Identification of broadly neutralizing antibodies effective against multiple variants

  • Functional screening before full production:

    • Testing antibody supernatants from single ASCs for binding and neutralization

    • Selection of only the most promising candidates for full recombinant production

    • Prioritization based on desired functional characteristics

As demonstrated in the COVID-19 pandemic, this approach enabled researchers to rapidly identify and characterize neutralizing antibodies against SARS-CoV-2. From the panel of 36 spike-specific monoclonal antibodies generated from convalescent patients, researchers identified antibodies capable of neutralizing both the Wuhan and Delta variants, though not the later-emerging Omicron variant . This demonstrates how recombinant antibody technology can provide rapid therapeutic candidates, diagnostic tools, and mechanistic insights during outbreaks.

What methodological approaches enable high-throughput sequencing and cataloging of hybridoma-derived antibodies?

High-throughput sequencing and cataloging of hybridoma-derived antibodies involves several sophisticated methodological approaches:

  • Direct hybridoma sequencing:

    • RNA extraction from hybridoma cells

    • Targeted amplification of immunoglobulin heavy and light chain variable domains

    • Next-generation sequencing to determine antibody sequences

  • Database development and management:

    • Creation of searchable DNA sequence databases with appropriate metadata

    • Integration of sequence data with functional characterization

    • Public availability of sequence information through dedicated platforms

  • Recombinant conversion validation:

    • Production of recombinant versions of hybridoma-derived antibodies

    • Functional comparison between original hybridoma antibodies and recombinant versions

    • Validation of epitope recognition and binding characteristics

The NeuroMabSeq initiative exemplifies this approach, having successfully sequenced a large collection of hybridoma-derived monoclonal antibodies validated for neuroscience research. This initiative created a publicly accessible database (neuromabseq.ucdavis.edu) for sharing, analysis, and downstream applications . This methodological framework enhances reproducibility and enables subsequent engineering of antibodies into alternate forms with distinct utility, including different detection modes for multiplexed labeling and miniaturized formats like scFvs .

What are common challenges in recombinant antibody expression and how can they be addressed methodologically?

Researchers frequently encounter several challenges when expressing recombinant antibodies that can be systematically addressed through targeted optimization strategies:

Challenge: Low expression yields

  • Methodological solution: Implement codon optimization for the expression host using computational tools to design DNA geneblocks with preferred codon usage .

  • Methodological solution: Add an appropriate signal peptide sequence to the N-terminus of antibody chains to enhance secretion efficiency .

  • Methodological solution: Optimize transfection conditions including DNA:transfection reagent ratio, cell density, and harvest timing for maximum yield.

Challenge: Misfolding or aggregation

  • Methodological solution: Modify culture temperature (typically lowering to 30-32°C) during expression to slow protein synthesis and improve folding.

  • Methodological solution: Incorporate stabilizing mutations identified through computational prediction or directed evolution approaches.

  • Methodological solution: For scFv constructs, optimize the length and composition of the linker between variable domains to improve proper folding .

Challenge: Chain pairing issues

  • Methodological solution: Implement knobs-into-holes mutations in the CH3 domain for difficult-to-express antibodies.

  • Methodological solution: Consider single-chain formats or creation of bispecific constructs using controlled assembly strategies.

Challenge: Loss of binding affinity

  • Methodological solution: Verify sequence integrity through complete sequencing before and after cloning.

  • Methodological solution: Perform affinity maturation through directed evolution if necessary to recover or enhance binding properties.

How can researchers evaluate and ensure the functional equivalence between original hybridoma antibodies and their recombinant versions?

Ensuring functional equivalence between hybridoma-derived antibodies and their recombinant versions requires systematic comparative analysis:

  • Binding characteristics comparison:

    • Perform side-by-side ELISA assays with serial dilutions of both antibody sources

    • Compare EC50 values and binding curves to detect any affinity differences

    • Use surface plasmon resonance (SPR) to quantitatively measure binding kinetics (kon and koff rates)

  • Epitope mapping validation:

    • Conduct competitive binding assays between hybridoma and recombinant antibodies

    • Perform epitope binning experiments to confirm identical epitope recognition

    • Use peptide arrays or alanine scanning mutagenesis to precisely map epitope boundaries

  • Application-specific functional testing:

    • Validate performance in the intended application contexts (immunohistochemistry, Western blot, immunoprecipitation, etc.)

    • Compare signal-to-noise ratios and specificity profiles across different sample types

    • Assess performance across different experimental conditions (fixatives, buffers, detergents)

  • Physicochemical characterization:

    • Analyze antibody stability using differential scanning fluorimetry or circular dichroism

    • Assess aggregation propensity using size exclusion chromatography

    • Confirm glycosylation patterns if relevant to function

These methodological approaches ensure that recombinant antibodies maintain the valuable properties of the original hybridoma-derived antibodies while gaining the advantages of recombinant production systems .

How might open-source sharing of antibody sequences and expression plasmids accelerate research innovation?

Open-source sharing of antibody sequences and expression plasmids offers transformative potential for accelerating research through several mechanisms:

  • Improved reproducibility across laboratories:

    • Researchers can access identical antibody reagents with defined sequences

    • Elimination of batch-to-batch variability issues that plague commercial antibodies

    • Enhanced ability to reproduce and build upon published findings

  • Democratized antibody engineering:

    • Similar to how the CRISPR community created hundreds of useful Cas9 variants through plasmid sharing

    • Researchers worldwide could generate higher affinity antibodies, modify antibody function, improve stability

    • Development of novel tools and applications that haven't been imagined yet

  • Reduced research costs and barriers:

    • Elimination of repeated antibody development against the same targets

    • Reduced dependence on expensive commercial antibodies

    • Lower barriers to entry for labs with limited resources

  • Accelerated iterative improvement:

    • Collective intelligence applied to antibody optimization

    • Rapid sharing of modifications that enhance performance

    • Community-driven validation and characterization

The current practice of keeping antibody sequences proprietary significantly hampers scientific progress. Open plasmid sharing would empower scientists to rapidly build upon existing tools rather than constantly "reinventing the wheel," allowing more research energy to be directed toward novel discoveries rather than reagent generation .

What emerging technologies are poised to further revolutionize recombinant antibody development and engineering?

Several cutting-edge technologies are positioned to transform recombinant antibody development:

  • AI-driven antibody design:

    • Machine learning algorithms for predicting optimal antibody sequences based on epitope structure

    • Computational tools for enhancing stability, solubility, and manufacturability

    • In silico maturation of antibody affinity and specificity

  • High-throughput single B-cell technologies:

    • Microfluidic platforms for isolating and analyzing thousands of individual B cells

    • Direct linkage of phenotypic screening with genotypic information

    • Rapid identification of rare antibodies with desired properties

  • Cell-free expression systems:

    • Rapid production of antibodies without cell culture

    • High-throughput parallel screening of multiple antibody candidates

    • Reduced time from sequence to functional protein

  • Synthetic antibody libraries with expanded chemical diversity:

    • Incorporation of non-canonical amino acids for enhanced function

    • Chemically diversified antibody libraries beyond natural amino acid constraints

    • Novel binding properties and catalytic activities

  • Genome editing for optimized expression hosts:

    • CRISPR-engineered cell lines with enhanced protein folding and secretion capacity

    • Glycoengineered expression hosts for optimized antibody effector functions

    • Removal of problematic proteases and modification of chaperone expression

These technologies, combined with standardized production methods and open sharing of resources, are poised to dramatically accelerate the pace of antibody engineering and expand the repertoire of antibody-based research tools available to the scientific community.

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.