Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 13318, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770098

ABSTRACT

Immunoassays based on sandwich immuno-complexes of capture and detection antibodies simultaneously binding to the target analytes have been powerful technologies in molecular analyses. Recent developments in single molecule detection technologies enable the detection limit of the sandwich immunoassays approaching femtomolar (10-15 M), driving the needs of developing sensitive and specific antibodies for ever-increasingly broad applications in detecting and quantifying biomarkers. The key components underlying the sandwich immunoassays are antibody-based affinity reagents, for which the conventional sources are mono- or poly-clonal antibodies from immunized animals. The downsides of the animal-based antibodies as affinity reagents arise from the requirement of months of development timespan and limited choices of antibody candidates due to immunodominance of humoral immune responses in animals. Hence, developing animal antibodies capable of distinguishing highly related antigens could be challenging. To overcome the limitation imposed by the animal immune systems, we developed an in vitro methodology based on phage-displayed synthetic antibody libraries for diverse antibodies as affinity reagents against closely related influenza virus nucleoprotein (NP) subtypes, aiming to differentiating avian influenza virus (H5N1) from seasonal influenza viruses (H1N1 and H3N2), for which the NPs are closely related by 90-94% in terms of pairwise amino acid sequence identity. We applied the methodology to attain, within four weeks, a panel of IgGs with distinguishable specificities against a group of representative NPs with pairwise amino acid sequence identities up to more than 90%, and the antibodies derived from the antibody libraries without further affinity refinement had comparable affinity of mouse antibodies to the NPs with the detection limit less than 1 nM of viral NP from lysed virus with sandwich ELISA. The panel of IgGs were capable of rapidly distinguishing infections due to virulent avian influenza virus from infections of seasonal flu, in responding to a probable emergency scenario where avian influenza virus would be transmissible among humans overlapping with the seasonal influenza infections. The results indicate that the in vitro antibody development methodology enables developing diagnostic antibodies that would not otherwise be available from animal-based antibody technologies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Influenza A virus/immunology , Peptide Library , Viral Core Proteins/immunology , Animals , Dogs , Enzyme-Linked Immunosorbent Assay , Humans , Influenza, Human/diagnosis , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Mice
2.
MAbs ; 11(2): 373-387, 2019.
Article in English | MEDLINE | ID: mdl-30526270

ABSTRACT

Antibodies provide immune protection by recognizing antigens of diverse chemical properties, but elucidating the amino acid sequence-function relationships underlying the specificity and affinity of antibody-antigen interactions remains challenging. We designed and constructed phage-displayed synthetic antibody libraries with enriched protein antigen-recognition propensities calculated with machine learning predictors, which indicated that the designed single-chain variable fragment variants were encoded with enhanced distributions of complementarity-determining region (CDR) hot spot residues with high protein antigen recognition propensities in comparison with those in the human antibody germline sequences. Antibodies derived directly from the synthetic antibody libraries, without affinity maturation cycles comparable to those in in vivo immune systems, bound to the corresponding protein antigen through diverse conformational or linear epitopes with specificity and affinity comparable to those of the affinity-matured antibodies from in vivo immune systems. The results indicated that more densely populated CDR hot spot residues were sustainable by the antibody structural frameworks and could be accompanied by enhanced functionalities in recognizing protein antigens. Our study results suggest that synthetic antibody libraries, which are not limited by the sequences found in antibodies in nature, could be designed with the guidance of the computational machine learning algorithms that are programmed to predict interaction propensities to molecules of diverse chemical properties, leading to antibodies with optimal characteristics pertinent to their medical applications.


Subject(s)
Machine Learning , Protein Engineering/methods , Single-Chain Antibodies/chemistry , Antibody Affinity , Antibody Specificity , Humans , Peptide Library , Structure-Activity Relationship
3.
MAbs ; 11(1): 153-165, 2019 01.
Article in English | MEDLINE | ID: mdl-30365359

ABSTRACT

HER2-ECD (human epidermal growth factor receptor 2 - extracellular domain) is a prominent therapeutic target validated for treating HER2-positive breast and gastric cancer, but HER2-specific therapeutic options for treating advanced gastric cancer remain limited. We have developed antibody-drug conjugates (ADCs), comprising IgG1 linked via valine-citrulline to monomethyl auristatin E, with potential to treat HER2-positive gastric cancer in humans. The antibodies optimally selected from the ADC discovery platform, which was developed to discover antibody candidates suitable for immunoconjugates from synthetic antibody libraries designed using antibody-antigen interaction principles, were demonstrated to be superior immunoconjugate targeting modules in terms of efficacy and off-target toxicity. In comparison with the two control humanized antibodies (trastuzumab and H32) derived from murine antibody repertoires, the antibodies derived from the synthetic antibody libraries had enhanced receptor-mediated internalization rate, which could result in ADCs with optimal efficacies. Along with the ADCs, two other forms of immunoconjugates (scFv-PE38KDEL and IgG1-AL1-PE38KDEL) were used to test the antibodies for delivering cytotoxic payloads to xenograft tumor models in vivo and to cultured cells in vitro. The in vivo experiments with the three forms of immunoconjugates revealed minimal off-target toxicities of the selected antibodies from the synthetic antibody libraries; the off-target toxicities of the control antibodies could have resulted from the antibodies' propensity to target the liver in the animal models. Our ADC discovery platform and the knowledge gained from our in vivo tests on xenograft models with the three forms of immunoconjugates could be useful to anyone developing optimal ADC cancer therapeutics.


Subject(s)
Aminobenzoates/pharmacology , Immunoconjugates/pharmacology , Molecular Targeted Therapy/methods , Oligopeptides/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/pathology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Humans , Mice , Xenograft Model Antitumor Assays
4.
Sci Rep ; 7(1): 14455, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089574

ABSTRACT

Pandemic and epidemic outbreaks of influenza A virus (IAV) infection pose severe challenges to human society. Passive immunotherapy with recombinant neutralizing antibodies can potentially mitigate the threats of IAV infection. With a high throughput neutralizing antibody discovery platform, we produced artificial anti-hemagglutinin (HA) IAV-neutralizing IgGs from phage-displayed synthetic scFv libraries without necessitating prior memory of antibody-antigen interactions or relying on affinity maturation essential for in vivo immune systems to generate highly specific neutralizing antibodies. At least two thirds of the epitope groups of the artificial anti-HA antibodies resemble those of natural protective anti-HA antibodies, providing alternatives to neutralizing antibodies from natural antibody repertoires. With continuing advancement in designing and constructing synthetic scFv libraries, this technological platform is useful in mitigating not only the threats of IAV pandemics but also those from other newly emerging viral infections.


Subject(s)
Antibodies, Neutralizing/immunology , Orthomyxoviridae/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Viral/immunology , Bacteriophages/immunology , Disease Outbreaks , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , High-Throughput Screening Assays/methods , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A virus/immunology , Influenza, Human/virology , Pandemics , Single-Chain Antibodies/immunology
5.
Sci Rep ; 5: 15053, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456860

ABSTRACT

Broadly neutralizing antibodies developed from the IGHV1-69 germline gene are known to bind to the stem region of hemagglutinin in diverse influenza viruses but the sequence determinants for the antigen recognition, including neutralization potency and binding affinity, are not clearly understood. Such understanding could inform designs of synthetic antibody libraries targeting the stem epitope on hemagglutinin, leading to artificially designed antibodies that are functionally advantageous over antibodies from natural antibody repertoires. In this work, the sequence space of the complementarity determining regions of a broadly neutralizing antibody (F10) targeting the stem epitope on the hemagglutinin of a strain of H1N1 influenza virus was systematically explored; the elucidated antibody-hemagglutinin recognition principles were used to design a phage-displayed antibody library, which was then used to discover neutralizing antibodies against another strain of H1N1 virus. More than 1000 functional antibody candidates were selected from the antibody library and were shown to neutralize the corresponding strain of influenza virus with up to 7 folds higher potency comparing with the parent F10 antibody. The antibody library could be used to discover functionally effective antibodies against other H1N1 influenza viruses, supporting the notion that target-specific antibody libraries can be designed and constructed with systematic sequence-function information.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Epitopes/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Peptide Library , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Cross Reactions , Dogs , Epitope Mapping , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Madin Darby Canine Kidney Cells , Molecular Sequence Data , Neutralization Tests , Protein Binding , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/immunology
6.
Sci Rep ; 5: 12411, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26202883

ABSTRACT

Humoral immunity against diverse pathogens is rapidly elicited from natural antibody repertoires of limited complexity. But the organizing principles underlying the antibody repertoires that facilitate this immunity are not well-understood. We used HER2 as a model immunogen and reverse-engineered murine antibody response through constructing an artificial antibody library encoded with rudimentary sequence and structural characteristics learned from high throughput sequencing of antibody variable domains. Antibodies selected in vitro from the phage-displayed synthetic antibody library bound to the model immunogen with high affinity and specificities, which reproduced the specificities of natural antibody responses. We conclude that natural antibody structural repertoires are shaped to allow functional antibodies to be encoded efficiently, within the complexity limit of an individual antibody repertoire, to bind to diverse protein antigens with high specificity and affinity. Phage-displayed synthetic antibody libraries, in conjunction with high-throughput sequencing, can thus be designed to replicate natural antibody responses and to generate novel antibodies against diverse antigens.


Subject(s)
Antigen-Antibody Reactions/immunology , Immunity, Innate/immunology , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Amino Acid Sequence , Animals , Binding Sites , Humans , Mice , Molecular Sequence Data , Protein Binding , Structure-Activity Relationship
7.
Structure ; 22(1): 22-34, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24268647

ABSTRACT

Protein structural stability and biological functionality are dictated by the formation of intradomain cores and interdomain interfaces, but the intricate sequence-structure-function interrelationships in the packing of protein cores and interfaces remain difficult to elucidate due to the intractability of enumerating all packing possibilities and assessing the consequences of all the variations. In this work, groups of ß strand residues of model antibody variable domains were randomized with saturated mutagenesis and the functional variants were selected for high-throughput sequencing and high-throughput thermal stability measurements. The results show that the sequence preferences of the intradomain hydrophobic core residues are strikingly flexible among hydrophobic residues, implying that these residues are coupled indirectly with antigen binding through energetic stabilization of the protein structures. By contrast, the interdomain interface residues are directly coupled with antigen binding. The interdomain interface should be treated as an integral part of the antigen-binding site.


Subject(s)
Immunoglobulin Variable Region/chemistry , Single-Chain Antibodies/chemistry , Vascular Endothelial Growth Factor A/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , High-Throughput Nucleotide Sequencing , High-Throughput Screening Assays , Humans , Hydrogen Bonding , Immunoglobulin Variable Region/immunology , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Binding , Protein Folding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Single-Chain Antibodies/immunology , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/immunology , Structure-Activity Relationship , Thermodynamics , Vascular Endothelial Growth Factor A/immunology
8.
PLoS One ; 7(3): e33340, 2012.
Article in English | MEDLINE | ID: mdl-22457753

ABSTRACT

Protein-protein interactions are critical determinants in biological systems. Engineered proteins binding to specific areas on protein surfaces could lead to therapeutics or diagnostics for treating diseases in humans. But designing epitope-specific protein-protein interactions with computational atomistic interaction free energy remains a difficult challenge. Here we show that, with the antibody-VEGF (vascular endothelial growth factor) interaction as a model system, the experimentally observed amino acid preferences in the antibody-antigen interface can be rationalized with 3-dimensional distributions of interacting atoms derived from the database of protein structures. Machine learning models established on the rationalization can be generalized to design amino acid preferences in antibody-antigen interfaces, for which the experimental validations are tractable with current high throughput synthetic antibody display technologies. Leave-one-out cross validation on the benchmark system yielded the accuracy, precision, recall (sensitivity) and specificity of the overall binary predictions to be 0.69, 0.45, 0.63, and 0.71 respectively, and the overall Matthews correlation coefficient of the 20 amino acid types in the 24 interface CDR positions was 0.312. The structure-based computational antibody design methodology was further tested with other antibodies binding to VEGF. The results indicate that the methodology could provide alternatives to the current antibody technologies based on animal immune systems in engineering therapeutic and diagnostic antibodies against predetermined antigen epitopes.


Subject(s)
Antigen-Antibody Reactions , Complementarity Determining Regions , Artificial Intelligence , Binding Sites, Antibody , Crystallography, X-Ray , Humans , Models, Molecular , Reproducibility of Results , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Vascular Endothelial Growth Factor A/immunology
9.
Biochem Biophys Res Commun ; 411(2): 348-53, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21741355

ABSTRACT

Phage-displayed single chain variable fragment (scFv) libraries are powerful tools in antibody engineering. Disulfide-stabilized scFv (sc-dsFv) with an interface disulfide bond is structure-wise more stable than the corresponding scFv. A set of recently discovered signal sequences replacing the wild type (pelB) signal peptidase cleavage site in the c-region has been shown to be effective in rescuing the expression of sc-dsFv libraries on the phage surface. However, the effects of the other regions of the signal sequence on the expression of the sc-dsFv libraries and on the formation of the interface disulfide bond in the phage-displayed sc-dsFv have not been clear. In this work, selected novel signal sequence variants in the h-region were shown to be equally effective in promoting sc-dsFv library expression on the phage surface; the expression level and complexity of the sc-dsFv libraries were comparable to the corresponding scFv libraries produced with the wild-type (pelB) signal sequence. The interface disulfide bond in the phage-displayed sc-dsFv was proven to form to a large extent in the library variant ensemble generated with signal sequence variants in both the h-region and the c-region. The sc-dsFv engineering platform established in this work can be applied to many of the known scFv molecules which are in need of a more stable version for the applications under harsh conditions or for longer shelf-life.


Subject(s)
Cysteine/chemistry , Peptide Library , Protein Sorting Signals , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Vascular Endothelial Growth Factor A/immunology , Amino Acid Sequence , Humans , Molecular Sequence Data , Protein Engineering , Protein Stability , Single-Chain Antibodies/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Plant Physiol ; 154(4): 1646-58, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20935176

ABSTRACT

Glutathione S-transferases (GSTs) have been well documented to be involved in diverse aspects of biotic and abiotic stresses, especially detoxification processes. Whether they regulate plant development remains unclear. Here, we report on our isolation by reverse transcription-polymerase chain reaction of a plant GST, AtGSTU17, from Arabidopsis (Arabidopsis thaliana) and demonstrate that its expression is regulated by multiple photoreceptors, especially phytochrome A (phyA) under all light conditions. Further physiological studies indicated that AtGSTU17 participates in various aspects of seedling development, including hypocotyl elongation, anthocyanin accumulation, and far-red light-mediated inhibition of greening with a requirement of functional phyA. The loss-of-function mutant of AtGSTU17 (atgstu17) resulted in reduced biomass of seedlings and number of lateral roots in the presence of auxin, as well as insensitivity to abscisic acid (ABA)-mediated inhibition of root elongation, with similarity to different phyA mutant alleles. Moreover, the root phenotype conferred by atgstu17 was reflected by histochemical ß-glucuronidase staining of AtGSTU17 promoter activity with the addition of auxin or ABA. Further microarray analysis of wild-type Columbia and atgstu17 seedlings treated with far-red irradiation or ABA revealed that AtGSTU17 might modulate hypocotyl elongation by positively regulating some light-signaling components and negatively regulating a group of auxin-responsive genes and modulate root development by negatively controlling an auxin transport protein in the presence of ABA. Therefore, our data reveal that AtGSTU17 participates in light signaling and might modulate various aspects of Arabidopsis development by affecting glutathione pools via a coordinated regulation with phyA and phytohormones.


Subject(s)
Arabidopsis/growth & development , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Plant/physiology , Glutathione Transferase/genetics , Light , Plant Growth Regulators/physiology , Arabidopsis/enzymology , Mutation , Plant Roots/genetics
11.
Mol Biosyst ; 6(7): 1307-15, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20424732

ABSTRACT

Phage-displayed single chain variable fragment (scFv) libraries have been powerful tools in antibody engineering. But the scFv structures are frequently unstable due to the dissociation of the dimeric interface between the two variable domains. One solution is the sc-dsFv construct, where the single chain variable domain fragment is stabilized with an additional interface disulfide bond, leading to stable and homogeneous dimeric interface for the sc-dsFv structure. However, the phagemid system that is capable of effective expression for both sc-dsFv-pIII fusion proteins on phage surface and secreted non-fusion sc-dsFv in bacterial culture medium has not been demonstrated. In this work, a biological combinatorial approach was applied to optimize the signal sequence N-terminal to the sc-dsFv-pIII fusion protein encoded in a phagemid. The optimized sc-dsFv phage display systems were compatible with both the phage-based directed evolution procedure and the high throughput screening of the soluble sc-dsFv. The utility of the phagemid systems was demonstrated in generating anti-VEGF sc-dsFv with VEGF-binding affinity one order of magnitude higher than the corresponding scFv, due only to the interface disulfide bond in the sc-dsFv. Moreover, the protein stability of the sc-dsFv construct was unmatched by the corresponding scFv. These advantages of the sc-dsFv were gained through the interface disulfide bond of the sc-dsFv and the novel signal sequence in the phagemid.


Subject(s)
Protein Sorting Signals/genetics , Single-Chain Antibodies/genetics , Vascular Endothelial Growth Factors/immunology , Antibody Specificity , Base Sequence , Binding, Competitive , Blotting, Western , Disulfides/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Vascular Endothelial Growth Factors/chemistry , Vascular Endothelial Growth Factors/metabolism
12.
J Biol Chem ; 285(11): 7880-91, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20068035

ABSTRACT

Phage display of antibody fragments from natural or synthetic antibody libraries with the single chain constructs combining the variable fragments (scFv) has been one of the most prominent technologies in antibody engineering. However, the nature of the artificial single chain constructs results in unstable proteins expressed on the phage surface or as soluble proteins secreted in the bacterial culture medium. The stability of the variable domain structures can be enhanced with interdomain disulfide bond, but the single chain disulfide-stabilized constructs (sc-dsFv) have yet to be established as a feasible format for bacterial phage display due to diminishing expression levels on the phage surface in known phage display systems. In this work, biological combinatorial searches were used to establish that the c-region of the signal sequence is critically responsible for effective expression and functional folding of the sc-dsFv on the phage surface. The optimum signal sequences increase the expression of functional sc-dsFv by 2 orders of magnitude compared with wild-type signal sequences, enabling the construction of phage-displayed synthetic antivascular endothelial growth factor sc-dsFv libraries. Comparison of the scFv and sc-dsFv variants selected from the phage-displayed libraries for vascular endothelial growth factor binding revealed the sequence preference differences resulting from the interdomain disulfide bond. These results underlie a new phage display format for antibody fragments with all the benefits from the scFv format but without the downside due to the instability of the dimeric interface in scFv.


Subject(s)
Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Peptide Library , Protein Engineering/methods , Vascular Endothelial Growth Factor A , Dimerization , Disulfides/chemistry , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Gene Expression , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Mutagenesis, Site-Directed , Protein Sorting Signals/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology
13.
Plant Physiol ; 143(3): 1189-202, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17220357

ABSTRACT

Far-red (FR) insensitive 219 (FIN219) was previously shown to be involved in phytochrome A-mediated FR light signaling. To further understand its function and regulatory relation with other light-signaling components, a yeast two-hybrid approach was used to isolate FIN219-interacting partners. Here, we demonstrate that FIN219-interacting protein 1 (FIP1) interacts with FIN219 in vitro and in vivo and is composed of 217 amino acids that belong to the tau class of the large glutathione S-transferase gene family. FIP1 was further shown to have glutathione S-transferase activity. The gain of function and partial loss of function of FIP1 resulted in a hyposensitive hypocotyl phenotype under continuous FR (cFR) light and a delayed flowering phenotype under long-day conditions, which suggests that FIP1 may exist in a complex to function in the regulation of Arabidopsis (Arabidopsis thaliana) development. In addition, FIP1 mRNA was down-regulated in the suppressor of phytochrome A-105 1 mutant and differentially expressed in constitutive photomorphogenic 1-4 (cop1-4) and cop1-5 mutants under cFR. Intriguingly, FIP1 expression was up-regulated in the fin219 mutant under all light conditions, except cFR. Furthermore, promoter activity assays revealed that FIP1 expression was light dependent, mainly associated with vascular tissues, and developmentally regulated. Subcellular localization studies revealed that the beta-glucuronidase-FIP1 fusion protein was localized in the nucleus and cytoplasm. Taken together, these data indicate that FIP1 may interact with FIN219 to regulate cell elongation and flowering in response to light.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Carrier Proteins/metabolism , Glutathione Transferase/metabolism , Phytochrome A/metabolism , Signal Transduction , Amino Acid Sequence , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Carrier Proteins/analysis , Carrier Proteins/chemistry , Flowers/enzymology , Flowers/genetics , Flowers/metabolism , Gene Expression Regulation, Plant , Glucuronidase/analysis , Glutathione Transferase/physiology , Light , Molecular Sequence Data , Plants, Genetically Modified/metabolism , RNA, Messenger/metabolism , Recombinant Fusion Proteins/analysis , Sequence Alignment , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...