416 results on '"Sachdev S Sidhu"'
Search Results
352. Selection of Streptomyces griseus protease B mutants with desired alterations in primary specificity using a library screening strategy
- Author
-
Sachdev S. Sidhu and Thor J. Borgford
- Subjects
Models, Molecular ,Proteases ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Mutant ,DNA Mutational Analysis ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,law.invention ,Substrate Specificity ,Structural Biology ,law ,Enzyme Stability ,medicine ,Escherichia coli ,Amino Acids ,Protein Precursors ,Streptogrisin B ,Protein precursor ,Molecular Biology ,Gene ,Gene Library ,Protease ,Base Sequence ,Serine Endopeptidases ,Streptomyces griseus ,Genetic Variation ,Milk Proteins ,Molecular biology ,Biochemistry ,Mutagenesis ,Mutation ,Recombinant DNA ,Bacillus subtilis ,Half-Life - Abstract
Streptomyces griseus protease B (SGPB) has primary specificity for large hydrophobic residues. The protease is secreted in a promature form, and autocatalytic removal of the propeptide is essential for activity. We genetically substituted the P1 Leu at the promature junction of SGPB with Phe, Met, or Val and monitored expression levels in Escherichia coli . Substitution with Phe had no effect on active SGPB production; substitution with Met or Val abolished proteolytic activity. An E. coli expression library containing 29,952 possible SGPB mutants was constructed with variations at seven sites involved in conferring primary specificity. A rapid, visual screening strategy was used to detect active protease secretion. The expression library was screened, in conjunction with the different promature junction sequences, for those variants producing increased proteolytic activity. The sequences of the isolated mutant genes were determined; the substrate specificities and thermostabilities of the corresponding proteases were investigated. Mutants isolated from the screen with the wild-type promature junction exhibited substrate specificities and thermostabilities similar to wild-type. The screen with Phe at the promature junction P1 site resulted in the isolation of mutant proteases with increased thermostabilities (up to an order of magnitude increase in half-life at 55°C), while a protease with broad substrate specificity was isolated from the Val screen. Proteases isolated from the screen with Met at the promature junction P1 site exhibited dramatic increases in activity towards a synthetic substrate with Met at the P1 site. The results suggest that the substrate specificity of recombinant SGPB is constrained by the sequence of the promature junction; active protease production is dependent on the efficiency of the self-processive promature junction cleavage. With an efficient screening strategy, this relationship can be used to isolate catalytically active proteases with desired specificities engineered at the promature junction.
- Published
- 1996
353. T Cell Receptor-Like Recognition of Tumor In Vivo by Synthetic Antibody Fragment
- Author
-
Edward J. Collins, Hong Yuan, Bryan C. Yoder, Sachdev S. Sidhu, Akiko Koide, Michael Jay, Brenda Leung, Keith R. Miller, Jonathan Fitzsimmons, and Shohei Koide
- Subjects
Radionuclide imaging ,Receptor, ErbB-2 ,Cancer Treatment ,PET imaging ,Antigen Processing and Recognition ,Plasma protein binding ,Major Histocompatibility Complex ,Mice ,0302 clinical medicine ,Cricetinae ,Receptor ,Immunoglobulin Fragments ,0303 health sciences ,Multidisciplinary ,3. Good health ,Synthetic antibody ,Oncology ,Medicine ,Antibody ,Radiology ,Research Article ,Protein Binding ,Tumor Immunology ,Science ,Immunology ,Receptors, Antigen, T-Cell ,CHO Cells ,Biology ,Major histocompatibility complex ,03 medical and health sciences ,Immune system ,Antibody Therapy ,Antigen ,Cell Line, Tumor ,HLA-A2 Antigen ,Cancer Detection and Diagnosis ,Animals ,Humans ,030304 developmental biology ,T-cell receptor ,Immunologic Subspecialties ,Molecular biology ,Nuclear medicine ,biology.protein ,Clinical Immunology ,Peptides ,030215 immunology - Abstract
A major difficulty in treating cancer is the inability to differentiate between normal and tumor cells. The immune system differentiates tumor from normal cells by T cell receptor (TCR) binding of tumor-associated peptides bound to Major Histocompatibility Complex (pMHC) molecules. The peptides, derived from the tumor-specific proteins, are presented by MHC proteins, which then serve as cancer markers. The TCR is a difficult protein to use as a recombinant protein because of production issues and has poor affinity for pMHC; therefore, it is not a good choice for use as a tumor identifier outside of the immune system. We constructed a synthetic antibody-fragment (Fab) library in the phage-display format and isolated antibody-fragments that bind pMHC with high affinity and specificity. One Fab, fE75, recognizes our model cancer marker, the Human Epidermal growth factor Receptor 2 (HER2/neu) peptide, E75, bound to the MHC called Human Leukocyte Antigen-A2 (HLA-A2), with nanomolar affinity. The fE75 bound selectively to E75/HLA-A2 positive cancer cell lines in vitro. The fE75 Fab conjugated with (64)Cu selectively accumulated in E75/HLA-A2 positive tumors and not in E75/HLA-A2 negative tumors in an HLA-A2 transgenic mouse as probed using positron emission tomography/computed tomography (PET/CT) imaging. Considering that hundreds to thousands of different peptides bound to HLA-A2 are present on the surface of each cell, the fact that fE75 arrives at the tumor at all shows extraordinary specificity. These antibody fragments have great potential for diagnosis and targeted drug delivery in cancer.
- Published
- 2012
354. Charged and Hydrophobic Surfaces on the A Chain of Shiga-Like Toxin 1 Recognize the C-Terminal Domain of Ribosomal Stalk Proteins
- Author
-
Sachdev S. Sidhu, Jean Gariépy, Eleonora Bolewska-Pedyczak, Andrew J. McCluskey, Gang Chen, and Nick Jarvik
- Subjects
Ribosomal Proteins ,Molecular Sequence Data ,Protein domain ,lcsh:Medicine ,Tripeptide ,Shiga Toxin 1 ,Toxicology ,Biochemistry ,Microbiology ,Ribosome ,03 medical and health sciences ,Ribosomal protein ,Catalytic Domain ,Two-Hybrid System Techniques ,Amino Acid Sequence ,lcsh:Science ,Biology ,Peptide sequence ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Sequence Homology, Amino Acid ,Tetrapeptide ,030306 microbiology ,Chemistry ,C-terminus ,lcsh:R ,Surface Plasmon Resonance ,Ribosomal RNA ,Recombinant Proteins ,Protein Structure, Tertiary ,Bacterial Pathogens ,RNA, Ribosomal ,Protein Biosynthesis ,Medicine ,lcsh:Q ,Hydrophobic and Hydrophilic Interactions ,Oligopeptides ,Ribosomes ,Research Article - Abstract
Shiga-like toxins are ribosome-inactivating proteins (RIP) produced by pathogenic E. coli strains that are responsible for hemorrhagic colitis and hemolytic uremic syndrome. The catalytic A(1) chain of Shiga-like toxin 1 (SLT-1), a representative RIP, first docks onto a conserved peptide SD[D/E]DMGFGLFD located at the C-terminus of all three eukaryotic ribosomal stalk proteins and halts protein synthesis through the depurination of an adenine base in the sarcin-ricin loop of 28S rRNA. Here, we report that the A(1) chain of SLT-1 rapidly binds to and dissociates from the C-terminal peptide with a monomeric dissociation constant of 13 µM. An alanine scan performed on the conserved peptide revealed that the SLT-1 A(1) chain interacts with the anionic tripeptide DDD and the hydrophobic tetrapeptide motif FGLF within its sequence. Based on these 2 peptide motifs, SLT-1 A(1) variants were generated that displayed decreased affinities for the stalk protein C-terminus and also correlated with reduced ribosome-inactivating activities in relation to the wild-type A(1) chain. The toxin-peptide interaction and subsequent toxicity were shown to be mediated by cationic and hydrophobic docking surfaces on the SLT-1 catalytic domain. These docking surfaces are located on the opposite face of the catalytic cleft and suggest that the docking of the A(1) chain to SDDDMGFGLFD may reorient its catalytic domain to face its RNA substrate. More importantly, both the delineated A(1) chain ribosomal docking surfaces and the ribosomal peptide itself represent a target and a scaffold, respectively, for the design of generic inhibitors to block the action of RIPs.
- Published
- 2012
355. Synthetic antibodies as tools to probe RNA-binding protein function
- Author
-
J. Timothy Westwood, Pietro Sollazzo, Craig A. Smibert, Sachdev S. Sidhu, Howard D. Lipshitz, Kristin Ancevicius, and John D. Laver
- Subjects
Genetics ,endocrine system ,Phage display ,Immunoprecipitation ,RNA-Binding Proteins ,RNA-binding protein ,Computational biology ,Biology ,Models, Biological ,Antibodies ,Synthetic antibody ,Repressor Proteins ,Gene Expression Regulation ,Molecular Probes ,RNA splicing ,Drosophila Proteins ,Humans ,Synthetic Biology ,Smaug ,Target protein ,Antigens ,Molecular Biology ,Function (biology) ,Biotechnology - Abstract
RNA-binding proteins (RBPs) have essential roles in post-transcriptional regulation of gene expression. They bind sequence elements in specific mRNAs and control their splicing, transport, localization, translation, and stability. A complete understanding of RBP function requires identification of the target RNAs that an RBP regulates, the mechanisms by which the RBP regulates these targets, and the biological consequences for the cell in which these transactions occur. Antibodies are key tools in such studies: first, mRNA targets of RBPs can be identified by co-immunoprecipitation of RBPs with their associated RNAs followed by microarray analysis or sequencing; second, partner proteins can be identified by immunoprecipitation of the RBP followed by mass spectrometry; third, the mechanisms and functions of RBPs can be inferred from loss-of-function studies employing antibodies that block RBP-RNA interactions. One potentially powerful approach to making antibodies for such studies is the generation of synthetic antibodies using phage display, which involves in vitro selection using a human-designed antibody library to generate antibodies that recognize a target protein. Using two well-characterized Drosophila RNA-binding proteins, Staufen and Smaug, for proof-of-principle, we demonstrate that synthetic antibodies can be generated and used either to perform RNA-coimmunoprecipitations (RIPs) to identify RBP-bound mRNAs, or to block RBP-RNA interactions. Given that synthetic antibody selection protocols are amenable to high-throughput antibody production, these results demonstrate that synthetic antibodies can be powerful tools for genome-wide studies of RBP function.
- Published
- 2012
356. MUSI: an integrated system for identifying multiple specificity from very large peptide or nucleic acid data sets
- Author
-
Haiming Huang, Sachdev S. Sidhu, Gary D. Bader, Marc S. Tyndel, Philip M. Kim, David Gfeller, and TaeHyung Kim
- Subjects
Cell signaling ,Phage display ,Computational biology ,Biology ,Ligands ,src Homology Domains ,Set (abstract data type) ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Peptide Library ,Sequence Analysis, Protein ,Genetics ,Animals ,Humans ,Position-Specific Scoring Matrices ,Protein Interaction Domains and Motifs ,Transcription factor ,Binding selectivity ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Microarray analysis techniques ,High-Throughput Nucleotide Sequencing ,Nucleic acid ,Methods Online ,DNA microarray ,Peptides ,Software ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Peptide recognition domains and transcription factors play crucial roles in cellular signaling. They bind linear stretches of amino acids or nucleotides, respectively, with high specificity. Experimental techniques that assess the binding specificity of these domains, such as microarrays or phage display, can retrieve thousands of distinct ligands, providing detailed insight into binding specificity. In particular, the advent of next-generation sequencing has recently increased the throughput of such methods by several orders of magnitude. These advances have helped reveal the presence of distinct binding specificity classes that co-exist within a set of ligands interacting with the same target. Here, we introduce a software system called MUSI that can rapidly analyze very large data sets of binding sequences to determine the relevant binding specificity patterns. Our pipeline provides two major advances. First, it can detect previously unrecognized multiple specificity patterns in any data set. Second, it offers integrated processing of very large data sets from next-generation sequencing machines. The results are visualized as multiple sequence logos describing the different binding preferences of the protein under investigation. We demonstrate the performance of MUSI by analyzing recent phage display data for human SH3 domains as well as microarray data for mouse transcription factors.
- Published
- 2011
357. Identification of specificity determining residues in peptide recognition domains using an information theoretic approach applied to large-scale binding maps
- Author
-
Xihao Hu, Simon Sitwell, Kevin Y. Yip, Lukas Utz, Benjamin E. Turk, Mark Gerstein, Philip M. Kim, and Sachdev S. Sidhu
- Subjects
Models, Molecular ,Physiology ,PDZ domain ,Information Theory ,Scale (descriptive set theory) ,Peptide ,Plant Science ,Plasma protein binding ,Computational biology ,Biology ,Information theory ,General Biochemistry, Genetics and Molecular Biology ,Structural Biology ,Humans ,Position-Specific Scoring Matrices ,Amino Acids ,lcsh:QH301-705.5 ,Ecology, Evolution, Behavior and Systematics ,Binding selectivity ,chemistry.chemical_classification ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Reproducibility of Results ,Cell Biology ,Protein Structure, Tertiary ,Identification (information) ,Biochemistry ,chemistry ,lcsh:Biology (General) ,Mutagenesis ,General Agricultural and Biological Sciences ,Peptides ,Algorithms ,Developmental Biology ,Biotechnology ,Research Article ,Protein Binding - Abstract
Background Peptide Recognition Domains (PRDs) are commonly found in signaling proteins. They mediate protein-protein interactions by recognizing and binding short motifs in their ligands. Although a great deal is known about PRDs and their interactions, prediction of PRD specificities remains largely an unsolved problem. Results We present a novel approach to identifying these Specificity Determining Residues (SDRs). Our algorithm generalizes earlier information theoretic approaches to coevolution analysis, to become applicable to this problem. It leverages the growing wealth of binding data between PRDs and large numbers of random peptides, and searches for PRD residues that exhibit strong evolutionary covariation with some positions of the statistical profiles of bound peptides. The calculations involve only information from sequences, and thus can be applied to PRDs without crystal structures. We applied the approach to PDZ, SH3 and kinase domains, and evaluated the results using both residue proximity in co-crystal structures and verified binding specificity maps from mutagenesis studies. Discussion Our predictions were found to be strongly correlated with the physical proximity of residues, demonstrating the ability of our approach to detect physical interactions of the binding partners. Some high-scoring pairs were further confirmed to affect binding specificity using previous experimental results. Combining the covariation results also allowed us to predict binding profiles with higher reliability than two other methods that do not explicitly take residue covariation into account. Conclusions The general applicability of our approach to the three different domain families demonstrated in this paper suggests its potential in predicting binding targets and assisting the exploration of binding mechanisms.
- Published
- 2011
358. The functional capacity of the natural amino acids for molecular recognition
- Author
-
Sara C. Birtalan, Robert D. Fisher, and Sachdev S. Sidhu
- Subjects
Receptor, ErbB-2 ,Stereochemistry ,Molecular Sequence Data ,Plasma protein binding ,Binding, Competitive ,DNA-binding protein ,Antibodies ,Cell Line ,Residue (chemistry) ,Molecular recognition ,Peptide Library ,Serine ,Animals ,Humans ,Amino Acid Sequence ,Amino Acids ,Antigens ,Peptide library ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,Binding Sites ,Sequence Homology, Amino Acid ,Flow Cytometry ,Complementarity Determining Regions ,Amino acid ,Synthetic antibody ,chemistry ,Biochemistry ,Tyrosine ,Protein Binding ,Biotechnology - Abstract
We tested the functional capacity of the natural amino acids for molecular recognition in a minimalist background of binary Tyr/Ser diversity. In phage-displayed synthetic antibody libraries, we replaced either Tyr or Ser with other residues. We find that Tyr is optimal for mediating contacts that contribute favourably to both affinity and specificity, but it can be replaced by Trp, which contributes favourably to affinity but is detrimental to specificity. Arg exhibited a limited capacity for mediating molecular recognition but was less effective than either Tyr or Trp, and moreover, was the major contributor to non-specific interactions. Nine other residue types (Phe, Leu, Ile, Asn, Thr, Pro, Cys, Ala, and Gly) were found to be ineffective as replacements for Tyr. By replacing Ser with Gly or Ala, we found that Gly is as effective as Ser for providing conformational flexibility that allows bulky Tyr residues to achieve optimal binding contacts, while Ala is less effective but still functional in this capacity. For some antigens, high affinity antibodies could be derived using only Tyr/Ser/Gly diversity, but for others, additional chemical diversity was required to achieve high affinity. Our results establish a minimal benchmark for the generation of synthetic antigen-binding sites with affinities comparable to those of natural antibodies. Moreover, our findings illuminate the fundamental principles underlying protein-protein interactions and provide valuable guidelines for engineering synthetic binding proteins with functions beyond the scope of natural proteins.
- Published
- 2010
359. Coevolution of PDZ domain–ligand interactions analyzed by high-throughput phage display and deep sequencing
- Author
-
Andreas Ernst, Somasekar Seshagiri, Zhengyan Kan, Sachdev S. Sidhu, David Gfeller, Gary D. Bader, and Philip M. Kim
- Subjects
Genetics ,Phage display ,PDZ domain ,PDZ Domains ,Cooperativity ,Computational biology ,Plasma protein binding ,Biology ,Ligands ,Deep sequencing ,Evolution, Molecular ,Peptide Library ,Bacteriophages ,Amino Acid Sequence ,Binding site ,Peptides ,Peptide library ,Molecular Biology ,Peptide sequence ,Protein Binding ,Biotechnology - Abstract
The determinants of binding specificities of peptide recognition domains and their evolution remain important problems in molecular systems biology. Here, we present a new methodology to analyze the coevolution between a domain and its ligands by combining high-throughput phage display with deep sequencing. First, from a library of PDZ domains with diversity introduced at ten positions in the binding site, we evolved domains for binding to 15 distinct peptide ligands. Interestingly, for a given peptide many different functional domains emerged, which exhibited only limited sequence homology, showing that many different binding sites can recognize a given peptide. Subsequently, we used peptide-phage libraries and deep sequencing to map the specificity profiles of these evolved domains at high resolution, and we found that the domains recognize their cognate peptides with high affinity but low specificity. Our analysis reveals two aspects of evolution of new binding specificities. First, we were able to identify some common features amongst domains raised against a common peptide. Second, our analysis suggests that cooperative interactions between multiple binding site residues lead to a diversity of binding profiles with considerable plasticity. The details of intramolecular cooperativity remain to be elucidated, but nonetheless, we have established a general methodology that can be used to explore protein evolution in a systematic yet rapid manner.
- Published
- 2010
360. Crtam regulates a late phase of T cell polarity and IFNγ/IL22 cytokine production
- Author
-
Andrew C. Chan, Jung-Hua Yeh, and Sachdev S. Sidhu
- Subjects
Chemistry ,Polarity (physics) ,medicine.medical_treatment ,T cell ,Biochemistry ,Cell biology ,Interleukin 22 ,Cytokine ,medicine.anatomical_structure ,Late phase ,Genetics ,medicine ,Molecular Biology ,Biotechnology - Published
- 2008
361. Phage Display In Biotechnology and Drug Discovery
- Author
-
Sachdev S. Sidhu, Clarence Ronald Geyer, Sachdev S. Sidhu, and Clarence Ronald Geyer
- Subjects
- Peptides--Biotechnology, Affinity chromatography, Biotechnology, Proteins--Biotechnology, Microbial biotechnology, Bacteriophages, Viral proteins, Pharmaceutical biotechnology, Bioengineering, Pharmaceutical chemistry
- Abstract
The first and only guide to showcase the impact of phage display technology on drug discovery, this reference details the theories, principles, and methods impacting the field and demonstrates applications for peptide phage display, protein phage display, and the development of novel antibodies. Highlighting the current and future role of phage dis
- Published
- 2005
362. Cover Picture: ChemBioChem 1/2003
- Author
-
Sachdev S. Sidhu, Kurt Deshayes, and Wayne J. Fairbrother
- Subjects
chemistry.chemical_classification ,biology ,Stereochemistry ,Chemistry ,Organic Chemistry ,Ligand (biochemistry) ,Biochemistry ,Cell membrane ,Transmembrane domain ,medicine.anatomical_structure ,Rhodopsin ,biology.protein ,medicine ,Molecular Medicine ,Nucleotide ,Receptor ,Molecular Biology ,Intracellular ,G protein-coupled receptor - Abstract
The cover picture shows bovine rhodopsin, the first G-protein-coupled receptor (GPCR) structure determined at atomic resolution. It is embedded into the cell membrane with the conserved seven transmembrane helices. Rhodopsin is unique among GPCRs due to its activation mechanism: The covalently bound ligand retinal (shown in red) undergoes a light-induced isomerization, thus activating the receptor. For other GPCRs intracellular responses are modulated by binding of extracellular signal molecules and drugs. Valsartan, fexofenadine, montelukast, and clapidogrel are examples of drug compounds acting on GPCRs that bind peptides, biogenic amines, lipids, and nucleotides, respectively. Further details can be found in the review article by Klabunde and Hessler on p. 928 ff.
- Published
- 2003
363. Getting what you want from phage display
- Author
-
Sachdev S. Sidhu
- Subjects
Microbiology (medical) ,Protein structure and function ,Alternative methods ,Genetics ,Protein function ,Phage display ,Combinatorial biology ,Biology ,Microbiology ,Genome ,Field (computer science) ,World Wide Web ,Infectious Diseases ,Virology ,Genomic information - Abstract
Phage Display: A Laboratory Manualedited by C.F. Barbas, III et al.Cold Spring Harbour Laboratory Press, 2001.$135.00 (hbk) (xvi + 736 pages)ISBN 0 87969 546 3Recent years have witnessed the complete sequencing of numerous genomes, including the all-important human genome itself. However, in the midst of this genomic revolution, the study of protein structure and function has lagged behind. The reason for this is simple: DNA manipulation is easy whereas protein manipulation is exceedingly difficult. Nevertheless, as we enter the post-genomic era, it must be remembered that we live in a protein world. The vast majority of biological processes are driven by proteins, and the benefits of genome sequences will only be realized if genomic information can be translated to the level of protein function. We are now faced with tens of thousands of proteins waiting to be studied, and there is a crucial need for technologies that can simplify this herculean task.Phage display is a powerful technology that has played a major role in protein engineering over the past decade, and this role will certainly expand in the coming years. This technology links proteins on the surfaces of M13 bacteriophages with their cognate genes inside the phage particles. Billions of different proteins can be screened en masse for particular functions, and individual protein sequences can be decoded from the sequences of their cognate DNA. In essence, phage display enables the study of protein function using simple molecular biology techniques that were previously applicable only to DNA. The history of phage display is marked by many celebrated successes and an even greater number of uncelebrated failures and, as a result, this technology has earned the reputation of a powerful but capricious pursuit. Although the complexity of the method cannot be denied, most phage failures can be attributed to inexperienced or naive practitioners, and there is a real need for literature that focuses on the practical aspects of phage display.Phage Display: A Laboratory Manual aims to satisfy this need by providing practical methods based mainly on the Cold Spring Harbor course ‘Phage Display of Combinatorial Antibody Libraries’. The manual emphasizes antibody and peptide libraries, providing detailed methods in major sections devoted entirely to these topics. The use of cDNA libraries is also covered in detail; other protein engineering applications are touched on lightly in the form of literature reviews. As an introduction, Robert Webster provides an excellent review of filamentous phage biology. Webster's chapter is highly recommended to all phage display practitioners because ultimately, current success and future innovation depends on this fundamental knowledge. Overall, the manual does a solid job of the topics chosen for coverage, providing detailed methods and references to other works in the field.Phage display is a rapidly expanding technology and many alternative methods have proven successful in particular applications. Although the current volume does a good job of covering the editors’ interests and views, no single work can cover the full breadth of the field, and there is certainly room on the shelf for other points of view. In this regard, other laboratory manuals are worth investigating1xSee all References, 2xSee all References. Also, a recently published Methods in Enzymology3xSee all References3 is highly recommended as it contains several chapters on phage display and excellent articles on alternative combinatorial biology methods. Finally, keeping up in the fast-paced world of phage display requires constant perusal of current reviews and original literature. With all of this knowledge in hand, you just might select what you want.
- Published
- 2001
364. Functional complexes between YAP2 and ZO-2 are PDZ domain-dependent, and regulate YAP2 nuclear localization and signalling1.
- Author
-
Tsutomu Oka, David Gfeller, Gary D. Bader, and Sachdev S. Sidhu
- Subjects
GENETIC regulation ,APOPTOSIS ,GENETIC transcription ,PROTEIN kinases ,BIOCHEMICAL mechanism of action ,CANCER treatment ,CELLULAR signal transduction ,CELL culture - Abstract
The Hippo pathway regulates the size of organs by controlling two opposing processes: proliferation and apoptosis. YAP2 (Yes kinase-associated protein 2), one of the three isoforms of YAP, is a WW domain-containing transcriptional co-activator that acts as the effector of the Hippo pathway in mammalian cells. In addition to WW domains, YAP2 has a PDZ-binding motif at its C-terminus. We reported previously that this motif was necessary for YAP2 localization in the nucleus and for promoting cell detachment and apoptosis. In the present study, we show that the tight junction protein ZO (zonula occludens)-2 uses its first PDZ domain to form a complex with YAP2. The endogenous ZO-2 and YAP2 proteins co-localize in the nucleus. We also found that ZO-2 facilitates the nuclear localization and pro-apoptotic function of YAP2, and that this activity of ZO-2 is PDZ-domain-dependent. The present paper is the first report on a PDZ-based nuclear translocation mechanism. Moreover, since the Hippo pathway acts as a tumour suppressor pathway, the YAP2–ZO-2 complex could represent a target for cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
365. Interaction Domains of Sos1/Grb2 Are Finely Tuned for Cooperative Control of Embryonic Stem Cell Fate
- Author
-
Monika Tucholska, Janet Rossant, Haiming Huang, Katie Cockburn, Lorne Taylor, Greg M. Findlay, Fredrik Lanner, Marilyn Hsiung, Mitshuhiko Ikura, Sachdev S. Sidhu, Evangelia Petsalaki, Shawn S.-C. Li, Tomonori Kaneko, Troy Ketela, Richard D. Bagshaw, Jason Moffat, David David D Bowtell, Gerald D. Gish, Tony Pawson, and Matthew J. Smith
- Subjects
Models, Molecular ,Lineage (genetic) ,Protein domain ,Molecular Sequence Data ,Cooperativity ,Saccharomyces cerevisiae ,Fibroblast growth factor ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Cell Lineage ,Amino Acid Sequence ,Embryonic Stem Cells ,030304 developmental biology ,GRB2 Adaptor Protein ,Genetics ,0303 health sciences ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Endoderm ,Eukaryota ,Embryo, Mammalian ,Embryonic stem cell ,Cell biology ,Protein Structure, Tertiary ,SOS1 ,biology.protein ,ras Guanine Nucleotide Exchange Factors ,GRB2 ,Guanine nucleotide exchange factor ,SOS1 Protein ,Sequence Alignment ,030217 neurology & neurosurgery - Abstract
SummaryMetazoan evolution involves increasing protein domain complexity, but how this relates to control of biological decisions remains uncertain. The Ras guanine nucleotide exchange factor (RasGEF) Sos1 and its adaptor Grb2 are multidomain proteins that couple fibroblast growth factor (FGF) signaling to activation of the Ras-Erk pathway during mammalian development and drive embryonic stem cells toward the primitive endoderm (PrE) lineage. We show that the ability of Sos1/Grb2 to appropriately regulate pluripotency and differentiation factors and to initiate PrE development requires collective binding of multiple Sos1/Grb2 domains to their protein and phospholipid ligands. This provides a cooperative system that only allows lineage commitment when all ligand-binding domains are occupied. Furthermore, our results indicate that the interaction domains of Sos1 and Grb2 have evolved so as to bind ligands not with maximal strength but with specificities and affinities that maintain cooperativity. This optimized system ensures that PrE lineage commitment occurs in a timely and selective manner during embryogenesis.
- Full Text
- View/download PDF
366. Ubiquitin Chain Editing Revealed by Polyubiquitin Linkage-Specific Antibodies
- Author
-
Vishva M. Dixit, Nathaniel C. Gordon, Deanne M. Compaan, Donald S. Kirkpatrick, Ronald E. Ferrando, Jenille Tan, Dorothy French, Marissa L. Matsumoto, Kim Newton, Debra L. Dugger, Ivan Bosanac, Ingrid E. Wertz, Christine Yu, Robert F. Kelley, Jennie R. Lill, László G. Kömüves, Sarah G. Hymowitz, Cynthia Lam, Frederic A. Fellouse, and Sachdev S. Sidhu
- Subjects
PROTEINS ,Lysine ,Mutant ,Saccharomyces cerevisiae ,Cellular homeostasis ,macromolecular substances ,environment and public health ,Antibodies ,Mass Spectrometry ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,Ubiquitin ,Peptide Library ,Schizosaccharomyces ,Animals ,Humans ,MOLIMMUNO ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,C-terminus ,Ubiquitination ,RNA-Binding Proteins ,biology.organism_classification ,Mice, Inbred C57BL ,Nuclear Pore Complex Proteins ,Interleukin-1 Receptor-Associated Kinases ,Biochemistry ,biology.protein ,CELLBIO ,Signal transduction - Abstract
Summary Posttranslational modification of proteins with polyubiquitin occurs in diverse signaling pathways and is tightly regulated to ensure cellular homeostasis. Studies employing ubiquitin mutants suggest that the fate of polyubiquitinated proteins is determined by which lysine within ubiquitin is linked to the C terminus of an adjacent ubiquitin. We have developed linkage-specific antibodies that recognize polyubiquitin chains joined through lysine 63 (K63) or 48 (K48). A cocrystal structure of an anti-K63 linkage Fab bound to K63-linked diubiquitin provides insight into the molecular basis for specificity. We use these antibodies to demonstrate that RIP1, which is essential for tumor necrosis factor-induced NF-κB activation, and IRAK1, which participates in signaling by interleukin-1β and Toll-like receptors, both undergo polyubiquitin editing in stimulated cells. Both kinase adaptors initially acquire K63-linked polyubiquitin, while at later times K48-linked polyubiquitin targets them for proteasomal degradation. Polyubiquitin editing may therefore be a general mechanism for attenuating innate immune signaling.
- Full Text
- View/download PDF
367. Synthetic antibodies block receptor binding and current‐inhibiting effects of α‐cobratoxin from Naja kaouthia
- Author
-
Shane Miersch, Guillermo de la Rosa, Rasmus Friis, Line Ledsgaard, Kim Boddum, Andreas H. Laustsen, and Sachdev S. Sidhu
- Subjects
Sheep ,Antivenins ,Full‐length Papers ,Neurotoxins ,Naja naja ,Animals ,Humans ,Horses ,Cobra Neurotoxin Proteins ,Molecular Biology ,Biochemistry - Abstract
Each year, thousands of people fall victim to envenomings caused by cobras. These incidents often result in death due to paralysis caused by α‐neurotoxins from the three‐finger toxin (3FTx) family, which are abundant in elapid venoms. Due to their small size, 3FTxs are among the snake toxins that are most poorly neutralized by current antivenoms, which are based on polyclonal antibodies of equine or ovine origin. While antivenoms have saved countless lives since their development in the late 18th century, an opportunity now exists to improve snakebite envenoming therapy via the application of new biotechnological methods, particularly by developing monoclonal antibodies against poorly neutralized α‐neurotoxins. Here, we describe the use of phage‐displayed synthetic antibody libraries and the development and characterization of six synthetic antibodies built on a human IgG framework and developed against α‐cobratoxin – the most abundant long‐chain α‐neurotoxin from Naja kaouthia venom. The synthetic antibodies exhibited sub‐nanomolar affinities to α‐cobratoxin and neutralized the curare‐mimetic effect of the toxin in vitro. These results demonstrate that phage display technology based on synthetic repertoires can be used to rapidly develop human antibodies with drug‐grade potencies as inhibitors of venom toxins.
- Full Text
- View/download PDF
368. Brain tumor is a sequence-specific RNA-binding protein that directs maternal mRNA clearance during the Drosophila maternal-to-zygotic transition
- Author
-
Noah A Hahn, Alexander Marsolais, Xiao Li, Debashish Ray, J. Timothy Westwood, Craig A. Smibert, John D. Laver, Karen Yy Fung, Syed Nabeel-Shah, Hua Luo, Sachdev S. Sidhu, Howard D. Lipshitz, Kate B. Cook, Timothy P. Hughes, Mariana Kekis, and Quaid Morris
- Subjects
Protein family ,RNA-binding protein ,Biology ,Epigenetic Repression ,Tissue Culture Techniques ,Animals ,Drosophila Proteins ,Transcription factor ,Genetic Association Studies ,Genetics ,Regulation of gene expression ,Messenger RNA ,Binding Sites ,Brain Neoplasms ,Research ,RNA ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,RNA-Binding Proteins ,DNA-Binding Proteins ,RNA, Messenger, Stored ,Mutation ,Maternal to zygotic transition ,Drosophila ,Female ,Drosophila Protein ,Transcription Factors - Abstract
Background Brain tumor (BRAT) is a Drosophila member of the TRIM-NHL protein family. This family is conserved among metazoans and its members function as post-transcriptional regulators. BRAT was thought to be recruited to mRNAs indirectly through interaction with the RNA-binding protein Pumilio (PUM). However, it has recently been demonstrated that BRAT directly binds to RNA. The precise sequence recognized by BRAT, the extent of BRAT-mediated regulation, and the exact roles of PUM and BRAT in post-transcriptional regulation are unknown. Results Genome-wide identification of transcripts associated with BRAT or with PUM in Drosophila embryos shows that they bind largely non-overlapping sets of mRNAs. BRAT binds mRNAs that encode proteins associated with a variety of functions, many of which are distinct from those implemented by PUM-associated transcripts. Computational analysis of in vitro and in vivo data identified a novel RNA motif recognized by BRAT that confers BRAT-mediated regulation in tissue culture cells. The regulatory status of BRAT-associated mRNAs suggests a prominent role for BRAT in post-transcriptional regulation, including a previously unidentified role in transcript degradation. Transcriptomic analysis of embryos lacking functional BRAT reveals an important role in mediating the decay of hundreds of maternal mRNAs during the maternal-to-zygotic transition. Conclusions Our results represent the first genome-wide analysis of the mRNAs associated with a TRIM-NHL protein and the first identification of an RNA motif bound by this protein family. BRAT is a prominent post-transcriptional regulator in the early embryo through mechanisms that are largely independent of PUM. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0659-4) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
369. Cell-based passive immunization for protection against SARS-CoV-2 infection
- Author
-
Evan Sawula, Shane Miersch, Eric D. Jong, Chengjin Li, Fang-Yu Chou, Jean Kit Tang, Reza Saberianfar, Jeffrey Harding, Sachdev S. Sidhu, and Andras Nagy
- Subjects
SARS-CoV-2 ,COVID-19 ,Neutralizing antibodies ,Passive immunization ,Cell therapy ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Immunologically impaired individuals respond poorly to vaccines, highlighting the need for additional strategies to protect these vulnerable populations from COVID-19. While monoclonal antibodies (mAbs) have emerged as promising tools to manage infectious diseases, the transient lifespan of neutralizing mAbs in patients limits their ability to confer lasting, passive prophylaxis from SARS-CoV-2. Here, we attempted to solve this problem by combining cell and mAb engineering in a way that provides durable immune protection against viral infection using safe and universal cell therapy. Methods Mouse embryonic stem cells equipped with our FailSafe™ and induced allogeneic cell tolerance technologies were engineered to express factors that potently neutralize SARS-CoV-2, which we call ‘neutralizing biologics’ (nBios). We subcutaneously transplanted the transgenic cells into mice and longitudinally assessed the ability of the cells to deliver nBios into circulation. To do so, we quantified plasma nBio concentrations and SARS-CoV-2 neutralizing activity over time in transplant recipients. Finally, using similar cell engineering strategies, we genetically modified FailSafe™ human-induced pluripotent stem cells to express SARS-CoV-2 nBios. Results Transgenic mouse embryonic stem cells engineered for safety and allogeneic-acceptance can secrete functional and potent SARS-CoV-2 nBios. As a dormant, subcutaneous tissue, the transgenic cells and their differentiated derivatives long-term deliver a supply of protective nBio titers in vivo. Moving toward clinical relevance, we also show that human-induced pluripotent stem cells, similarly engineered for safety, can secrete highly potent nBios. Conclusions Together, these findings show the promise and potential of using ‘off-the-shelf’ cell products that secrete neutralizing antibodies for sustained protective immunity against current and future viral pathogens of public health significance.
- Published
- 2023
- Full Text
- View/download PDF
370. Author Correction: Discovery of an exosite on the SOCS2-SH2 domain that enhances SH2 binding to phosphorylated ligands
- Author
-
Edmond M. Linossi, Kunlun Li, Gianluca Veggiani, Cyrus Tan, Farhad Dehkhoda, Colin Hockings, Dale J. Calleja, Narelle Keating, Rebecca Feltham, Andrew J. Brooks, Shawn S. Li, Sachdev S. Sidhu, Jeffrey J. Babon, Nadia J. Kershaw, and Sandra E. Nicholson
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
371. Single-cell transcriptomics identifies a WNT7A-FZD5 signaling axis that maintains fallopian tube stem cells in patient-derived organoids
- Author
-
Abdulkhaliq Alsaadi, Mara Artibani, Zhiyuan Hu, Nina Wietek, Matteo Morotti, Laura Santana Gonzalez, Moiad Alazzam, Jason Jiang, Beena Abdul, Hooman Soleymani majd, Levi L. Blazer, Jarret Adams, Francesca Silvestri, Sachdev S. Sidhu, Joan S. Brugge, and Ahmed Ashour Ahmed
- Subjects
CP: Stem cell research ,Biology (General) ,QH301-705.5 - Abstract
Summary: The study of fallopian tube (FT) function in health and disease has been hampered by limited knowledge of FT stem cells and lack of in vitro models of stem cell renewal and differentiation. Using optimized organoid culture conditions to address these limitations, we find that FT stem cell renewal is highly dependent on WNT/β-catenin signaling and engineer endogenous WNT/β-catenin signaling reporter organoids to biomark, isolate, and characterize these cells. Using functional approaches, as well as bulk and single-cell transcriptomics analyses, we show that an endogenous hormonally regulated WNT7A-FZD5 signaling axis is critical for stem cell renewal and that WNT/β-catenin pathway-activated cells form a distinct transcriptomic cluster of FT cells enriched in extracellular matrix (ECM) remodeling and integrin signaling pathways. Overall, we provide a deep characterization of FT stem cells and their molecular requirements for self-renewal, paving the way for mechanistic work investigating the role of stem cells in FT health and disease.
- Published
- 2023
- Full Text
- View/download PDF
372. Nebulization of pharmacological solutions with an innovative medical device based on microvaporization
- Author
-
Bruno Brandimarte, Lino Di Rienzo Businco, Francesco Cappello, Roberto Fiore, Giuseppe Bastone, Gianfranco Gualdi, Saadi Sollaku, Emanuele Casciani, Federica Tortorella, Pasquale Longo, Eleonora Centanini, Silva Pavaci, Federica Sangiuolo, Maria Patrizia Patrizi, Shane Miersch, Sachdev S. Sidhu, Virgilio Sacchini, and Giuseppe Novelli
- Subjects
Microdrop ,Induction energy ,Drug delivery device ,Aerosol ,COVID-19 ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
The currently available nebulization devices have a slow aerosol flow and produce vapor with large microdrops. Improved devices that achieve higher airflow and produce smaller microdrops are needed to improve the clinical care of patients. To address this critical need, we developed a novel system for the molecular vaporization of liquids. This device vaporizes an active pharmacological substance dissolved in water, alcohol, or a mixture of water and alcohol using two energy sources at the same time: high-frequency ultrasound and thermal induction. Application of energy to a solution contained in the device’s tank allows, within tens of seconds, for the vaporization of the solution itself, with the generation of a vapor consisting of microdrops of very small diameter (0.2–0.3 μm). In this article, we illustrate the technology used, the main verification tests performed, and the primary fields of application for this device. In particular, the advantages of both the aerosol delivery system and the administration system are highlighted.
- Published
- 2023
- Full Text
- View/download PDF
373. Discovery of an exosite on the SOCS2-SH2 domain that enhances SH2 binding to phosphorylated ligands
- Author
-
Edmond M. Linossi, Kunlun Li, Gianluca Veggiani, Cyrus Tan, Farhad Dehkhoda, Colin Hockings, Dale J. Calleja, Narelle Keating, Rebecca Feltham, Andrew J. Brooks, Shawn S. Li, Sachdev S. Sidhu, Jeffrey J. Babon, Nadia J. Kershaw, and Sandra E. Nicholson
- Subjects
Science - Abstract
Abstract Suppressor of cytokine signaling (SOCS)2 protein is a key negative regulator of the growth hormone (GH) and Janus kinase (JAK)-Signal Transducers and Activators of Transcription (STAT) signaling cascade. The central SOCS2-Src homology 2 (SH2) domain is characteristic of the SOCS family proteins and is an important module that facilitates recognition of targets bearing phosphorylated tyrosine (pTyr) residues. Here we identify an exosite on the SOCS2-SH2 domain which, when bound to a non-phosphorylated peptide (F3), enhances SH2 affinity for canonical phosphorylated ligands. Solution of the SOCS2/F3 crystal structure reveals F3 as an α-helix which binds on the opposite side of the SH2 domain to the phosphopeptide binding site. F3:exosite binding appears to stabilise the SOCS2-SH2 domain, resulting in slower dissociation of phosphorylated ligands and consequently, enhances binding affinity. This biophysical enhancement of SH2:pTyr binding affinity translates to increase SOCS2 inhibition of GH signaling.
- Published
- 2021
- Full Text
- View/download PDF
374. Monoclonal antibodies binding data for SARS-CoV-2 proteins
- Author
-
Nawneet Mishra, Joan Teyra, RuthMabel Boytz, Shane Miersch, Trudy N. Merritt, Lia Cardarelli, Maryna Gorelik, Filip Mihalic, Per Jemth, Robert A. Davey, Sachdev S. Sidhu, Daisy W. Leung, and Gaya K. Amarasinghe
- Subjects
ELISA ,B.L.I. ,SARS-CoV-2 ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
SARS-CoV-2 pandemic opens up the curiosity of understanding the coronavirus. This demand for the development of the regent, which can be used for academic and therapeutic applications. The present data provide the biochemical characterization of synthetically developed monoclonal antibodies for the SARS-CoV-2 proteins. The antibodies from phage-displayed antibody libraries were selected with the SARS-CoV-2 proteins immobilized in microwell plates. The clones which bind to the antigen in Fab-phage ELISA were selected, and a two-point competitive phage ELISA was performed. Antibodies binding kinetic of IgGs for SARS-CoV2 proteins further carried with B.L.I. Systematic analysis of binding with different control proteins and purified SARS-CoV-2 ensured the robustness of the antibodies.
- Published
- 2022
- Full Text
- View/download PDF
375. CellectSeq: In silico discovery of antibodies targeting integral membrane proteins combining in situ selections and next-generation sequencing
- Author
-
Abdellali Kelil, Eugenio Gallo, Sunandan Banerjee, Jarrett J. Adams, and Sachdev S. Sidhu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Kelil, Gallo et al. develop an antibody discovery strategy, termed CellectSeq, for targeting integral membrane proteins on native cells. This strategy uses motif-based scoring and sequencing error-filtering algorithms to identify antibodies with high diversity and specificity even at extremely low abundances in NGS pools.
- Published
- 2021
- Full Text
- View/download PDF
376. Inhibition of HECT E3 ligases as potential therapy for COVID-19
- Author
-
Giuseppe Novelli, Jing Liu, Michela Biancolella, Tonino Alonzi, Antonio Novelli, J. J. Patten, Dario Cocciadiferro, Emanuele Agolini, Vito Luigi Colona, Barbara Rizzacasa, Rosalinda Giannini, Benedetta Bigio, Delia Goletti, Maria Rosaria Capobianchi, Sandro Grelli, Justin Mann, Trevor D. McKee, Ke Cheng, Fatima Amanat, Florian Krammer, Andrea Guarracino, Gerardo Pepe, Carlo Tomino, Yacine Tandjaoui-Lambiotte, Yurdagul Uzunhan, Sarah Tubiana, Jade Ghosn, COVID Human Genetic Effort, French COVID Cohort Study Group, CoV-Contact Cohort, Luigi D. Notarangelo, Helen C. Su, Laurent Abel, Aurélie Cobat, Gai Elhanan, Joseph J. Grzymski, Andrea Latini, Sachdev S. Sidhu, Suresh Jain, Robert A. Davey, Jean-Laurent Casanova, Wenyi Wei, and Pier Paolo Pandolfi
- Subjects
Cytology ,QH573-671 - Abstract
Abstract SARS-CoV-2 is responsible for the ongoing world-wide pandemic which has already taken more than two million lives. Effective treatments are urgently needed. The enzymatic activity of the HECT-E3 ligase family members has been implicated in the cell egression phase of deadly RNA viruses such as Ebola through direct interaction of its VP40 Protein. Here we report that HECT-E3 ligase family members such as NEDD4 and WWP1 interact with and ubiquitylate the SARS-CoV-2 Spike protein. Furthermore, we find that HECT family members are overexpressed in primary samples derived from COVID-19 infected patients and COVID-19 mouse models. Importantly, rare germline activating variants in the NEDD4 and WWP1 genes are associated with severe COVID-19 cases. Critically, I3C, a natural NEDD4 and WWP1 inhibitor from Brassicaceae, displays potent antiviral effects and inhibits viral egression. In conclusion, we identify the HECT family members of E3 ligases as likely novel biomarkers for COVID-19, as well as new potential targets of therapeutic strategy easily testable in clinical trials in view of the established well-tolerated nature of the Brassicaceae natural compounds.
- Published
- 2021
- Full Text
- View/download PDF
377. A Quantitative Assay for Ca2+ Uptake through Normal and Pathological Hemichannels
- Author
-
Chiara Nardin, Abraham Tettey-Matey, Viola Donati, Daniela Marazziti, Chiara Di Pietro, Chiara Peres, Marcello Raspa, Francesco Zonta, Guang Yang, Maryna Gorelik, Serena Singh, Lia Cardarelli, Sachdev S. Sidhu, and Fabio Mammano
- Subjects
connexins ,genetically encoded calcium indicators ,monoclonal antibodies ,drug discovery ,genodermatoses ,cancer ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Connexin (Cx) hemichannels (HCs) are large pore hexameric structures that allow the exchange of ions, metabolites and a variety of other molecules between the cell cytoplasm and extracellular milieu. HC inhibitors are attracting growing interest as drug candidates because deregulated fluxes through HCs have been implicated in a plethora of genetic conditions and other diseases. HC activity has been mainly investigated by electrophysiological methods and/or using HC-permeable dye uptake measurements. Here, we present an all-optical assay based on fluorometric measurements of ionized calcium (Ca2+) uptake with a Ca2+-selective genetically encoded indicator (GCaMP6s) that permits the optical tracking of cytosolic Ca2+ concentration ([Ca2+]cyt) changes with high sensitivity. We exemplify use of the assay in stable pools of HaCaT cells overexpressing human Cx26, Cx46, or the pathological mutant Cx26G45E, under control of a tetracycline (Tet) responsive element (TRE) promoter (Tet-on). We demonstrate the usefulness of the assay for the characterization of new monoclonal antibodies (mAbs) targeting the extracellular domain of the HCs. Although we developed the assay on a spinning disk confocal fluorescence microscope, the same methodology can be extended seamlessly to high-throughput high-content platforms to screen other kinds of inhibitors and/or to probe HCs expressed in primary cells and microtissues.
- Published
- 2022
- Full Text
- View/download PDF
378. A T cell redirection platform for co-targeting dual antigens on solid tumors
- Author
-
Leonie Enderle, Karim H. Shalaby, Maryna Gorelik, Alexander Weiss, Levi L. Blazer, Marcin Paduch, Lia Cardarelli, Anthony Kossiakoff, Jarrett J. Adams, and Sachdev S. Sidhu
- Subjects
Antibody/immunotherapy/pancreatic cancer/solid tumors/T-cell redirection/CD133/EPHA2/EPCAM/EPHB2/dual targeting ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
In order to direct T cells to specific features of solid cancer cells, we engineered a bispecific antibody format, named Dual Antigen T cell Engager (DATE), by fusing a single-chain variable fragment targeting CD3 to a tumor-targeting antigen-binding fragment. In this format, multiple novel paratopes against different tumor antigens were able to recruit T-cell cytotoxicity to tumor cells in vitro and in an in vivo pancreatic ductal adenocarcinoma xenograft model. Since unique surface antigens in solid tumors are limited, in order to enhance selectivity, we further engineered “double-DATEs” targeting two tumor antigens simultaneously. The double-DATE contains an additional autonomous variable heavy-chain domain, which binds a second tumor antigen without itself eliciting a cytotoxic response. This novel modality provides a strategy to enhance the selectivity of immune redirection through binary targeting of native tumor antigens. The modularity and use of a common, stable human framework for all components enables a pipeline approach to rapidly develop a broad repertoire of tailored DATEs and double-DATEs with favorable biophysical properties and high potencies and selectivities.
- Published
- 2021
- Full Text
- View/download PDF
379. Two Different Therapeutic Approaches for SARS-CoV-2 in hiPSCs-Derived Lung Organoids
- Author
-
Paola Spitalieri, Federica Centofanti, Michela Murdocca, Maria Giovanna Scioli, Andrea Latini, Silvia Di Cesare, Gennaro Citro, Antonio Rossi, Augusto Orlandi, Shane Miersch, Sachdev S. Sidhu, Pier Paolo Pandolfi, Annalisa Botta, Federica Sangiuolo, and Giuseppe Novelli
- Subjects
hiPSCs ,hLORGs ,SARS-CoV2 pseudovirus ,neutralizing monoclonal antibody ,synthetic peptide ,Cytology ,QH573-671 - Abstract
The global health emergency for SARS-CoV-2 (COVID-19) created an urgent need to develop new treatments and therapeutic drugs. In this study, we tested, for the first time on human cells, a new tetravalent neutralizing antibody (15033-7) targeting Spike protein and a synthetic peptide homologous to dipeptidyl peptidase-4 (DPP4) receptor on host cells. Both could represent powerful immunotherapeutic candidates for COVID-19 treatment. The infection begins in the proximal airways, namely the alveolar type 2 (AT2) cells of the distal lung, which express both ACE2 and DPP4 receptors. Thus, to evaluate the efficacy of both approaches, we developed three-dimensional (3D) complex lung organoid structures (hLORGs) derived from human-induced pluripotent stem cells (iPSCs) and resembling the in vivo organ. Afterward, hLORGs were infected by different SARS-CoV-2 S pseudovirus variants and treated by the Ab15033-7 or DPP4 peptide. Using both approaches, we observed a significant reduction of viral entry and a modulation of the expression of genes implicated in innate immunity and inflammatory response. These data demonstrate the efficacy of such approaches in strongly reducing the infection efficiency in vitro and, importantly, provide proof-of-principle evidence that hiPSC-derived hLORGs represent an ideal in vitro system for testing both therapeutic and preventive modalities against COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
380. The RNA-Binding Protein Rasputin/G3BP Enhances the Stability and Translation of Its Target mRNAs
- Author
-
John D. Laver, Jimmy Ly, Allison K. Winn, Angelo Karaiskakis, Sichun Lin, Kun Nie, Giulia Benic, Nima Jaberi-Lashkari, Wen Xi Cao, Alireza Khademi, J. Timothy Westwood, Sachdev S. Sidhu, Quaid Morris, Stephane Angers, Craig A. Smibert, and Howard D. Lipshitz
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: G3BP RNA-binding proteins are important components of stress granules (SGs). Here, we analyze the role of the Drosophila G3BP Rasputin (RIN) in unstressed cells, where RIN is not SG associated. Immunoprecipitation followed by microarray analysis identifies over 550 mRNAs that copurify with RIN. The mRNAs found in SGs are long and translationally silent. In contrast, we find that RIN-bound mRNAs, which encode core components of the transcription, splicing, and translation machinery, are short, stable, and highly translated. We show that RIN is associated with polysomes and provide evidence for a direct role for RIN and its human homologs in stabilizing and upregulating the translation of their target mRNAs. We propose that when cells are stressed, the resulting incorporation of RIN/G3BPs into SGs sequesters them away from their short target mRNAs. This would downregulate the expression of these transcripts, even though they are not incorporated into stress granules. : Laver et al. show that in early embryos, Rasputin, the Drosophila G3BP ortholog, binds short mRNAs and is associated with polysomes. They provide evidence for a direct role for Rasputin and its human homologs in stabilizing and upregulating the translation of their target mRNAs. Keywords: RNA-binding protein, post-transcriptional regulation, mRNA translation, mRNA stability, stress granule, Drosophila, embryo, G3BP, Rasputin
- Published
- 2020
- Full Text
- View/download PDF
381. In situ antibody phage display yields optimal inhibitors of integrin α11/β1
- Author
-
Eugenio Gallo, Abdellali Kelil, Peter E. Bayliss, Ajitha Jeganathan, Olga Egorova, Lynda Ploder, Jarrett J. Adams, Patricia Giblin, and Sachdev S. Sidhu
- Subjects
Enhanced diversity ,antibody selections ,cellular selections ,ITGA11 ,integrin-α11/β1 receptor ,cancer therapeutics ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Integrins are transmembrane multi-conformation receptors that mediate interactions with the extracellular matrix. In cancer, integrins influence metastasis, proliferation, and survival. Collagen-binding integrin-α11/β1, a marker of aggressive tumors that is involved in stroma-tumor crosstalk, may be an attractive target for anti-cancer therapeutic antibodies. We performed selections with phage-displayed synthetic antibody libraries for binding to either purified integrin-α11/β1 or in situ on live cells. The in-situ strategy yielded many diverse antibodies, and strikingly, most of these antibodies did not recognize purified integrin-α11/β1. Conversely, none of the antibodies selected for binding to purified integrin-α11/β1 were able to efficiently recognize native cell-surface antigen. Most importantly, only the in-situ selection yielded functional antibodies that were able to compete with collagen-I for binding to cell-surface integrin-α11/β1, and thus inhibited cell adhesion. In-depth characterization of a subset of in situ-derived clones as full-length immunoglobulins revealed high affinity cellular binding and inhibitory activities in the single-digit nanomolar range. Moreover, the antibodies showed high selectivity for integrin-α11/β1 with minimal cross-reactivity for close homologs. Taken together, our findings highlight the advantages of in-situ selections for generation of anti-integrin antibodies optimized for recognition and inhibition of native cell-surface proteins, and our work establishes general methods that could be extended to many other membrane proteins.
- Published
- 2020
- Full Text
- View/download PDF
382. The Deleterious Effects of Shiga Toxin Type 2 Are Neutralized In Vitro by FabF8:Stx2 Recombinant Monoclonal Antibody
- Author
-
Daniela Luz, Fernando D. Gómez, Raíssa L. Ferreira, Bruna S. Melo, Beatriz E. C. Guth, Wagner Quintilio, Ana Maria Moro, Agostina Presta, Flavia Sacerdoti, Cristina Ibarra, Gang Chen, Sachdev S. Sidhu, María Marta Amaral, and Roxane M. F. Piazza
- Subjects
STEC ,Stx2 ,antibody fragment ,monoclonal antibody ,Medicine - Abstract
Hemolytic Uremic Syndrome (HUS) associated with Shiga-toxigenic Escherichia coli (STEC) infections is the principal cause of acute renal injury in pediatric age groups. Shiga toxin type 2 (Stx2) has in vitro cytotoxic effects on kidney cells, including human glomerular endothelial (HGEC) and Vero cells. Neither a licensed vaccine nor effective therapy for HUS is available for humans. Recombinant antibodies against Stx2, produced in bacteria, appeared as the utmost tool to prevent HUS. Therefore, in this work, a recombinant FabF8:Stx2 was selected from a human Fab antibody library by phage display, characterized, and analyzed for its ability to neutralize the Stx activity from different STEC-Stx2 and Stx1/Stx2 producing strains in a gold standard Vero cell assay, and the Stx2 cytotoxic effects on primary cultures of HGEC. This recombinant Fab showed a dissociation constant of 13.8 nM and a half maximum effective concentration (EC50) of 160 ng/mL to Stx2. Additionally, FabF8:Stx2 neutralized, in different percentages, the cytotoxic effects of Stx2 and Stx1/2 from different STEC strains on Vero cells. Moreover, it significantly prevented the deleterious effects of Stx2 in a dose-dependent manner (up to 83%) in HGEC and protected this cell up to 90% from apoptosis and necrosis. Therefore, this novel and simple anti-Stx2 biomolecule will allow further investigation as a new therapeutic option that could improve STEC and HUS patient outcomes.
- Published
- 2021
- Full Text
- View/download PDF
383. Peptide Platform as a Powerful Tool in the Fight against COVID-19
- Author
-
Michela Murdocca, Gennaro Citro, Isabella Romeo, Antonio Lupia, Shane Miersch, Bruno Amadio, Alessia Bonomo, Antonio Rossi, Sachdev S. Sidhu, Pier Paolo Pandolfi, Stefano Alcaro, Federica Carla Sangiuolo, and Giuseppe Novelli
- Subjects
COVID-19 ,peptides ,protein–protein docking ,conserved RBD region ,SARS-CoV-2 variants ,VSVpp.SARS-2S ,Microbiology ,QR1-502 - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a global pandemic causing over 195 million infections and more than 4 million fatalities as of July 2021.To date, it has been demonstrated that a number of mutations in the spike glycoprotein (S protein) of SARS-CoV-2 variants of concern abrogate or reduce the neutralization potency of several therapeutic antibodies and vaccine-elicited antibodies. Therefore, the development of additional vaccine platforms with improved supply and logistic profile remains a pressing need. In this work, we have validated the applicability of a peptide-based strategy focused on a preventive as well as a therapeutic purpose. On the basis of the involvement of the dipeptidyl peptidase 4 (DPP4), in addition to the angiotensin converting enzyme 2 (ACE2) receptor in the mechanism of virus entry, we analyzed peptides bearing DPP4 sequences by protein–protein docking and assessed their ability to block pseudovirus infection in vitro. In parallel, we have selected and synthetized peptide sequences located within the highly conserved receptor-binding domain (RBD) of the S protein, and we found that RBD-based vaccines could better promote elicitation of high titers of neutralizing antibodies specific against the regions of interest, as confirmed by immunoinformatic methodologies and in vivo studies. These findings unveil a key antigenic site targeted by broadly neutralizing antibodies and pave the way to the design of pan-coronavirus vaccines.
- Published
- 2021
- Full Text
- View/download PDF
384. The influence of microRNAs and poly(A) tail length on endogenous mRNA–protein complexes
- Author
-
Olivia S. Rissland, Alexander O. Subtelny, Miranda Wang, Andrew Lugowski, Beth Nicholson, John D. Laver, Sachdev S. Sidhu, Craig A. Smibert, Howard D. Lipshitz, and David P. Bartel
- Subjects
MicroRNAs ,Poly(A) tail ,mRNA–protein complexes ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background All mRNAs are bound in vivo by proteins to form mRNA–protein complexes (mRNPs), but changes in the composition of mRNPs during posttranscriptional regulation remain largely unexplored. Here, we have analyzed, on a transcriptome-wide scale, how microRNA-mediated repression modulates the associations of the core mRNP components eIF4E, eIF4G, and PABP and of the decay factor DDX6 in human cells. Results Despite the transient nature of repressed intermediates, we detect significant changes in mRNP composition, marked by dissociation of eIF4G and PABP, and by recruitment of DDX6. Furthermore, although poly(A)-tail length has been considered critical in post-transcriptional regulation, differences in steady-state tail length explain little of the variation in either PABP association or mRNP organization more generally. Instead, relative occupancy of core components correlates best with gene expression. Conclusions These results indicate that posttranscriptional regulatory factors, such as microRNAs, influence the associations of PABP and other core factors, and do so without substantially affecting steady-state tail length.
- Published
- 2017
- Full Text
- View/download PDF
385. Blockade of TGF-β signaling with novel synthetic antibodies limits immune exclusion and improves chemotherapy response in metastatic ovarian cancer models
- Author
-
Daniel Newsted, Sunandan Banerjee, Kathleen Watt, Sarah Nersesian, Peter Truesdell, Levi L. Blazer, Lia Cardarelli, Jarrett J. Adams, Sachdev S. Sidhu, and Andrew W. Craig
- Subjects
ovarian cancer ,tgf-β ,emt ,immune exclusion ,synthetic antibodies ,combination therapies ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Epithelial ovarian cancer (EOC) is a leading cause of cancer-related death in women. EOC is often diagnosed at late stages, with peritoneal metastases and ascites production. Current surgery and platinum-based chemotherapy regimes fail to prevent recurrence in most patients. High levels of Transforming growth factor-β (TGF-β) within ascites has been linked to poor prognosis. TGF-β signaling promotes epithelial-mesenchymal transition (EMT) in EOC tumor cells, and immune suppression within the tumor microenvironment, with both contributing to chemotherapy resistance and metastasis. The goal of this study was to develop specific synthetic inhibitory antibodies to the Type II TGF-β receptor (TGFBR2), and test these antibodies in EOC cell and tumor models. Following screening of a phage-displayed synthetic antigen-binding fragment (Fab) library with the extracellular domain of TGFBR2, we identified a lead inhibitory Fab that suppressed TGF-β signaling in mouse and human EOC cell lines. Affinity maturation of the lead inhibitory Fab resulted in several derivative Fabs with increased affinity for TGFBR2 and efficacy as suppressors of TGF-β signaling, EMT and EOC cell invasion. In EOC xenograft and syngeneic tumor models, blockade of TGFBR2 with our lead antibodies led to improved chemotherapy response. This correlated with reversal of EMT and immune exclusion in these tumor models with TGFBR2 blockade. Together, these results describe new inhibitors of the TGF-β pathway that improve antitumor immunity, and response to chemotherapy in preclinical EOC models.
- Published
- 2019
- Full Text
- View/download PDF
386. Biosynthetic Oligoclonal Antivenom (BOA) for Snakebite and Next-Generation Treatments for Snakebite Victims
- Author
-
R. Manjunatha Kini, Sachdev S. Sidhu, and Andreas Hougaard Laustsen
- Subjects
snakebite envenoming ,neglected tropical diseases ,antivenom ,next-generation antivenom ,recombinant antivenom ,small molecule inhibitors ,Medicine - Abstract
Snakebite envenoming is a neglected tropical disease that each year claims the lives of 80,000⁻140,000 victims worldwide. The only effective treatment against envenoming involves intravenous administration of antivenoms that comprise antibodies that have been isolated from the plasma of immunized animals, typically horses. The drawbacks of such conventional horse-derived antivenoms include their propensity for causing allergenic adverse reactions due to their heterologous and foreign nature, an inability to effectively neutralize toxins in distal tissue, a low content of toxin-neutralizing antibodies, and a complex manufacturing process that is dependent on husbandry and procurement of snake venoms. In recent years, an opportunity to develop a fundamentally novel type of antivenom has presented itself. By using modern antibody discovery strategies, such as phage display selection, and repurposing small molecule enzyme inhibitors, next-generation antivenoms that obviate the drawbacks of existing plasma-derived antivenoms could be developed. This article describes the conceptualization of a novel therapeutic development strategy for biosynthetic oligoclonal antivenom (BOA) for snakebites based on recombinantly expressed oligoclonal mixtures of human monoclonal antibodies, possibly combined with repurposed small molecule enzyme inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
387. Host Protein BAG3 is a Negative Regulator of Lassa VLP Egress
- Author
-
Ziying Han, Michael P. Schwoerer, Philip Hicks, Jingjing Liang, Gordon Ruthel, Corbett T. Berry, Bruce D. Freedman, Cari A. Sagum, Mark T. Bedford, Sachdev S. Sidhu, Marius Sudol, and Ronald N. Harty
- Subjects
Lassa fever virus ,VLPs ,budding ,BAG3 ,L-domain ,virus-host interaction ,Ebola ,WW-domain ,autophagy ,Medicine - Abstract
Lassa fever virus (LFV) belongs to the Arenaviridae family and can cause acute hemorrhagic fever in humans. The LFV Z protein plays a central role in virion assembly and egress, such that independent expression of LFV Z leads to the production of virus-like particles (VLPs) that mimic egress of infectious virus. LFV Z contains both PTAP and PPPY L-domain motifs that are known to recruit host proteins that are important for mediating efficient virus egress and spread. The viral PPPY motif is known to interact with specific host WW-domain bearing proteins. Here we identified host WW-domain bearing protein BCL2 Associated Athanogene 3 (BAG3) as a LFV Z PPPY interactor using our proline-rich reading array of WW-domain containing mammalian proteins. BAG3 is a stress-induced molecular co-chaperone that functions to regulate cellular protein homeostasis and cell survival via Chaperone-Assisted Selective Autophagy (CASA). Similar to our previously published findings for the VP40 proteins of Ebola and Marburg viruses, our results using VLP budding assays, BAG3 knockout cells, and confocal microscopy indicate that BAG3 is a WW-domain interactor that negatively regulates egress of LFV Z VLPs, rather than promoting VLP release. Our results suggest that CASA and specifically BAG3 may represent a novel host defense mechanism, whereby BAG3 may dampen egress of several hemorrhagic fever viruses by interacting and interfering with the budding function of viral PPxY-containing matrix proteins.
- Published
- 2018
- Full Text
- View/download PDF
388. Structural Changes in Stx1 Engineering Monoclonal Antibody Improves Its Functionality as Diagnostic Tool for a Rapid Latex Agglutination Test
- Author
-
Daniela Luz, Emerson A. Shiga, Gang Chen, Wagner Quintilio, Fernanda B. Andrade, Andrea Q. Maranhão, Bruna A. Caetano, Thaís Mitsunari, Míriam A. Silva, Letícia B. Rocha, Ana M. Moro, Sachdev S. Sidhu, and Roxane M. F. Piazza
- Subjects
antibody ,scFv ,Stx1 ,STEC ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Stx1 toxin is one of the AB5 toxins of Shiga toxin-producing Escherichia coli (STEC) responsible for foodborne intoxication during outbreaks. The single-chain variable fragment (scFv) is the most common recombinant antibody format; it consists of both variable chains connected by a peptide linker with conserved specificity and affinity for antigen. The drawbacks of scFv production in bacteria are the heterologous expression, conformation and stability of the molecule, which could change the affinity for the antigen. In this work, we obtained a stable and functional scFv-Stx1 in bacteria, starting from IgG produced by hybridoma cells. After structural modifications, i.e., change in protein orientation, vector and linker, its solubility for expression in bacteria was increased as well as the affinity for its antigen, demonstrated by a scFv dissociation constant (KD) of 2.26 × 10−7 M. Also, it was able to recognize purified Stx1 and cross-reacted with Stx2 toxin by ELISA (Enzyme-Linked Immunosorbent Assay), and detected 88% of Stx1-producing strains using a rapid latex agglutination test. Thus, the scFv fragment obtained in the present work is a bacteria-produced tool for use in a rapid diagnosis test, providing an alternative for STEC diagnosis.
- Published
- 2018
- Full Text
- View/download PDF
389. Antigenic mapping reveals sites of vulnerability on α-HCoV spike protein.
- Author
-
Xiang J, Su J, Lan Q, Zhao W, Zhou Y, Xu Y, Niu J, Xia S, Qi Q, Sidhu S, Lu L, Miersch S, and Yang B
- Subjects
- Humans, Spike Glycoprotein, Coronavirus genetics, SARS-CoV-2, Antigens, Viral, Epitopes, Antibodies, Neutralizing, COVID-19, Coronavirus 229E, Human
- Abstract
Understanding the antigenic signatures of all human coronaviruses (HCoVs) Spike (S) proteins is imperative for pan-HCoV epitopes identification and broadly effective vaccine development. To depict the currently elusive antigenic signatures of α-HCoVs S proteins, we isolated a panel of antibodies against the HCoV-229E S protein and characterized their epitopes and neutralizing potential. We found that the N-terminal domain of HCoV-229E S protein is antigenically dominant wherein an antigenic supersite is present and appears conserved in HCoV-NL63, which holds potential to serve as a pan-α-HCoVs epitope. In the receptor binding domain, a neutralizing epitope is captured in the end distal to the receptor binding site, reminiscent of the locations of the SARS-CoV-2 RBD cryptic epitopes. We also identified a neutralizing antibody that recognizes the connector domain, thus representing the first S2-directed neutralizing antibody against α-HCoVs. The unraveled HCoVs S proteins antigenic similarities and variances among genera highlight the challenges faced by pan-HCoV vaccine design while supporting the feasibility of broadly effective vaccine development against a subset of HCoVs., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
390. Rapid On-Cell Selection of High-Performance Human Antibodies.
- Author
-
Philpott DN, Gomis S, Wang H, Atwal R, Kelil A, Sack T, Morningstar B, Burnie C, Sargent EH, Angers S, Sidhu S, and Kelley SO
- Abstract
Phage display is a critical tool for developing antibodies. However, existing approaches require many time-consuming rounds of biopanning and screening of potential candidates due to a high rate of failure during validation. Herein, we present a rapid on-cell phage display platform which recapitulates the complex in vivo binding environment to produce high-performance human antibodies in a short amount of time. Selection is performed in a highly stringent heterogeneous mixture of cells to quickly remove nonspecific binders. A microfluidic platform then separates antigen-presenting cells with high throughput and specificity. An unsupervised machine learning algorithm analyzes sequences of phage from all pools to identify the structural trends that contribute to affinity and proposes ideal candidates for validation. In a proof-of-concept screen against human Frizzled-7, a key ligand in the Wnt signaling pathway, antibodies with picomolar affinity were discovered in two rounds of selection that outperformed current gold-standard reagents. This approach, termed μCellect, is low cost, high throughput, and compatible with a wide variety of cell types, enabling widespread adoption for antibody development., Competing Interests: The authors declare no competing financial interest., (© 2021 The Authors. Published by American Chemical Society.)
- Published
- 2022
- Full Text
- View/download PDF
391. Human ACE2 receptor polymorphisms and altered susceptibility to SARS-CoV-2.
- Author
-
Suryamohan K, Diwanji D, Stawiski EW, Gupta R, Miersch S, Liu J, Chen C, Jiang YP, Fellouse FA, Sathirapongsasuti JF, Albers PK, Deepak T, Saberianfar R, Ratan A, Washburn G, Mis M, Santhosh D, Somasekar S, Hiranjith GH, Vargas D, Mohan S, Phalke S, Kuriakose B, Antony A, Ustav M Jr, Schuster SC, Sidhu S, Junutula JR, Jura N, and Seshagiri S
- Subjects
- Amino Acid Sequence, Angiotensin-Converting Enzyme 2 chemistry, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 metabolism, COVID-19 virology, Host-Pathogen Interactions, Humans, Models, Molecular, Protein Binding, Protein Domains, Receptors, Virus chemistry, Receptors, Virus metabolism, SARS-CoV-2 metabolism, SARS-CoV-2 physiology, Sequence Homology, Amino Acid, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus metabolism, Virus Internalization, Angiotensin-Converting Enzyme 2 genetics, COVID-19 genetics, Genetic Predisposition to Disease genetics, Mutation, Missense genetics, Polymorphism, Genetic, Receptors, Virus genetics
- Abstract
COVID-19 is a respiratory illness caused by a novel coronavirus called SARS-CoV-2. The viral spike (S) protein engages the human angiotensin-converting enzyme 2 (ACE2) receptor to invade host cells with ~10-15-fold higher affinity compared to SARS-CoV S-protein, making it highly infectious. Here, we assessed if ACE2 polymorphisms can alter host susceptibility to SARS-CoV-2 by affecting this interaction. We analyzed over 290,000 samples representing >400 population groups from public genomic datasets and identified multiple ACE2 protein-altering variants. Using reported structural data, we identified natural ACE2 variants that could potentially affect virus-host interaction and thereby alter host susceptibility. These include variants S19P, I21V, E23K, K26R, T27A, N64K, T92I, Q102P and H378R that were predicted to increase susceptibility, while variants K31R, N33I, H34R, E35K, E37K, D38V, Y50F, N51S, M62V, K68E, F72V, Y83H, G326E, G352V, D355N, Q388L and D509Y were predicted to be protective variants that show decreased binding to S-protein. Using biochemical assays, we confirmed that K31R and E37K had decreased affinity, and K26R and T92I variants showed increased affinity for S-protein when compared to wildtype ACE2. Consistent with this, soluble ACE2 K26R and T92I were more effective in blocking entry of S-protein pseudotyped virus suggesting that ACE2 variants can modulate susceptibility to SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
392. Neutralizing Antibody and Soluble ACE2 Inhibition of a Replication-Competent VSV-SARS-CoV-2 and a Clinical Isolate of SARS-CoV-2.
- Author
-
Case JB, Rothlauf PW, Chen RE, Liu Z, Zhao H, Kim AS, Bloyet LM, Zeng Q, Tahan S, Droit L, Ilagan MXG, Tartell MA, Amarasinghe G, Henderson JP, Miersch S, Ustav M, Sidhu S, Virgin HW, Wang D, Ding S, Corti D, Theel ES, Fremont DH, Diamond MS, and Whelan SPJ
- Subjects
- Angiotensin-Converting Enzyme 2, Animals, Betacoronavirus genetics, Betacoronavirus physiology, COVID-19, Chlorocebus aethiops, Coronavirus Infections genetics, Coronavirus Infections immunology, Coronavirus Infections virology, Green Fluorescent Proteins genetics, Host Microbial Interactions immunology, Humans, Immunization, Passive, Neutralization Tests, Pandemics, Pneumonia, Viral immunology, Pneumonia, Viral virology, SARS-CoV-2, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vero Cells, Vesicular stomatitis Indiana virus genetics, Vesicular stomatitis Indiana virus immunology, Virus Internalization, Virus Replication, COVID-19 Serotherapy, Antibodies, Neutralizing blood, Antibodies, Viral blood, Betacoronavirus immunology, Coronavirus Infections therapy, Peptidyl-Dipeptidase A immunology, Pneumonia, Viral therapy
- Abstract
Antibody-based interventions against SARS-CoV-2 could limit morbidity, mortality, and possibly transmission. An anticipated correlate of such countermeasures is the level of neutralizing antibodies against the SARS-CoV-2 spike protein, which engages with host ACE2 receptor for entry. Using an infectious molecular clone of vesicular stomatitis virus (VSV) expressing eGFP as a marker of infection, we replaced the glycoprotein gene (G) with the spike protein of SARS-CoV-2 (VSV-eGFP-SARS-CoV-2) and developed a high-throughput-imaging-based neutralization assay at biosafety level 2. We also developed a focus-reduction neutralization test with a clinical isolate of SARS-CoV-2 at biosafety level 3. Comparing the neutralizing activities of various antibodies and ACE2-Fc soluble decoy protein in both assays revealed a high degree of concordance. These assays will help define correlates of protection for antibody-based countermeasures and vaccines against SARS-CoV-2. Additionally, replication-competent VSV-eGFP-SARS-CoV-2 provides a tool for testing inhibitors of SARS-CoV-2 mediated entry under reduced biosafety containment., Competing Interests: Declaration of Interests M.S.D. is a consultant for Inbios, Eli Lilly, Vir Biotechnology, and NGM Biopharmaceuticals and is on the Scientific Advisory Board of Moderna. The Diamond laboratory has received unrelated funding under sponsored research agreements from Moderna and Emergent BioSolutions. D.C. and H.W.V. are employees of Vir Biotechnology Inc. and may hold shares in Vir Biotechnology Inc. S.P.J.W. and P.W.R. have filed a disclosure with Washington University for the recombinant VSV., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
393. Surface Loops in a Single SH2 Domain Are Capable of Encoding the Spectrum of Specificity of the SH2 Family.
- Author
-
Liu H, Huang H, Voss C, Kaneko T, Qin WT, Sidhu S, and Li SS
- Subjects
- Animals, Crystallography, X-Ray, Genetic Variation, Humans, Ligands, Models, Molecular, Peptide Library, Protein Structure, Secondary, Proto-Oncogene Proteins c-fyn genetics, src Homology Domains, Proto-Oncogene Proteins c-fyn chemistry
- Abstract
Src homology 2 (SH2) domains play an essential role in cellular signal transduction by binding to proteins phosphorylated on Tyr residue. Although Tyr phosphorylation (pY) is a prerequisite for binding for essentially all SH2 domains characterized to date, different SH2 domains prefer specific sequence motifs C-terminal to the pY residue. Because all SH2 domains adopt the same structural fold, it is not well understood how different SH2 domains have acquired the ability to recognize distinct sequence motifs. We have shown previously that the EF and BG loops that connect the secondary structure elements on an SH2 domain dictate its specificity. In this study, we investigated if these surface loops could be engineered to encode diverse specificities. By characterizing a group of SH2 variants selected by different pY peptides from phage-displayed libraries, we show that the EF and BG loops of the Fyn SH2 domain can encode a wide spectrum of specificities, including all three major specificity classes ( p + 2, p + 3 and p + 4) of the SH2 domain family. Furthermore, we found that the specificity of a given variant correlates with the sequence feature of the bait peptide used for its isolation, suggesting that an SH2 domain may acquire specificity by co-evolving with its ligand. Intriguingly, we found that the SH2 variants can employ a variety of different mechanisms to confer the same specificity, suggesting the EF and BG loops are highly flexible and adaptable. Our work provides a plausible mechanism for the SH2 domain to acquire the wide spectrum of specificity observed in nature through loop variation with minimal disturbance to the SH2 fold. It is likely that similar mechanisms may have been employed by other modular interaction domains to generate diversity in specificity., (© 2019 Liu et al.)
- Published
- 2019
- Full Text
- View/download PDF
394. Cotargeting Ephrin Receptor Tyrosine Kinases A2 and A3 in Cancer Stem Cells Reduces Growth of Recurrent Glioblastoma.
- Author
-
Qazi MA, Vora P, Venugopal C, Adams J, Singh M, Hu A, Gorelik M, Subapanditha MK, Savage N, Yang J, Chokshi C, London M, Gont A, Bobrowski D, Grinshtein N, Brown KR, Murty NK, Nilvebrant J, Kaplan D, Moffat J, Sidhu S, and Singh SK
- Subjects
- Animals, Biomarkers, Tumor genetics, Carcinogenesis genetics, Cell Differentiation genetics, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Ephrin-A2 antagonists & inhibitors, Gene Expression Regulation, Neoplastic genetics, Gene Knockdown Techniques, Glioblastoma drug therapy, Glioblastoma pathology, Glioblastoma radiotherapy, Humans, Mice, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local radiotherapy, Neoplastic Stem Cells pathology, Prognosis, Radiation, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor, EphA3, Receptors, Eph Family antagonists & inhibitors, Receptors, Eph Family genetics, Temozolomide pharmacology, Xenograft Model Antitumor Assays, Ephrin-A2 genetics, Glioblastoma genetics, Neoplasm Recurrence, Local genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM. Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
395. Corrigendum: Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors.
- Author
-
Steinhart Z, Pavlovic Z, Chandrashekhar M, Hart T, Wang X, Zhang X, Robitaille M, Brown KR, Jaksani S, Overmeer R, Boj SF, Adams J, Pan J, Clevers H, Sidhu S, Moffat J, and Angers S
- Abstract
This corrects the article DOI: 10.1038/nm.4219.
- Published
- 2017
- Full Text
- View/download PDF
396. Syp1 regulates the clathrin-mediated and clathrin-independent endocytosis of multiple cargo proteins through a novel sorting motif.
- Author
-
Apel AR, Hoban K, Chuartzman S, Tonikian R, Sidhu S, Schuldiner M, Wendland B, and Prosser D
- Subjects
- Adaptor Proteins, Vesicular Transport metabolism, Biological Transport, Carrier Proteins genetics, Cell Membrane metabolism, Clathrin metabolism, Endocytosis physiology, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Plasmids, Protein Transport, SNARE Proteins metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Carrier Proteins metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Internalization of proteins from the plasma membrane (PM) allows for cell-surface composition regulation, signaling of network modulation, and nutrient uptake. Clathrin-mediated endocytosis (CME) is a major internalization route for PM proteins. During CME, endocytic adaptor proteins bind cargoes at the cell surface and link them to the PM and clathrin coat. Muniscins are a conserved family of endocytic adaptors, including Syp1 in budding yeast and its mammalian orthologue, FCHo1. These adaptors bind cargo via a C-terminal μ -homology domain ( μ HD); however, few cargoes exhibiting muniscin-dependent endocytosis have been identified, and the sorting sequence recognized by the µ HD is unknown. To reveal Syp1 cargo-sorting motifs, we performed a phage display screen and used biochemical methods to demonstrate that the Syp1 µ HD binds DxY motifs in the previously identified Syp1 cargo Mid2 and the v-SNARE Snc1. We also executed an unbiased visual screen, which identified the peptide transporter Ptr2 and the ammonium permease Mep3 as Syp1 cargoes containing DxY motifs. Finally, we determined that, in addition to regulating cargo entry through CME, Syp1 can promote internalization of Ptr2 through a recently identified clathrin-independent endocytic pathway that requires the Rho1 GTPase. These findings elucidate the mechanism of Syp1 cargo recognition and its role in trafficking., (© 2017 Apel, Hoban, et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2017
- Full Text
- View/download PDF
397. Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors.
- Author
-
Steinhart Z, Pavlovic Z, Chandrashekhar M, Hart T, Wang X, Zhang X, Robitaille M, Brown KR, Jaksani S, Overmeer R, Boj SF, Adams J, Pan J, Clevers H, Sidhu S, Moffat J, and Angers S
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Animals, Carcinoma, Pancreatic Ductal metabolism, Cell Line, Tumor, Clustered Regularly Interspaced Short Palindromic Repeats, Colorectal Neoplasms metabolism, Flow Cytometry, Fluorescent Antibody Technique, Frizzled Receptors metabolism, Humans, Mice, Mice, SCID, Molecular Targeted Therapy, Neoplasm Transplantation, Organoids drug effects, Organoids metabolism, Pancreatic Neoplasms metabolism, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Ubiquitin-Protein Ligases, Wnt Signaling Pathway genetics, Antibodies pharmacology, Carcinoma, Pancreatic Ductal genetics, Cell Cycle Checkpoints drug effects, Colorectal Neoplasms genetics, DNA-Binding Proteins genetics, Frizzled Receptors antagonists & inhibitors, Oncogene Proteins genetics, Pancreatic Neoplasms genetics, Wnt Signaling Pathway drug effects
- Abstract
Forward genetic screens with CRISPR-Cas9 genome editing enable high-resolution detection of genetic vulnerabilities in cancer cells. We conducted genome-wide CRISPR-Cas9 screens in RNF43-mutant pancreatic ductal adenocarcinoma (PDAC) cells, which rely on Wnt signaling for proliferation. Through these screens, we discovered a unique requirement for a Wnt signaling circuit: engaging FZD5, one of the ten Frizzled receptors encoded in the human genome. Our results uncover an underappreciated level of context-dependent specificity at the Wnt receptor level. We further derived a panel of recombinant antibodies that reports the expression of nine FZD proteins and confirms that FZD5 functional specificity cannot be explained by protein expression patterns. Additionally, antibodies that specifically bind FZD5 and FZD8 robustly inhibited the growth of RNF43-mutant PDAC cells grown in vitro and as xenografts in vivo, providing orthogonal support for the functional specificity observed genetically. Proliferation of a patient-derived PDAC cell line harboring an RNF43 variant was also selectively inhibited by the FZD5 antibodies, further demonstrating their use as a potential targeted therapy. Tumor organoid cultures from colorectal carcinoma patients that carried RNF43 mutations were also sensitive to the FZD5 antibodies, highlighting the potential generalizability of these findings beyond PDAC. Our results show that CRIPSR-based genetic screens can be leveraged to identify and validate cell surface targets for antibody development and therapy.
- Published
- 2017
- Full Text
- View/download PDF
398. High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities.
- Author
-
Hart T, Chandrashekhar M, Aregger M, Steinhart Z, Brown KR, MacLeod G, Mis M, Zimmermann M, Fradet-Turcotte A, Sun S, Mero P, Dirks P, Sidhu S, Roth FP, Rissland OS, Durocher D, Angers S, and Moffat J
- Subjects
- Bayes Theorem, CRISPR-Cas Systems, Cell Line, Tumor, Gene Knockout Techniques, Gene Library, Humans, Mutation, Genes, Essential
- Abstract
The ability to perturb genes in human cells is crucial for elucidating gene function and holds great potential for finding therapeutic targets for diseases such as cancer. To extend the catalog of human core and context-dependent fitness genes, we have developed a high-complexity second-generation genome-scale CRISPR-Cas9 gRNA library and applied it to fitness screens in five human cell lines. Using an improved Bayesian analytical approach, we consistently discover 5-fold more fitness genes than were previously observed. We present a list of 1,580 human core fitness genes and describe their general properties. Moreover, we demonstrate that context-dependent fitness genes accurately recapitulate pathway-specific genetic vulnerabilities induced by known oncogenes and reveal cell-type-specific dependencies for specific receptor tyrosine kinases, even in oncogenic KRAS backgrounds. Thus, rigorous identification of human cell line fitness genes using a high-complexity CRISPR-Cas9 library affords a high-resolution view of the genetic vulnerabilities of a cell., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
399. A High Through-put Platform for Recombinant Antibodies to Folded Proteins.
- Author
-
Hornsby M, Paduch M, Miersch S, Sääf A, Matsuguchi T, Lee B, Wypisniak K, Doak A, King D, Usatyuk S, Perry K, Lu V, Thomas W, Luke J, Goodman J, Hoey RJ, Lai D, Griffin C, Li Z, Vizeacoumar FJ, Dong D, Campbell E, Anderson S, Zhong N, Gräslund S, Koide S, Moffat J, Sidhu S, Kossiakoff A, and Wells J
- Subjects
- Antigens genetics, Antigens immunology, Escherichia coli genetics, High-Throughput Screening Assays, Protein Folding, RNA, Small Interfering genetics, Recombinant Proteins genetics, Recombinant Proteins immunology, Antibodies genetics, Antibodies immunology, Immunoglobulin Fab Fragments genetics, Immunoglobulin Fab Fragments immunology, Transcription Factors genetics, Transcription Factors immunology
- Abstract
Antibodies are key reagents in biology and medicine, but commercial sources are rarely recombinant and thus do not provide a permanent and renewable resource. Here, we describe an industrialized platform to generate antigens and validated recombinant antibodies for 346 transcription factors (TFs) and 211 epigenetic antigens. We describe an optimized automated phage display and antigen expression pipeline that in aggregate produced about 3000 sequenced Fragment antigen-binding domain that had high affinity (typically EC50<20 nm), high stability (Tm∼80 °C), good expression in E. coli (∼5 mg/L), and ability to bind antigen in complex cell lysates. We evaluated a subset of Fabs generated to homologous SCAN domains for binding specificities. These Fragment antigen-binding domains were monospecific to their target SCAN antigen except in rare cases where they cross-reacted with a few highly related antigens. Remarkably, immunofluorescence experiments in six cell lines for 270 of the TF antigens, each having multiple antibodies, show that ∼70% stain predominantly in the cytosol and ∼20% stain in the nucleus which reinforces the dominant role that translocation plays in TF biology. These cloned antibody reagents are being made available to the academic community through our web site recombinant-antibodies.org to allow a more system-wide analysis of TF and chromatin biology. We believe these platforms, infrastructure, and automated approaches will facilitate the next generation of renewable antibody reagents to the human proteome in the coming decade., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
400. Development and characterization of recombinant antibody fragments that recognize and neutralize in vitro Stx2 toxin from Shiga toxin-producing Escherichia coli.
- Author
-
Luz D, Chen G, Maranhão AQ, Rocha LB, Sidhu S, and Piazza RM
- Subjects
- Animals, Cell Line, Humans, Hybridomas immunology, Mice, Peptide Library, Sensitivity and Specificity, Shiga-Toxigenic Escherichia coli immunology, Single-Chain Antibodies genetics, Antibodies, Neutralizing metabolism, Immunoglobulin Fab Fragments metabolism, Shiga Toxin 2 antagonists & inhibitors, Shiga-Toxigenic Escherichia coli metabolism, Single-Chain Antibodies metabolism
- Abstract
Background: Stx toxin is a member of the AB5 family of bacterial toxins: the active A subunit has N-glycosidase activity against 28S rRNA, resulting in inhibition of protein synthesis in eukaryotic cells, and the pentamer ligand B subunits (StxB) bind to globotria(tetra)osylceramide receptors (Gb3/Gb4) on the cell membrane. Shiga toxin-producing Escherichia coli strains (STEC) may produce Stx1 and/or Stx2 and variants. Strains carrying Stx2 are considered more virulent and related to the majority of outbreaks, besides being usually associated with hemolytic uremic syndrome in humans. The development of tools for the detection and/or neutralization of these toxins is a turning point for early diagnosis and therapeutics. Antibodies are an excellent paradigm for the design of high-affinity, protein-based binding reagents used for these purposes., Methods and Findings: In this work, we developed two recombinant antibodies; scFv fragments from mouse hybridomas and Fab fragments by phage display technology using a human synthetic antibody library. Both fragments showed high binding affinity to Stx2, and they were able to bind specifically to the GKIEFSKYNEDDTF region of the Stx2 B subunit and to neutralize in vitro the cytotoxicity of the toxin up to 80%. Furthermore, the scFv fragments showed 79% sensitivity and 100% specificity in detecting STEC strains by ELISA., Conclusion: In this work, we developed and characterized two recombinant antibodies against Stx2, as promising tools to be used in diagnosis or therapeutic approaches against STEC, and for the first time, we showed a human monovalent molecule, produced in bacteria, able to neutralize the cytotoxicity of Stx2 in vitro.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.