Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
MAbs ; 15(1): 2195517, 2023.
Article in English | MEDLINE | ID: mdl-37074212

ABSTRACT

Single-chain fragment variable (scFv) domains play an important role in antibody-based therapeutic modalities, such as bispecifics, multispecifics and chimeric antigen receptor T cells or natural killer cells. However, scFv domains exhibit lower stability and increased risk of aggregation due to transient dissociation ("breathing") and inter-molecular reassociation of the two domains (VL and VH). We designed a novel strategy, referred to as stapling, that introduces two disulfide bonds between the scFv linker and the two variable domains to minimize scFv breathing. We named the resulting molecules stapled scFv (spFv). Stapling increased thermal stability (Tm) by an average of 10°C. In multiple scFv/spFv multispecifics, the spFv molecules display significantly improved stability, minimal aggregation and superior product quality. These spFv multispecifics retain binding affinity and functionality. Our stapling design was compatible with all antibody variable regions we evaluated and may be widely applicable to stabilize scFv molecules for designing biotherapeutics with superior biophysical properties.


Subject(s)
Antibodies , Immunoglobulin Variable Region , Immunoglobulin Variable Region/chemistry , Immunoglobulin Fragments
2.
ACS Infect Dis ; 4(4): 620-634, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29411968

ABSTRACT

As obligate, intracellular parasites, Plasmodium spp. rely on invasion of host cells in order to replicate and continue their life cycle. The parasite needs to traverse the dermis and endothelium of blood vessels, invade hepatocytes and red blood cells, traverse the mosquito midgut, and enter the salivary glands to continue the cycle of infection and transmission. To traverse and invade cells, the parasite employs an actomyosin motor at the core of a larger invasion machinery complex known as the glideosome. The complex is comprised of multiple protein-protein interactions linking the motor to the internal cytoskeletal network of the parasite and to the extracellular adhesins, which directly contact the host tissue or cell surface. One key interaction is between the cytoplasmic tails of the thrombospondin related anonymous protein (TRAP) and aldolase, a bridging protein to the motor. Here, we present results from screening the Medicines for Malaria Venture (MMV) library of 400 compounds against this key protein-protein interaction. Using a surface plasmon resonance screen, we have identified several compounds that modulate the dynamics of the interaction between TRAP and aldolase. These compounds have been validated in vitro by studying their effects on sporozoite gliding motility and hepatocyte invasion. One of the MMV compounds identified reduced invasion levels by 89% at the lowest concentration tested (16 µM) and severely inhibited gliding at even lower concentrations (5 µM). By targeting protein-protein interactions, we investigated an under-explored area of parasite biology and general drug development, to identify potential antimalarial lead compounds.


Subject(s)
Antimalarials/isolation & purification , Endocytosis/drug effects , Fructose-Bisphosphate Aldolase/metabolism , Locomotion/drug effects , Plasmodium/drug effects , Protozoan Proteins/metabolism , Sporozoites/drug effects , Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Hepatocytes/parasitology , Plasmodium/physiology , Protein Binding/drug effects , Sporozoites/physiology , Surface Plasmon Resonance
3.
PLoS Pathog ; 12(7): e1005763, 2016 07.
Article in English | MEDLINE | ID: mdl-27467575

ABSTRACT

A major cause of the paucity of new starting points for drug discovery is the lack of interaction between academia and industry. Much of the global resource in biology is present in universities, whereas the focus of medicinal chemistry is still largely within industry. Open source drug discovery, with sharing of information, is clearly a first step towards overcoming this gap. But the interface could especially be bridged through a scale-up of open sharing of physical compounds, which would accelerate the finding of new starting points for drug discovery. The Medicines for Malaria Venture Malaria Box is a collection of over 400 compounds representing families of structures identified in phenotypic screens of pharmaceutical and academic libraries against the Plasmodium falciparum malaria parasite. The set has now been distributed to almost 200 research groups globally in the last two years, with the only stipulation that information from the screens is deposited in the public domain. This paper reports for the first time on 236 screens that have been carried out against the Malaria Box and compares these results with 55 assays that were previously published, in a format that allows a meta-analysis of the combined dataset. The combined biochemical and cellular assays presented here suggest mechanisms of action for 135 (34%) of the compounds active in killing multiple life-cycle stages of the malaria parasite, including asexual blood, liver, gametocyte, gametes and insect ookinete stages. In addition, many compounds demonstrated activity against other pathogens, showing hits in assays with 16 protozoa, 7 helminths, 9 bacterial and mycobacterial species, the dengue fever mosquito vector, and the NCI60 human cancer cell line panel of 60 human tumor cell lines. Toxicological, pharmacokinetic and metabolic properties were collected on all the compounds, assisting in the selection of the most promising candidates for murine proof-of-concept experiments and medicinal chemistry programs. The data for all of these assays are presented and analyzed to show how outstanding leads for many indications can be selected. These results reveal the immense potential for translating the dispersed expertise in biological assays involving human pathogens into drug discovery starting points, by providing open access to new families of molecules, and emphasize how a small additional investment made to help acquire and distribute compounds, and sharing the data, can catalyze drug discovery for dozens of different indications. Another lesson is that when multiple screens from different groups are run on the same library, results can be integrated quickly to select the most valuable starting points for subsequent medicinal chemistry efforts.


Subject(s)
Antimalarials/therapeutic use , Datasets as Topic , Drug Discovery/methods , Malaria/drug therapy , Neglected Diseases/drug therapy , Drug Evaluation, Preclinical , Humans , Small Molecule Libraries
4.
J Biol Chem ; 291(8): 3860-70, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26697886

ABSTRACT

Protein modification by small ubiquitin-related modifiers (SUMOs) is essential and conserved in the malaria parasite, Plasmodium falciparum. We have previously shown that interactions between the SUMO E1-activating and E2-conjugating enzyme in P. falciparum are distinct compared with human, suggesting a potential target for development of parasite-specific inhibitors of SUMOylation. The parasite asexual trophozoite stage is susceptible to iron-induced oxidative stress and is subsequently a target for many of the current anti-malarial drugs. Here, we provide evidence that SUMOylation plays a role in the parasite response to oxidative stress during red blood cell stages, indicative of a protective role seen in other organisms. Using x-ray crystallography, we solved the structure of the human SUMO E1 ubiquitin fold domain in complex with the E2, Ubc9. The interface defined in this structure guided in silico modeling, mutagenesis, and in vitro biochemical studies of the P. falciparum SUMO E1 and E2 enzymes, resulting in the identification of surface residues that explain species-specific interactions. Our findings suggest that parasite-specific inhibitors of SUMOylation could be developed and used in combination therapies with drugs that induce oxidative stress.


Subject(s)
Models, Molecular , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Humans , Oxidative Stress/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Species Specificity , Sumoylation/physiology , Trophozoites/chemistry , Trophozoites/enzymology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
5.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 6): 718-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057801

ABSTRACT

THB1 is one of several group 1 truncated hemoglobins (TrHb1s) encoded in the genome of the unicellular green alga Chlamydomonas reinhardtii. THB1 expression is under the control of NIT2, the master regulator of nitrate assimilation, which also controls the expression of the only nitrate reductase in the cell, NIT1. In vitro and physiological evidence suggests that THB1 converts the nitric oxide generated by NIT1 into nitrate. To aid in the elucidation of the function and mechanism of THB1, the structure of the protein was solved in the ferric state. THB1 resembles other TrHb1s, but also exhibits distinct features associated with the coordination of the heme iron by a histidine (proximal) and a lysine (distal). The new structure illustrates the versatility of the TrHb1 fold, suggests factors that stabilize the axial ligation of a lysine, and highlights the difficulty of predicting the identity of the distal ligand, if any, in this group of proteins.


Subject(s)
Algal Proteins/chemistry , Chlamydomonas reinhardtii/chemistry , Heme/chemistry , Histidine/chemistry , Lysine/chemistry , Nitrate Reductase/chemistry , Truncated Hemoglobins/chemistry , Algal Proteins/genetics , Amino Acid Motifs , Chlamydomonas reinhardtii/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Iron/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Nitrate Reductase/genetics , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structural Homology, Protein , Truncated Hemoglobins/genetics
6.
PLoS One ; 10(3): e0121074, 2015.
Article in English | MEDLINE | ID: mdl-25803609

ABSTRACT

Bactofilins are novel cytoskeleton proteins that are widespread in Gram-negative bacteria. Myxococcus xanthus, an important predatory soil bacterium, possesses four bactofilins of which one, BacM (Mxan_7475) plays an important role in cell shape maintenance. Electron and fluorescence light microscopy, as well as studies using over-expressed, purified BacM, indicate that this protein polymerizes in vivo and in vitro into ~3 nm wide filaments that further associate into higher ordered fibers of about 10 nm. Here we use a multipronged approach combining secondary structure determination, molecular modeling, biochemistry, and genetics to identify and characterize critical molecular elements that enable BacM to polymerize. Our results indicate that the bactofilin-determining domain DUF583 folds into an extended ß-sheet structure, and we hypothesize a left-handed ß-helix with polymerization into 3 nm filaments primarily via patches of hydrophobic amino acid residues. These patches form the interface allowing head-to-tail polymerization during filament formation. Biochemical analyses of these processes show that folding and polymerization occur across a wide variety of conditions and even in the presence of chaotropic agents such as one molar urea. Together, these data suggest that bactofilins are comprised of a structure unique to cytoskeleton proteins, which enables robust polymerization.


Subject(s)
Bacterial Proteins/chemistry , Cytoskeletal Proteins/chemistry , Hydrophobic and Hydrophilic Interactions , Myxococcus xanthus , Amino Acid Motifs , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary , Sequence Analysis , Surface Properties
7.
J Struct Biol ; 190(2): 93-114, 2015 May.
Article in English | MEDLINE | ID: mdl-25764948

ABSTRACT

The apicomplexan family of pathogens, which includes Plasmodium spp. and Toxoplasma gondii, are primarily obligate intracellular parasites and invade multiple cell types. These parasites express extracellular membrane protein receptors, adhesins, to form specific pathogen-host cell interaction complexes. Various adhesins are used to invade a variety of cell types. The receptors are linked to an actomyosin motor, which is part of a complex comprised of many proteins known as the invasion machinery or glideosome. To date, reviews on invasion have focused primarily on the molecular pathways and signals of invasion, with little or no structural information presented. Over 75 structures of parasite receptors and glideosome proteins have been deposited with the Protein Data Bank. These structures include adhesins, motor proteins, bridging proteins, inner membrane complex and cytoskeletal proteins, as well as co-crystal structures with peptides and antibodies. These structures provide information regarding key interactions necessary for target receptor engagement, machinery complex formation, how force is transmitted, and the basis of inhibitory antibodies. Additionally, these structures can provide starting points for the development of antibodies and inhibitory molecules targeting protein-protein interactions, with the aim to inhibit invasion. This review provides an overview of the parasite adhesin protein families, the glideosome components, glideosome architecture, and discuss recent work regarding alternative models.


Subject(s)
Actomyosin/metabolism , Apicomplexa/genetics , Apicomplexa/metabolism , Host-Pathogen Interactions/physiology , Membrane Proteins/metabolism , Models, Molecular , Multiprotein Complexes/metabolism , Databases, Protein , Host-Pathogen Interactions/genetics , Membrane Proteins/genetics , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary
8.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 9): 1186-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25195889

ABSTRACT

The apicomplexan parasite Toxoplasma gondii must invade host cells to continue its lifecycle. It invades different cell types using an actomyosin motor that is connected to extracellular adhesins via the bridging protein fructose-1,6-bisphosphate aldolase. During invasion, aldolase serves in the role of a structural bridging protein, as opposed to its normal enzymatic role in the glycolysis pathway. Crystal structures of the homologous Plasmodium falciparum fructose-1,6-bisphosphate aldolase have been described previously. Here, T. gondii fructose-1,6-bisphosphate aldolase has been crystallized in space group P22121, with the biologically relevant tetramer in the asymmetric unit, and the structure has been determined via molecular replacement to a resolution of 2.0 Å. An analysis of the quality of the model and of the differences between the four chains in the asymmetric unit and a comparison between the T. gondii and P. falciparum aldolase structures is presented.


Subject(s)
Fructose-Bisphosphate Aldolase/chemistry , Fructosediphosphates/chemistry , Toxoplasma/enzymology , Amino Acid Sequence , Animals , Crystallography, X-Ray , Molecular Sequence Data , Protein Conformation
9.
J Mol Recognit ; 26(10): 496-500, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23996492

ABSTRACT

We have developed an expression system capable of producing large quantities of low cost, specific peptides that are either His12 -tagged, biotinylated, or unlabeled. The flexibility of this peptide system is suitable for interaction studies via surface plasmon resonance (SPR), co-crystallization, and enzyme-linked immunosorbent assay. Gene blocks containing peptide sequences of interest in addition to a 15 amino acid AviTag™, were cloned into a vector expressing an N-terminal maltose binding protein. The constructs were expressed and purified, and the molecular weights of the recombinant proteins were estimated by analytical size exclusion chromatography. Successful in situ biotinylation of the AviTag was confirmed by anti-biotin western blot and was used for coupling to the surface plasmon resonance chip. We were able to validate, as a proof of concept study, the specific protein-protein interaction of Plasmodium falciparum aldolase (PfAldolase) with three different cytoplasmic adhesin tail peptides from the family of thrombospondin-related anonymous proteins (TRAPs), and to determine their affinities. This method of peptide production enables high yield production of peptides in a two-day, cost effective manner. This tool will allow us to screen for protein-protein interaction inhibitors directed toward the liver stage and blood stage complexes of the glideosome in Plasmodium species. Adaptation of this tool will allow researchers to pursue their own studies of protein-protein interactions.


Subject(s)
Fructose-Bisphosphate Aldolase/chemistry , Peptides/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Base Sequence , Biotinylation , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Fructose-Bisphosphate Aldolase/genetics , Lab-On-A-Chip Devices , Molecular Sequence Data , Peptides/genetics , Plasmids , Plasmodium falciparum/metabolism , Protein Binding , Protein Interaction Mapping , Protozoan Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Staining and Labeling , Surface Plasmon Resonance
10.
J Biol Chem ; 288(39): 27724-36, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23943616

ABSTRACT

Small ubiquitin-related modifiers (SUMOs) are post-translationally conjugated to other proteins and are thereby essential regulators of a wide range of cellular processes. Sumoylation, and enzymes of the sumoylation pathway, are conserved in the malaria causing parasite, Plasmodium falciparum. However, the specific functions of sumoylation in P. falciparum, and the degree of functional conservation between enzymes of the human and P. falciparum sumoylation pathways, have not been characterized. Here, we demonstrate that sumoylation levels peak during midstages of the intra-erythrocyte developmental cycle, concomitant with hemoglobin consumption and elevated oxidative stress. In vitro studies revealed that P. falciparum E1- and E2-conjugating enzymes interact effectively to recognize and modify RanGAP1, a model mammalian SUMO substrate. However, in heterologous reactions, P. falciparum E1 and E2 enzymes failed to interact with cognate human E2 and E1 partners, respectively, to modify RanGAP1. Structural analysis, binding studies, and functional assays revealed divergent amino acid residues within the E1-E2 binding interface that define organism-specific enzyme interactions. Our studies identify sumoylation as a potentially important regulator of oxidative stress response during the P. falciparum intra-erythrocyte developmental cycle, and define E1 and E2 interactions as a promising target for development of parasite-specific inhibitors of sumoylation and parasite replication.


Subject(s)
Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Amino Acid Sequence , Erythrocytes/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Molecular Conformation , Molecular Sequence Data , Oxidative Stress , Protein Binding , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sumoylation , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...