Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Chem Rev ; 121(6): 3238-3270, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33410674

ABSTRACT

Drug resistance is prevalent across many diseases, rendering therapies ineffective with severe financial and health consequences. Rather than accepting resistance after the fact, proactive strategies need to be incorporated into the drug design and development process to minimize the impact of drug resistance. These strategies can be derived from our experience with viral disease targets where multiple generations of drugs had to be developed to combat resistance and avoid antiviral failure. Significant efforts including experimental and computational structural biology, medicinal chemistry, and machine learning have focused on understanding the mechanisms and structural basis of resistance against direct-acting antiviral (DAA) drugs. Integrated methods show promise for being predictive of resistance and potency. In this review, we give an overview of this research for human immunodeficiency virus type 1, hepatitis C virus, and influenza virus and the lessons learned from resistance mechanisms of DAAs. These lessons translate into rational strategies to avoid resistance in drug design, which can be generalized and applied beyond viral targets. While resistance may not be completely avoidable, rational drug design can and should incorporate strategies at the outset of drug development to decrease the prevalence of drug resistance.


Subject(s)
Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Pharmaceutical Preparations/chemistry , Viral Proteins/chemistry , Virus Diseases/drug therapy , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Computational Biology , Drug Design , Drug Resistance, Viral , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , HIV-1/drug effects , Hepacivirus/drug effects , Humans , Machine Learning , Mutation , Orthomyxoviridae/drug effects , Pharmaceutical Preparations/metabolism , Protein Binding , Signal Transduction , Structure-Activity Relationship
2.
J Chem Inf Model ; 60(7): 3489-3498, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32539379

ABSTRACT

A tremendous research and development effort was exerted toward combating chronic hepatitis C, ultimately leading to curative oral treatments, all of which are targeting viral proteins. Despite the advantage of numerous targets allowing for broad hepatitis C virus (HCV) genotype coverage, the only host target inhibitors that advanced into clinical development were Cyclosporin A based cyclophilin inhibitors. While cyclosporin-based molecules typically require a fermentation process, Gilead successfully pursued a fully synthetic, oral program based on Sanglifehrin A. The drug discovery process, though greatly helped by facile crystallography, was still hampered by the limitations in the accuracy of predictive computational methods for prioritizing compound ideas. Recent advances in accuracy and speed of free energy perturbation (FEP) methods, however, are attractive for prioritizing and derisking synthetically challenging molecules and potentially could have had a significant impact on the speed of the development of this program. Here in our simulated prospective study, the binding free energies of 26 macrocyclic cyclophilin inhibitors were blindly predicted using FEP+ to test this hypothesis. The predictions had a low mean unsigned error (MUE) (1.1 kcal/mol) and accurately reproduced many design decisions from the program, suggesting that FEP+ has the potential to drive synthetic chemistry efforts by more accurately ranking compounds with nonintuitive structure-activity relationships (SARs).


Subject(s)
Drug Discovery , Entropy , Prospective Studies , Structure-Activity Relationship , Thermodynamics
3.
J Chem Theory Comput ; 14(5): 2784-2796, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29570286

ABSTRACT

Water is essential in many biological processes, and the hydration structure plays a critical role in facilitating protein folding, dynamics, and ligand binding. A variety of biophysical spectroscopic techniques have been used to probe the water solvating proteins, often complemented with molecular dynamics (MD) simulations to resolve the spatial and dynamic features of the hydration shell, but comparing relative water structure is challenging. In this study 1 µs MD simulations were performed to identify and characterize hydration sites around HIV-1 protease bound to an inhibitor, darunavir (DRV). The water density, hydration site occupancy, extent and anisotropy of fluctuations, coordinated water molecules, and hydrogen bonds were characterized and compared to the properties of bulk water. The water density of the principal hydration shell was found to be higher than bulk, dependent on the topology and physiochemical identity of the biomolecular surface. The dynamics of water molecules occupying principal hydration sites was highly dependent on the number of water-water interactions and inversely correlated with hydrogen bonds to the protein-inhibitor complex. While many waters were conserved following the symmetry of homodimeric HIV protease, the asymmetry induced by DRV resulted in asymmetric lower-occupancy hydration sites at the concave surface of the active site. Key interactions between water molecules and the protease, that stabilize the protein in the inhibited form, were altered in a drug resistant variant of the protease indicating that modulation of solvent-solute interactions might play a key role in conveying drug resistance. Our analysis provides insights into the interplay between an enzyme inhibitor complex and the hydration shell and has implications in elucidating water structure in a variety of biological processes and applications including ligand binding, inhibitor design, and resistance.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , HIV-1/enzymology , Catalytic Domain , Drug Resistance, Viral , HIV Protease/drug effects , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Hydrogen Bonding , Molecular Dynamics Simulation , Surface Properties , Water/chemistry
4.
Biochemistry ; 57(10): 1652-1662, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29457713

ABSTRACT

In the era of state-of-the-art inhibitor design and high-resolution structural studies, detection of significant but small protein structural differences in the inhibitor-bound forms is critical to further developing the inhibitor. Here, we probed differences in HIV-1 protease (PR) conformation among darunavir and four analogous inhibitor-bound forms and compared them with a drug-resistant mutant using nuclear magnetic resonance chemical shifts. Changes in amide chemical shifts of wild-type (WT) PR among these inhibitor-bound forms, ΔCSP, were subtle but detectable and extended >10 Å from the inhibitor-binding site, asymmetrically between the two subunits of PR. Molecular dynamics simulations revealed differential local hydrogen bonding as the molecular basis of this remote asymmetric change. Inhibitor-bound forms of the drug-resistant mutant also showed a similar long-range ΔCSP pattern. Differences in ΔCSP values of the WT and the mutant (ΔΔCSPs) were observed at the inhibitor-binding site and in the surrounding region. Comparing chemical shift changes among highly analogous inhibitors and ΔΔCSPs effectively eliminated local environmental effects stemming from different chemical groups and enabled exploitation of these sensitive parameters to detect subtle protein conformational changes and to elucidate asymmetric and remote conformational effects upon inhibitor interaction.


Subject(s)
Drug Resistance, Viral , HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , Mutation , Nuclear Magnetic Resonance, Biomolecular/methods , Amino Acid Sequence , HIV Protease/drug effects , HIV Protease/genetics , Molecular Dynamics Simulation , Protein Conformation
5.
Proc Natl Acad Sci U S A ; 114(44): 11751-11756, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078326

ABSTRACT

Developing tools to accurately predict the clinical prevalence of drug-resistant mutations is a key step toward generating more effective therapeutics. Here we describe a high-throughput CRISPR-Cas9-based saturated mutagenesis approach to generate comprehensive libraries of point mutations at a defined genomic location and systematically study their effect on cell growth. As proof of concept, we mutagenized a selected region within the leukemic oncogene BCR-ABL1 Using bulk competitions with a deep-sequencing readout, we analyzed hundreds of mutations under multiple drug conditions and found that the effects of mutations on growth in the presence or absence of drug were critical for predicting clinically relevant resistant mutations, many of which were cancer adaptive in the absence of drug pressure. Using this approach, we identified all clinically isolated BCR-ABL1 mutations and achieved a prediction score that correlated highly with their clinical prevalence. The strategy described here can be broadly applied to a variety of oncogenes to predict patient mutations and evaluate resistance susceptibility in the development of new therapeutics.


Subject(s)
CRISPR-Cas Systems/genetics , Drug Resistance, Neoplasm/genetics , Mutagenesis/genetics , Animals , Antineoplastic Agents/pharmacology , CRISPR-Cas Systems/drug effects , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/genetics , Leukemia/drug therapy , Leukemia/genetics , Mice , Mutagenesis/drug effects , Oncogenes/genetics , Point Mutation/drug effects , Point Mutation/genetics
6.
Sci Immunol ; 2(12)2017 Jun 09.
Article in English | MEDLINE | ID: mdl-28783661

ABSTRACT

Many citrullinated proteins are known autoantigens in rheumatoid arthritis, a disease mediated by inflammatory cytokines, such as tumor necrosis factor-α (TNFα). Citrullinated proteins are generated by converting peptidylarginine to peptidylcitrulline, a process catalyzed by the peptidylarginine deiminases (PADs), including PAD1 to PAD4 and PAD6. Several major risk factors for rheumatoid arthritis are associated with heightened citrullination. However, the physiological role of citrullination in immune cells is poorly understood. We report that suppression of PAD activity attenuates Toll-like receptor-induced expression of interleukin-1ß (IL-1ß) and TNFα by neutrophils in vivo and in vitro but not their global transcription activity. Mechanistically, PAD4 directly citrullinates nuclear factor κB (NF-κB) p65 and enhances the interaction of p65 with importin α3, which brings p65 into the nucleus. The citrullination-enhanced interaction of p65 with importin α3 and its nuclear translocation and transcriptional activity can be attributed to citrullination of four arginine residues located in the Rel homology domain of p65. Furthermore, a rheumatoid arthritis-prone variant of PAD4, carrying three missense mutations, is more efficient in interacting with p65 and enhancing NF-κB activity. Together, these data not only demonstrate a critical role of citrullination in an NF-κB-dependent expression of IL-1ß and TNFα but also provide a molecular mechanism by which heightened citrullination propagates inflammation in rheumatoid arthritis. Accordingly, attenuating p65-mediated production of IL-1ß and TNFα by blocking the citrullination of p65 has great therapeutic potential in rheumatoid arthritis.

7.
J Chem Theory Comput ; 13(5): 2300-2309, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28358514

ABSTRACT

Molecular recognition is a highly interdependent process. Subsite couplings within the active site of proteases are most often revealed through conditional amino acid preferences in substrate recognition. However, the potential effect of these couplings on inhibition and thus inhibitor design is largely unexplored. The present study examines the interdependency of subsites in HIV-1 protease using a focused library of protease inhibitors, to aid in future inhibitor design. Previously a series of darunavir (DRV) analogs was designed to systematically probe the S1' and S2' subsites. Co-crystal structures of these analogs with HIV-1 protease provide the ideal opportunity to probe subsite interdependency. All-atom molecular dynamics simulations starting from these structures were performed and systematically analyzed in terms of atomic fluctuations, intermolecular interactions, and water structure. These analyses reveal that the S1' subsite highly influences other subsites: the extension of the hydrophobic P1' moiety results in 1) reduced van der Waals contacts in the P2' subsite, 2) more variability in the hydrogen bond frequencies with catalytic residues and the flap water, and 3) changes in the occupancy of conserved water sites both proximal and distal to the active site. In addition, one of the monomers in this homodimeric enzyme has atomic fluctuations more highly correlated with DRV than the other monomer. These relationships intricately link the HIV-1 protease subsites and are critical to understanding molecular recognition and inhibitor binding. More broadly, the interdependency of subsite recognition within an active site requires consideration in the selection of chemical moieties in drug design; this strategy is in contrast to what is traditionally done with independent optimization of chemical moieties of an inhibitor.


Subject(s)
Darunavir/analogs & derivatives , Darunavir/pharmacology , Drug Design , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease/chemistry , HIV-1/enzymology , Humans , Hydrogen Bonding/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation/drug effects , Water/chemistry
8.
Proc Natl Acad Sci U S A ; 112(50): 15354-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621728

ABSTRACT

Although humanized antibodies have been highly successful in the clinic, all current humanization techniques have potential limitations, such as: reliance on rodent hosts, immunogenicity due to high non-germ-line amino acid content, v-domain destabilization, expression and formulation issues. This study presents a technology that generates stable, soluble, ultrahumanized antibodies via single-step complementarity-determining region (CDR) germ-lining. For three antibodies from three separate key immune host species, binary substitution CDR cassettes were inserted into preferred human frameworks to form libraries in which only the parental or human germ-line destination residue was encoded at each position. The CDR-H3 in each case was also augmented with 1 ± 1 random substitution per clone. Each library was then screened for clones with restored antigen binding capacity. Lead ultrahumanized clones demonstrated high stability, with affinity and specificity equivalent to, or better than, the parental IgG. Critically, this was mainly achieved on germ-line frameworks by simultaneously subtracting up to 19 redundant non-germ-line residues in the CDRs. This process significantly lowered non-germ-line sequence content, minimized immunogenicity risk in the final molecules and provided a heat map for the essential non-germ-line CDR residue content of each antibody. The ABS technology therefore fully optimizes the clinical potential of antibodies from rodents and alternative immune hosts, rendering them indistinguishable from fully human in a simple, single-pass process.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Complementarity Determining Regions/immunology , Germ Cells/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity/immunology , Clone Cells , Complementarity Determining Regions/chemistry , Computer Simulation , Enzyme-Linked Immunosorbent Assay , Epitopes, T-Lymphocyte/immunology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Peptide Library , Protein Stability , Protein Structure, Tertiary , Rats , Sequence Alignment , Sequence Analysis, Protein , tau Proteins/chemistry , tau Proteins/immunology
9.
ACS Chem Biol ; 10(4): 1118-27, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25636004

ABSTRACT

Oncogenic transcriptional coregulators C-terminal Binding Protein (CtBP) 1 and 2 possess regulatory d-isomer specific 2-hydroxyacid dehydrogenase (D2-HDH) domains that provide an attractive target for small molecule intervention. Findings that the CtBP substrate 4-methylthio 2-oxobutyric acid (MTOB) can interfere with CtBP oncogenic activity in cell culture and in mice confirm that such inhibitors could have therapeutic benefit. Recent crystal structures of CtBP 1 and 2 revealed that MTOB binds in an active site containing a dominant tryptophan and a hydrophilic cavity, neither of which are present in other D2-HDH family members. Here, we demonstrate the effectiveness of exploiting these active site features for the design of high affinity inhibitors. Crystal structures of two such compounds, phenylpyruvate (PPy) and 2-hydroxyimino-3-phenylpropanoic acid (HIPP), show binding with favorable ring stacking against the CtBP active site tryptophan and alternate modes of stabilizing the carboxylic acid moiety. Moreover, ITC experiments show that HIPP binds to CtBP with an affinity greater than 1000-fold over that of MTOB, and enzymatic assays confirm that HIPP substantially inhibits CtBP catalysis. These results, thus, provide an important step, and additional insights, for the development of highly selective antineoplastic CtBP inhibitors.


Subject(s)
Alcohol Oxidoreductases/chemistry , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nerve Tissue Proteins/chemistry , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Binding Sites , Co-Repressor Proteins , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/metabolism , Humans , Hydroxylamines/chemistry , Hydroxylamines/metabolism , Hydroxylamines/pharmacology , Ligands , Models, Molecular , NAD/chemistry , NAD/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Phenylpropionates/chemistry , Phenylpropionates/metabolism , Phenylpropionates/pharmacology , Phenylpyruvic Acids/chemistry , Phenylpyruvic Acids/metabolism , Phenylpyruvic Acids/pharmacology , Protein Conformation , Structure-Activity Relationship , Thermodynamics
10.
J Chem Theory Comput ; 10(8): 3438-3448, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25136270

ABSTRACT

Under the selective pressure of therapy, HIV-1 protease mutants resistant to inhibitors evolve to confer drug resistance. Such mutations can impact both the dynamics and structures of the bound and unbound forms of the enzyme. Flap+ is a multidrug-resistant variant of HIV-1 protease with a combination of primary and secondary resistance mutations (L10I, G48V, I54V, V82A) and a strikingly altered thermodynamic profile for darunavir (DRV) binding relative to the wild-type protease. We elucidated the impact of these mutations on protein dynamics in the DRV-bound state using molecular dynamics simulations and NMR relaxation experiments. Both methods concur in that the conformational ensemble and dynamics of protease are impacted by the drug resistance mutations in Flap+ variant. Surprisingly this change in ensemble dynamics is different from that observed in the unliganded form of the same variant (Cai, Y. et al. J. Chem. Theory Comput.2012, 8, 3452-3462). Our comparative analysis of both inhibitor-free and bound states presents a comprehensive picture of the altered dynamics in drug-resistant mutant HIV-1 protease and underlies the importance of incorporating dynamic analysis of the whole system, including the unliganded state, into revealing drug resistance mechanisms.

11.
J Med Chem ; 57(6): 2643-56, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24568657

ABSTRACT

Species of Candida, primarily C. albicans and with increasing prevalence, C. glabrata, are responsible for the majority of fungal bloodstream infections that cause morbidity, especially among immune compromised patients. While the development of new antifungal agents that target the essential enzyme, dihydrofolate reductase (DHFR), in both Candida species would be ideal, previous attempts have resulted in antifolates that exhibit inconsistencies between enzyme inhibition and antifungal properties. In this article, we describe the evaluation of pairs of propargyl-linked antifolates that possess similar physicochemical properties but different shapes. All of these compounds are effective at inhibiting the fungal enzymes and the growth of C. glabrata; however, the inhibition of the growth of C. albicans is shape-dependent with extended para-linked compounds proving more effective than compact, meta-linked compounds. Using crystal structures of DHFR from C. albicans and C. glabrata bound to lead compounds, 13 new para-linked compounds designed to inhibit both species were synthesized. Eight of these compounds potently inhibit the growth of both fungal species with three compounds displaying dual MIC values less than 1 µg/mL. Analysis of the active compounds shows that shape and distribution of polar functionality is critical in achieving dual antifungal activity.


Subject(s)
Antifungal Agents , Candida albicans/drug effects , Candida glabrata/drug effects , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Morphinans/chemical synthesis , Morphinans/pharmacology , Chromatography, High Pressure Liquid , Crystallization , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Morphinans/chemistry , NADP/chemistry , Solubility , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/drug effects , X-Ray Diffraction
12.
MAbs ; 5(6): 882-95, 2013.
Article in English | MEDLINE | ID: mdl-23995618

ABSTRACT

While myriad molecular formats for bispecific antibodies have been examined to date, the simplest structures are often based on the scFv. Issues with stability and manufacturability in scFv-based bispecific molecules, however, have been a significant hindrance to their development, particularly for high-concentration, stable formulations that allow subcutaneous delivery. Our aim was to generate a tetravalent bispecific molecule targeting two inflammatory mediators for synergistic immune modulation. We focused on an scFv-Fc-scFv format, with a flexible (A4T)3 linker coupling an additional scFv to the C-terminus of an scFv-Fc. While one of the lead scFvs isolated directly from a naïve library was well-behaved and sufficiently potent, the parental anti-CXCL13 scFv 3B4 required optimization for affinity, stability, and cynomolgus ortholog cross-reactivity. To achieve this, we eschewed framework-based stabilizing mutations in favor of complementarity-determining region (CDR) mutagenesis and re-selection for simultaneous improvements in both affinity and thermal stability. Phage-displayed 3B4 CDR-mutant libraries were used in an aggressive "hammer-hug" selection strategy that incorporated thermal challenge, functional, and biophysical screening. This approach identified leads with improved stability and>18-fold, and 4,100-fold higher affinity for both human and cynomolgus CXCL13, respectively. Improvements were exclusively mediated through only 4 mutations in VL-CDR3. Lead scFvs were reformatted into scFv-Fc-scFvs and their biophysical properties ranked. Our final candidate could be formulated in a standard biopharmaceutical platform buffer at 100 mg/ml with<2% high molecular weight species present after 7 weeks at 4 °C and viscosity<15 cP. This workflow has facilitated the identification of a truly manufacturable scFv-based bispecific therapeutic suitable for subcutaneous administration.


Subject(s)
Antibodies, Bispecific/genetics , Complementarity Determining Regions/genetics , Protein Engineering , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Animals , Bacteriophages/genetics , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Subcutaneous , Peptide Library , Protein Stability , Rats , Single-Chain Antibodies/genetics , Temperature
13.
PLoS One ; 8(5): e62616, 2013.
Article in English | MEDLINE | ID: mdl-23700410

ABSTRACT

Strong genetic data link the Tyrosine kinase receptor B (TrkB) and its major endogenous ligand brain-derived neurotrophic factor (BDNF) to the regulation of energy homeostasis, with loss-of-function mutations in either gene causing severe obesity in both mice and humans. It has previously been reported that peripheral administration of the endogenous TrkB agonist ligand neurotrophin-4 (NT-4) profoundly decreases food intake and body weight in rodents, while paradoxically increasing these same parameters in monkeys. We generated a humanized TrkB agonist antibody, TAM-163, and characterized its therapeutic potential in several models of type 2 diabetes and obesity. In vitro, TAM-163 bound to human and rodent TrkB with high affinity, activated all aspects of the TrkB signaling cascade and induced TrkB internalization and degradation in a manner similar to BDNF. In vivo, peripheral administration of TAM-163 decreased food intake and/or body weight in mice, rats, hamsters, and dogs, but increased food intake and body weight in monkeys. The magnitude of weight change was similar in rodents and non-human primates, occurred at doses where there was no appreciable penetration into deep structures of the brain, and could not be explained by differences in exposures between species. Rather, peripherally administered TAM-163 localized to areas in the hypothalamus and the brain stem located outside the blood-brain barrier in a similar manner between rodents and non-human primates, suggesting differences in neuroanatomy across species. Our data demonstrate that a TrkB agonist antibody, administered peripherally, causes species-dependent effects on body weight similar to the endogenous TrkB ligand NT-4. The possible clinical utility of TrkB agonism in treating weight regulatory disorder, such as obesity or cachexia, will require evaluation in man.


Subject(s)
Antibodies, Monoclonal/pharmacology , Appetite Depressants/pharmacology , Body Weight/drug effects , Receptor, trkB/agonists , Administration, Intravenous , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacokinetics , Cricetinae , Diet, High-Fat/adverse effects , Dogs , Drug Evaluation, Preclinical , Energy Intake/drug effects , Female , HEK293 Cells , Humans , Hypothalamus/metabolism , Macaca fascicularis , Macaca mulatta , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/pathology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptor, trkB/metabolism , Tissue Distribution
14.
J Mol Biol ; 425(10): 1712-30, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23429058

ABSTRACT

We have generated large libraries of single-chain Fv antibody fragments (>10(10) transformants) containing unbiased amino acid diversity that is restricted to the central combining site of the stable, well-expressed DP47 and DPK22 germline V-genes. Library WySH2A was constructed to examine the potential for synthetic complementarity-determining region (CDR)-H3 diversity to act as the lone source of binding specificity. Library WySH2B was constructed to assess the necessity for diversification in both the H3 and L3. Both libraries provided diverse, specific antibodies, yielding a total of 243 unique hits against 7 different targets, but WySH2B produced fewer hits than WySH2A when selected in parallel. WySH2A also consistently produced hits of similar quality to WySH2B, demonstrating that the diversification of the CDR-L3 reduces library fitness. Despite the absence of deliberate bias in the library design, CDR length was strongly associated with the number of hits produced, leading to a functional loop length distribution profile that mimics the biases observed in the natural repertoire. A similar trend was also observed for the CDR-L3. After target selections, several key amino acids were enriched in the CDR-H3 (e.g., small and aromatic residues) while others were reduced (e.g., strongly charged residues) in a manner that was specific to position, preferentially occurred in CDR-H3 stem positions, and tended towards residues associated with loop stabilization. As proof of principle for the WySH2 libraries to produce viable lead candidate antibodies, 114 unique hits were produced against Delta-like ligand 4 (DLL4). Leads exhibited nanomolar binding affinities, highly specific staining of DLL4+ cells, and biochemical neutralization of DLL4-NOTCH1 interaction.


Subject(s)
Antibody Specificity , Complementarity Determining Regions/immunology , Complementarity Determining Regions/therapeutic use , Peptide Library , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Antibody Specificity/genetics , Calcium-Binding Proteins , Cloning, Molecular , Complementarity Determining Regions/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Mice , Models, Molecular , Mutation , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Single-Chain Antibodies/genetics
15.
Bioorg Med Chem Lett ; 23(5): 1279-84, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23375226

ABSTRACT

A novel strategy for targeting the pathogenic organisms Candida albicans and Candida glabrata focuses on the development of potent and selective antifolates effective against dihydrofolate reductase. Crystal structure analysis suggested that an essential loop at the active site (Thr 58-Phe 66) differs from the analogous residues in the human enzyme, potentially providing a mechanism for achieving selectivity. In order to probe the role of this loop, we employed chemical synthesis, crystal structure determination and molecular dynamics simulations. The results of these analyses show that the loop residues undergo ligand-induced conformational changes that are similar among the fungal and human species.


Subject(s)
Candida/enzymology , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Structure-Activity Relationship
16.
J Biol Chem ; 287(53): 44425-34, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23148212

ABSTRACT

Highly specific antibodies to phosphoepitopes are valuable tools to study phosphorylation in disease states, but their discovery is largely empirical, and the molecular mechanisms mediating phosphospecific binding are poorly understood. Here, we report the generation and characterization of extremely specific recombinant chicken antibodies to three phosphoepitopes on the Alzheimer disease-associated protein tau. Each antibody shows full specificity for a single phosphopeptide. The chimeric IgG pT231/pS235_1 exhibits a K(D) of 0.35 nm in 1:1 binding to its cognate phosphopeptide. This IgG is murine ortholog-cross-reactive, specifically recognizing the pathological form of tau in brain samples from Alzheimer patients and a mouse model of tauopathy. To better understand the underlying binding mechanisms allowing such remarkable specificity, we determined the structure of pT231/pS235_1 Fab in complex with its cognate phosphopeptide at 1.9 Å resolution. The Fab fragment exhibits novel complementarity determining region (CDR) structures with a "bowl-like" conformation in CDR-H2 that tightly and specifically interacts with the phospho-Thr-231 phosphate group, as well as a long, disulfide-constrained CDR-H3 that mediates peptide recognition. This binding mechanism differs distinctly from either peptide- or hapten-specific antibodies described to date. Surface plasmon resonance analyses showed that pT231/pS235_1 binds a truly compound epitope, as neither phosphorylated Ser-235 nor free peptide shows any measurable binding affinity.


Subject(s)
Alzheimer Disease/metabolism , Antibodies/immunology , Epitopes/immunology , tau Proteins/immunology , Alzheimer Disease/genetics , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/genetics , Brain/metabolism , Chickens , Epitopes/chemistry , Epitopes/genetics , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Phosphorylation , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/metabolism
17.
Dev Comp Immunol ; 36(4): 665-79, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22040740

ABSTRACT

The cartilaginous fish (chimeras, sharks, skates and rays) are the oldest group relative to mammals in which an adaptive immune system founded upon immunoglobulins has been found. In this manuscript we characterize the immunoglobulins of the spiny dogfish (Squalus acanthias) at both the molecular and expressed protein levels. Despite the presence of hundreds of IgM clusters in this species the serum levels of this isotype are comparatively low. However, analysis of cDNA sequences and serum protein suggests microheterogeneity in the IgM heavy chains and supports the proposal that different clusters are preferentially used in the two forms (monomer or pentamer) of this isotype. We also found that the IgNAR isotype in this species exists in a previously unknown multimeric format in serum. Finally, we identified a new form of the IgW isotype (the shark IgD orthologue), in which the leader is spliced directly to the first constant domain, resulting in a molecule lacking an antigen-binding domain.


Subject(s)
Immunoglobulins/chemistry , Immunoglobulins/immunology , Squalus acanthias/immunology , Amino Acid Sequence , Animals , Immunoglobulins/genetics , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sharks/genetics , Sharks/immunology , Squalus acanthias/genetics
18.
J Immunol ; 188(1): 322-33, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22131336

ABSTRACT

Examination of 1269 unique naive chicken V(H) sequences showed that the majority of positions in the framework (FW) regions were maintained as germline, with high mutation rates observed in the CDRs. Many FW mutations could be clearly related to the modulation of CDR structure or the V(H)-V(L) interface. CDRs 1 and 2 of the V(H) exhibited frequent mutation in solvent-exposed positions, but conservation of common structural residues also found in human CDRs at the same positions. In comparison with humans and mice, the chicken CDR3 repertoire was skewed toward longer sequences, was dominated by small amino acids (G/S/A/C/T), and had higher cysteine (chicken, 9.4%; human, 1.6%; and mouse, 0.25%) but lower tyrosine content (chicken, 9.2%; human, 16.8%; and mouse 26.4%). A strong correlation (R(2) = 0.97) was observed between increasing CDR3 length and higher cysteine content. This suggests that noncanonical disulfides are strongly favored in chickens, potentially increasing CDR stability and complexity in the topology of the combining site. The probable formation of disulfide bonds between CDR3 and CDR1, FW2, or CDR2 was also observed, as described in camelids. All features of the naive repertoire were fully replicated in the target-selected, phage-displayed repertoire. The isolation of a chicken Fab with four noncanonical cysteines in the V(H) that exhibits 64 nM (K(D)) binding affinity for its target proved these constituents to be part of the humoral response, not artifacts. This study supports the hypothesis that disulfide bond-constrained CDR3s are a structural diversification strategy in the restricted germline v-gene repertoire of chickens.


Subject(s)
Amino Acid Substitution , Chickens/genetics , Complementarity Determining Regions/genetics , Immunoglobulin Heavy Chains/genetics , Mutation , Animals , Antibody Affinity/genetics , Camelus/genetics , Camelus/immunology , Chickens/immunology , Complementarity Determining Regions/immunology , Disulfides/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Mice , Protein Stability , Species Specificity
19.
Chem Biol Drug Des ; 78(4): 505-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21726415

ABSTRACT

Candida albicans and Candida glabrata cause fungal bloodstream infections that are associated with significant mortality. As part of an effort to develop potent and selective antifolates that target dihydrofolate reductase (DHFR) from Candida species, we report three ternary crystal structures of C. albicans DHFR (CaDHFR) bound to novel propargyl-linked analogs. Consistent with earlier modeling results, these structures show that hydrophobic pockets in the binding site may be exploited to increase ligand potency. The crystal structures also confirm that loop residues Thr 58- Phe 66, which flank the active site and influence ligand potency and selectivity, adopt multiple conformations. To aid the development of a dual Candida spp. inhibitor, three new crystal structures of C. glabrata DHFR (CgDHFR) bound to similar ligands as those bound in the ternary structures of CaDHFR are also reported here. Loop residues 58-66 in CgDHFR and human DHFR are 1 and 3 Å closer to the folate binding site, respectively, than loop residues in CaDHFR, suggesting that a properly size ligand could be a potent and selective dual inhibitor of CaDHFR and CgDHFR.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Candida albicans/enzymology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Candida albicans/chemistry , Candida albicans/drug effects , Candidiasis/drug therapy , Catalytic Domain , Crystallography, X-Ray , Drug Design , Humans , Ligands , Models, Molecular , Pargyline/analogs & derivatives , Protein Binding , Tetrahydrofolate Dehydrogenase/chemistry
20.
J Mol Graph Model ; 29(5): 608-13, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21146434

ABSTRACT

Dihydrofolate reductase (DHFR) has been a well-recognized target for the development of therapeutics for human cancers for several decades. Classical inhibitors of DHFR use an active transport mechanism to gain access to the cell; disabling this mechanism creates a pathway for resistance. In response, recent research focuses on nonclassical lipid-soluble DHFR inhibitors that are designed to passively diffuse through the membrane. Here, a new series of propargyl-linked antifolates are investigated as potential nonclassical human DHFR inhibitors. Several of these compounds exhibit potent enzyme inhibition with 50% inhibition concentration values under 500 nM. Molecular docking investigations show that the compounds maintain conserved hydrogen bonds between the pyrimidine ring and the enzyme as well as form van der Waals interactions with critical residues in the active site. Interestingly, the most potent compound, 2,4-diamino-5-(3-(3,4,5-trimethoxyphenyl)prop-1-ynyl)-6-ethylpyrimidine (compound 35), is 3500-fold more potent than trimethoprim, a potent inhibitor of bacterial DHFR but weak inhibitor of human DHFR. The two structural differences between compound 35 and trimethoprim show that the propargyl linkage and the substitution at C6 of the pyrimidine ring are critical to the formation of contacts with Thr 56, Ser 59, Ile 60, Leu 22, Phe 31 and Phe 34 and hence, to enhancing potency. The propargyl-linked antifolates are efficient ligands with a high ratio of potency to the number of non-hydrogen atoms and represent a potentially fruitful avenue for future development of antineoplastic agents.


Subject(s)
Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Tetrahydrofolate Dehydrogenase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL