Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Commun ; 10(1): 4606, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601808

ABSTRACT

The current leading Zika vaccine candidates in clinical testing are based on live or killed virus platforms, which have safety issues, especially in pregnant women. Zika subunit vaccines, however, have shown poor performance in preclinical studies, most likely because the antigens tested do not display critical quaternary structure epitopes present on Zika E protein homodimers that cover the surface of the virus. Here, we produce stable recombinant E protein homodimers that are recognized by strongly neutralizing Zika specific monoclonal antibodies. In mice, the dimeric antigen stimulate strongly neutralizing antibodies that target epitopes that are similar to epitopes recognized by human antibodies following natural Zika virus infection. The monomer antigen stimulates low levels of E-domain III targeting neutralizing antibodies. In a Zika challenge model, only E dimer antigen stimulates protective antibodies, not the monomer. These results highlight the importance of mimicking the highly structured flavivirus surface when designing subunit vaccines.


Subject(s)
Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Vaccines/immunology , Zika Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Epitopes/immunology , Female , Humans , Mice, Inbred C57BL , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vero Cells , Viral Envelope Proteins/genetics , Zika Virus/genetics , Zika Virus Infection/immunology , Zika Virus Infection/virology
2.
Biomater Sci ; 6(11): 3063-3074, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30298866

ABSTRACT

P-glycoprotein (Pgp) has been considered as a major cause of cancer multidrug resistance; however, clinical solutions to overcome this drug resistance do not exist despite the tremendous endeavors. The lack of cancer specificity is a main reason for clinical failure of conventional approaches. Targeted photodynamic therapy (PDT) is highly cancer specific by combining antibody targeting and locoregional light irradiation. We aimed to develop Pgp-targeted PDT using antibody-photosensitizer conjugates made of a recombinant Fab fragment. We prepared the photosensitizer conjugates by expressing a recombinant Fab fragment and specifically linking IR700-maleimide at the C-terminal of the Fab heavy chain. In vitro studies showed that the Fab conjugates specifically bind to Pgp. Their phototoxicity was comparable to full antibody conjugates when assayed with conventional 2-D cell culture, but they outperformed the full antibody conjugates in a 3-D tumor spheroid model. In a mouse xenograft model of chemoresistant tumors, Fab conjugates showed Pgp specific delivery to chemoresistant tumors. Upon irradiation with near-infrared light, they caused rapid tumor shrinkage and significantly prolonged the survival of tumor-bearing mice. Compared to the full antibody conjugates, Fab conjugates took a shorter time to reach peak tumor levels and achieved a more homogeneous tumor distribution. This allows light irradiation to be initiated at a shorter time interval after the conjugate injection, and thus may facilitate clinical translation. We conclude that our targeted PDT approach provides a highly cancer-specific approach to combat chemoresistant tumors, and that the conjugates made of recombinant antibody fragments are superior to full antibody conjugates for targeted PDT.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibodies/chemistry , Antineoplastic Agents/chemistry , Drug Resistance, Neoplasm , Immunoglobulin Fab Fragments/chemistry , Photochemotherapy/methods , 3T3 Cells , Animals , Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Combined Modality Therapy/methods , Drug Resistance, Multiple , Female , Heterografts , Humans , Immunoglobulin Fab Fragments/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms/pathology , Neoplasms/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tissue Distribution
3.
PLoS Negl Trop Dis ; 12(9): e0006793, 2018 09.
Article in English | MEDLINE | ID: mdl-30248097

ABSTRACT

Dengue virus (DENV) is the causative agent of dengue fever and dengue hemorrhagic shock syndrome. Dengue vaccine development is challenging because of the need to induce protection against four antigenically distinct DENV serotypes. Recent studies indicate that tetravalent DENV vaccines must induce balanced, serotype-specific neutralizing antibodies to achieve durable protective immunity against all 4 serotypes. With the leading live attenuated tetravalent DENV vaccines, it has been difficult to achieve balanced and type-specific responses to each serotype, most likely because of unbalanced replication of vaccine viral strains. Here we evaluate a tetravalent DENV protein subunit vaccine, based on recombinant envelope protein (rE) adsorbed to the surface of poly (lactic-co-glycolic acid) (PLGA) nanoparticles for immunogenicity in mice. In monovalent and tetravalent formulations, we show that particulate rE induced higher neutralizing antibody titers compared to the soluble rE antigen alone. Importantly, we show the trend that tetravalent rE adsorbed to nanoparticles stimulated a more balanced serotype specific antibody response to each DENV serotype compared to soluble antigens. Our results demonstrate that tetravalent DENV subunit vaccines displayed on nanoparticles have the potential to overcome unbalanced immunity observed for leading live-attenuated vaccine candidates.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dengue Vaccines/immunology , Dengue Virus/immunology , Nanoparticles/administration & dosage , Viral Structural Proteins/immunology , Animals , Dengue Vaccines/administration & dosage , Female , Mice, Inbred BALB C , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
4.
J Biol Chem ; 293(23): 8922-8933, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29678884

ABSTRACT

The spread of dengue (DENV) and Zika virus (ZIKV) is a major public health concern. The primary target of antibodies that neutralize DENV and ZIKV is the envelope (E) glycoprotein, and there is interest in using soluble recombinant E (sRecE) proteins as subunit vaccines. However, the most potent neutralizing antibodies against DENV and ZIKV recognize epitopes on the virion surface that span two or more E proteins. Therefore, to create effective DENV and ZIKV vaccines, presentation of these quaternary epitopes may be necessary. The sRecE proteins from DENV and ZIKV crystallize as native-like dimers, but studies in solution suggest that these dimers are marginally stable. To better understand the challenges associated with creating stable sRecE dimers, we characterized the thermostability of sRecE proteins from ZIKV and three DENV serotypes, DENV2-4. All four proteins irreversibly unfolded at moderate temperatures (46-53 °C). At 23 °C and low micromolar concentrations, DENV2 and ZIKV were primarily dimeric, and DENV3-4 were primarily monomeric, whereas at 37 °C, all four proteins were predominantly monomeric. We further show that the dissociation constant for DENV2 dimerization is very temperature-sensitive, ranging from <1 µm at 25 °C to 50 µm at 41 °C, due to a large exothermic enthalpy of binding of -79 kcal/mol. We also found that quaternary epitope antibody binding to DENV2-4 and ZIKV sRecE is reduced at 37 °C. Our observation of reduced sRecE dimerization at physiological temperature highlights the need for stabilizing the dimer as part of its development as a subunit vaccine.


Subject(s)
Dengue Virus/chemistry , Protein Multimerization , Viral Envelope Proteins/chemistry , Zika Virus/chemistry , Body Temperature , Dengue/virology , Humans , Protein Stability , Recombinant Proteins/chemistry , Vaccines, Subunit/chemistry , Viral Vaccines/chemistry , Zika Virus Infection/virology
5.
Bioconjug Chem ; 29(5): 1544-1552, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29701995

ABSTRACT

The dengue virus (DENV) causes over 350 million infections, resulting in ∼25,000 deaths per year globally. An effective dengue vaccine requires generation of strong and balanced neutralizing antibodies against all four antigenically distinct serotypes of DENV. The leading live-attenuated tetravalent dengue virus vaccine platform has shown partial efficacy, with an unbalanced response across the four serotypes in clinical trials. DENV subunit vaccine platforms are being developed because they provide a strong safety profile and are expected to avoid the unbalanced immunization issues associated with live multivalent vaccines. Subunit vaccines often lack immunogenicity, requiring either a particulate or adjuvanted formulation. Particulate formulations adsorbing monomeric DENV-E antigen to the particle surface incite a strong immune response, but have no control of antigen presentation. Highly neutralizing epitopes are displayed by DENV-E quaternary structures. To control the display of DENV-E and produce quaternary structures, particulate formulations that covalently attach DENV-E to the particle surface are needed. Here we develop a surface attached DENV2-E particulate formulation, as well as analysis tools, using PEG hydrogel nanoparticles created with particle replication in nonwetting templates (PRINT) technology. We found that adding Tween-20 to the conjugation buffer controls DENV-E adsorption to the particle surface during conjugation, improving both protein stability and epitope display. Immunizations with the anionic but not the cationic DENV2-E conjugated particles were able to produce DENV-specific and virus neutralizing antibody in mice. This work optimized the display of DENV-E conjugated to the surface of a nanoparticle through EDC/NHS chemistry, establishing a platform that can be expanded upon in future work to fully control the display of DENV-E.


Subject(s)
Antibodies, Neutralizing/immunology , Dengue Vaccines/immunology , Dengue Virus/immunology , Dengue/prevention & control , Immobilized Proteins/immunology , Nanoparticles , Viral Envelope Proteins/immunology , Adsorption , Animals , Antibodies, Viral/immunology , Antibody Formation , Chlorocebus aethiops , Dengue/immunology , Dengue Vaccines/administration & dosage , Dengue Vaccines/chemistry , Dengue Virus/chemistry , Female , Immobilized Proteins/administration & dosage , Immobilized Proteins/chemistry , Immunization , Mice, Inbred BALB C , Models, Molecular , Nanoparticles/chemistry , Vero Cells , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/chemistry
6.
Sci Rep ; 7(1): 4524, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28674411

ABSTRACT

Zika virus (ZIKV) and the 4 dengue virus (DENV) serotypes are mosquito-borne Flaviviruses that are associated with severe neuronal and hemorrhagic syndromes. The mature flavivirus infectious virion has 90 envelope (E) protein homo-dimers that pack tightly to form a smooth protein coat with icosahedral symmetry. Human antibodies that strongly neutralize ZIKV and DENVs recognize complex quaternary structure epitopes displayed on E-homo-dimers and higher order structures. The ZIKV and DENV E protein expressed as a soluble protein is mainly a monomer that does not display quaternary epitopes, which may explain the modest success with soluble recombinant E (sRecE) as a vaccine and diagnostic antigen. New strategies are needed to design recombinant immunogens that display these critical immune targets. Here we present two novel methods for building or stabilizing in vitro E-protein homo-dimers that display quaternary epitopes. In the first approach we immobilize sRecE to enable subsequent dimer generation. As an alternate method, we describe the use of human mAbs to stabilize homo-dimers in solution. The ability to produce recombinant E protein dimers displaying quaternary structure epitopes is an important advance with applications in flavivirus diagnostics and vaccine development.


Subject(s)
Dengue Virus , Protein Multimerization , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Assembly , Zika Virus , Animals , Antibodies, Viral/immunology , Binding Sites , Cells, Cultured , Dengue Virus/classification , Dengue Virus/physiology , Epitopes/immunology , Humans , Neutralization Tests , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Proteins , Serogroup , Structure-Activity Relationship , Viral Envelope Proteins/immunology , Zika Virus/classification , Zika Virus/physiology
7.
PLoS Negl Trop Dis ; 11(5): e0005554, 2017 May.
Article in English | MEDLINE | ID: mdl-28505154

ABSTRACT

Dengue viruses (DENVs) are mosquito-borne flaviviruses and the causative agents of dengue fever and dengue hemorrhagic fever. As there are four serotypes of DENV (DENV1-4), people can be infected multiple times, each time with a new serotype. Primary infections stimulate antibodies that mainly neutralize the serotype of infection (type-specific), whereas secondary infections stimulate responses that cross-neutralize 2 or more serotypes. Previous studies have demonstrated that neutralizing antibodies induced by primary infections recognize tertiary and quaternary structure epitopes on the viral envelope (E) protein that are unique to each serotype. The goal of the current study was to determine the properties of neutralizing antibodies induced after secondary infection with a different (heterotypic) DENV serotypes. We evaluated whether polyclonal neutralizing antibody responses after secondary infections consist of distinct populations of type-specific antibodies to each serotype encountered or a new population of broadly cross-neutralizing antibodies. We observed two types of responses: in some individuals exposed to secondary infections, DENV neutralization was dominated by cross-reactive antibodies, whereas in other individuals both type-specific and cross-reactive antibodies contributed to neutralization. To better understand the origins of type-specific and cross-reactive neutralizing antibodies, we analyzed sera from individuals with well-documented sequential infections with two DENV serotypes only. These individuals had both type-specific and cross-reactive neutralizing antibodies to the 2 serotypes responsible for infection and only cross-reactive neutralizing antibodies to other serotypes. Collectively, the results demonstrate that the quality of neutralizing (and presumably protective) antibodies are different in individuals depending on the number of previous exposures to different DENV serotypes. We propose a model in which low affinity, cross-reactive antibody secreting B-cell clones induced by primary exposure evolve during each secondary infection to secrete higher affinity and more broadly neutralizing antibodies.


Subject(s)
Antibodies, Viral/blood , Antibody Formation , Coinfection/blood , Dengue Virus/classification , Dengue/immunology , Adolescent , Antibodies, Neutralizing/blood , Child , Child, Preschool , Cohort Studies , Cross Reactions , Dengue/epidemiology , Dengue/virology , Humans , Infant , Serogroup , Travel
8.
ChemMedChem ; 12(3): 207-213, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28032464

ABSTRACT

Macrocycles have attracted significant attention in drug discovery recently. In fact, a few de novo designed macrocyclic kinase inhibitors are currently in clinical trials with good potency and selectivity for their intended target. In this study, we successfully engaged a structure-based drug design approach to discover macrocyclic pyrimidines as potent Mer tyrosine kinase (MerTK)-specific inhibitors. An enzyme-linked immunosorbent assay (ELISA) in 384-well format was employed to evaluate the inhibitory activity of macrocycles in a cell-based assay assessing tyrosine phosphorylation of MerTK. Through structure-activity relationship (SAR) studies, analogue 11 [UNC2541; (S)-7-amino-N-(4-fluorobenzyl)-8-oxo-2,9,16-triaza-1(2,4)-pyrimidinacyclohexadecaphane-1-carboxamide] was identified as a potent and MerTK-specific inhibitor that exhibits sub-micromolar inhibitory activity in the cell-based ELISA. In addition, an X-ray structure of MerTK protein in complex with 11 was resolved to show that these macrocycles bind in the MerTK ATP pocket.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrimidines/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme-Linked Immunosorbent Assay , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Macrocyclic Compounds/chemistry , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Pyrimidines/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , c-Mer Tyrosine Kinase
9.
ACS Med Chem Lett ; 7(12): 1044-1049, 2016 Dec 08.
Article in English | MEDLINE | ID: mdl-27994735

ABSTRACT

Mer tyrosine kinase (MerTK) is aberrantly elevated in various tumor cells and has a normal anti-inflammatory role in the innate immune system. Inhibition of MerTK may provide dual effects against these MerTK-expressing tumors through reducing cancer cell survival and redirecting the innate immune response. Recently, we have designed novel and potent macrocyclic pyrrolopyrimidines as MerTK inhibitors using a structure-based approach. The most active macrocycles had an EC50 below 40 nM in a cell-based MerTK phosphor-protein ELISA assay. The X-ray structure of macrocyclic analogue 3 complexed with MerTK was also resolved and demonstrated macrocycles binding in the ATP binding pocket of the MerTK protein as anticipated. In addition, the lead compound 16 (UNC3133) had a 1.6 h half-life and 16% oral bioavailability in a mouse PK study.

10.
PLoS Negl Trop Dis ; 10(10): e0005071, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27764114

ABSTRACT

Dengue virus (DENV) is the causative agent of dengue fever and dengue hemorrhagic fever. The virus is endemic in over 120 countries, causing over 350 million infections per year. Dengue vaccine development is challenging because of the need to induce simultaneous protection against four antigenically distinct DENV serotypes and evidence that, under some conditions, vaccination can enhance disease due to specific immunity to the virus. While several live-attenuated tetravalent dengue virus vaccines display partial efficacy, it has been challenging to induce balanced protective immunity to all 4 serotypes. Instead of using whole-virus formulations, we are exploring the potentials for a particulate subunit vaccine, based on DENV E-protein displayed on nanoparticles that have been precisely molded using Particle Replication in Non-wetting Template (PRINT) technology. Here we describe immunization studies with a DENV2-nanoparticle vaccine candidate. The ectodomain of DENV2-E protein was expressed as a secreted recombinant protein (sRecE), purified and adsorbed to poly (lactic-co-glycolic acid) (PLGA) nanoparticles of different sizes and shape. We show that PRINT nanoparticle adsorbed sRecE without any adjuvant induces higher IgG titers and a more potent DENV2-specific neutralizing antibody response compared to the soluble sRecE protein alone. Antigen trafficking indicate that PRINT nanoparticle display of sRecE prolongs the bio-availability of the antigen in the draining lymph nodes by creating an antigen depot. Our results demonstrate that PRINT nanoparticles are a promising platform for delivering subunit vaccines against flaviviruses such as dengue and Zika.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Dengue Vaccines/immunology , Dengue Virus/immunology , Nanoparticles , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/immunology , Chlorocebus aethiops , Dengue/immunology , Dengue/prevention & control , Dengue Vaccines/administration & dosage , Humans , Immunoglobulin G/blood , Lactic Acid/chemistry , Lymph Nodes/immunology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Recombinant Proteins/administration & dosage , Recombinant Proteins/isolation & purification , Serogroup , Surface Properties , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
11.
Mol Hum Reprod ; 22(6): 410-26, 2016 06.
Article in English | MEDLINE | ID: mdl-26921398

ABSTRACT

STUDY HYPOTHESIS: Detailed structural comparisons of sperm-specific glyceraldehyde 3-phosphate dehydrogenase, spermatogenic (GAPDHS) and the somatic glyceraldehyde 3-phosphate dehydrogenase (GAPDH) isozyme should facilitate the identification of selective GAPDHS inhibitors for contraceptive development. STUDY FINDING: This study identified a small-molecule GAPDHS inhibitor with micromolar potency and >10-fold selectivity that exerts the expected inhibitory effects on sperm glycolysis and motility. WHAT IS KNOWN ALREADY: Glycolytic ATP production is required for sperm motility and male fertility in many mammalian species. Selective inhibition of GAPDHS, one of the glycolytic isozymes with restricted expression during spermatogenesis, is a potential strategy for the development of a non-hormonal contraceptive that directly blocks sperm function. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Homology modeling and x-ray crystallography were used to identify structural features that are conserved in GAPDHS orthologs in mouse and human sperm, but distinct from the GAPDH orthologs present in somatic tissues. We identified three binding pockets surrounding the substrate and cofactor in these isozymes and conducted a virtual screen to identify small-molecule compounds predicted to bind more tightly to GAPDHS than to GAPDH. Following the production of recombinant human and mouse GAPDHS, candidate compounds were tested in dose-response enzyme assays to identify inhibitors that blocked the activity of GAPDHS more effectively than GAPDH. The effects of a selective inhibitor on the motility of mouse and human sperm were monitored by computer-assisted sperm analysis, and sperm lactate production was measured to assess inhibition of glycolysis in the target cell. MAIN RESULTS AND THE ROLE OF CHANCE: Our studies produced the first apoenzyme crystal structures for human and mouse GAPDHS and a 1.73 Å crystal structure for NAD(+)-bound human GAPDHS, facilitating the identification of unique structural features of this sperm isozyme. In dose-response assays T0501_7749 inhibited human GAPDHS with an IC50 of 1.2 µM compared with an IC50 of 38.5 µM for the somatic isozyme. This compound caused significant reductions in mouse sperm lactate production (P= 0.017 for 100 µM T0501_7749 versus control) and in the percentage of motile mouse and human sperm (P values from <0.05 to <0.0001, depending on incubation conditions). LIMITATIONS, REASONS FOR CAUTION: The chemical properties of T0501_7749, including limited solubility and nonspecific protein binding, are not optimal for drug development. WIDER IMPLICATIONS OF THE FINDINGS: This study provides proof-of-principle evidence that GAPDHS can be selectively inhibited, causing significant reductions in sperm glycolysis and motility. These results highlight the utility of structure-based drug design and support further exploration of GAPDHS, and perhaps other sperm-specific isozymes in the glycolytic pathway, as contraceptive targets. LARGE SCALE DATA: None. Coordinates and data files for three GAPDHS crystal structures were deposited in the RCSB Protein Data Bank (http://www.rcsb.org). STUDY FUNDING AND COMPETING INTERESTS: This work was supported by grants from the National Institutes of Health (NIH), USA, including U01 HD060481 and cooperative agreement U54 HD35041 as part of the Specialized Cooperative Centers Program in Reproduction and Infertility Research from the Eunice Kennedy Shriver National Institute of Child Health and Human Development, and TW/HD00627 from the NIH Fogarty International Center. Additional support was provided by subproject CIG-05-109 from CICCR, a program of CONRAD, Eastern Virginia Medical School, USA. There are no conflicts of interest.


Subject(s)
Enzyme Inhibitors/pharmacology , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Spermatozoa/drug effects , Spermatozoa/enzymology , Adenosine Triphosphate/metabolism , Animals , Crystallography, X-Ray , Glycolysis/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice , Sperm Motility/drug effects
12.
Cancer Discov ; 4(12): 1418-29, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25252692

ABSTRACT

UNLABELLED: NRAS mutation at codons 12, 13, or 61 is associated with transformation; yet, in melanoma, such alterations are nearly exclusive to codon 61. Here, we compared the melanoma susceptibility of an NrasQ61R knock-in allele to similarly designed KrasG12D and NrasG12D alleles. With concomitant p16INK4a inactivation, KrasG12D or NrasQ61R expression efficiently promoted melanoma in vivo, whereas NrasG12D did not. In addition, NrasQ61R mutation potently cooperated with Lkb1/Stk11 loss to induce highly metastatic disease. Functional comparisons of NrasQ61R and NrasG12D revealed little difference in the ability of these proteins to engage PI3K or RAF. Instead, NrasQ61R showed enhanced nucleotide binding, decreased intrinsic GTPase activity, and increased stability when compared with NrasG12D. This work identifies a faithful model of human NRAS-mutant melanoma, and suggests that the increased melanomagenecity of NrasQ61R over NrasG12D is due to heightened abundance of the active, GTP-bound form rather than differences in the engagement of downstream effector pathways. SIGNIFICANCE: This work explains the curious predominance in human melanoma of mutations of codon 61 of NRAS over other oncogenic NRAS mutations. Using conditional "knock-in" mouse models, we show that physiologic expression of NRASQ61R, but not NRASG12D, drives melanoma formation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Codon , Genes, ras , Melanoma/genetics , Mutation , AMP-Activated Protein Kinase Kinases , Alleles , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Deletion , Gene Order , Genetic Loci , Genotype , Guanosine Triphosphate/metabolism , Humans , Melanoma/metabolism , Melanoma/mortality , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tumor Burden
13.
J Med Chem ; 56(23): 9693-700, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24219778

ABSTRACT

The role of Mer kinase in regulating the second phase of platelet activation generates an opportunity to use Mer inhibitors for preventing thrombosis with diminished likelihood for bleeding as compared to current therapies. Toward this end, we have discovered a novel, Mer kinase specific substituted-pyrimidine scaffold using a structure-based drug design and a pseudo ring replacement strategy. The cocrystal structure of Mer with two compounds (7 and 22) possessing distinct activity have been determined. Subsequent SAR studies identified compound 23 (UNC2881) as a lead compound for in vivo evaluation. When applied to live cells, 23 inhibits steady-state Mer kinase phosphorylation with an IC50 value of 22 nM. Treatment with 23 is also sufficient to block EGF-mediated stimulation of a chimeric receptor containing the intracellular domain of Mer fused to the extracellular domain of EGFR. In addition, 23 potently inhibits collagen-induced platelet aggregation, suggesting that this class of inhibitors may have utility for prevention and/or treatment of pathologic thrombosis.


Subject(s)
Cyclohexanols/chemical synthesis , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/therapeutic use , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thrombosis/drug therapy , Thrombosis/prevention & control , Cyclohexanols/therapeutic use , Drug Design , Humans , Models, Molecular , Pyrimidines/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , c-Mer Tyrosine Kinase
14.
J Med Chem ; 56(23): 9683-92, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24195762

ABSTRACT

Abnormal activation or overexpression of Mer receptor tyrosine kinase has been implicated in survival signaling and chemoresistance in many human cancers. Consequently, Mer is a promising novel cancer therapeutic target. A structure-based drug design approach using a pseudo-ring replacement strategy was developed and validated to discover a new family of pyridinepyrimidine analogues as potent Mer inhibitors. Through SAR studies, 10 (UNC2250) was identified as the lead compound for further investigation based on high selectivity against other kinases and good pharmacokinetic properties. When applied to live cells, 10 inhibited steady-state phosphorylation of endogenous Mer with an IC50 of 9.8 nM and blocked ligand-stimulated activation of a chimeric EGFR-Mer protein. Treatment with 10 also resulted in decreased colony-forming potential in rhabdoid and NSCLC tumor cells, thereby demonstrating functional antitumor activity. The results provide a rationale for further investigation of this compound for therapeutic application in patients with cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclohexanols/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclization , Cyclohexanols/pharmacology , Drug Design , Humans , Hydrogen Bonding , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Structure-Activity Relationship , c-Mer Tyrosine Kinase
15.
ACS Med Chem Lett ; 3(2): 129-134, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22662287

ABSTRACT

Ectopic Mer expression promotes pro-survival signaling and contributes to leukemogenesis and chemoresistance in childhood acute lymphoblastic leukemia (ALL). Consequently, Mer kinase inhibitors may promote leukemic cell death and further act as chemosensitizers increasing efficacy and reducing toxicities of current ALL regimens. We have applied a structure-based design approach to discover novel small molecule Mer kinase inhibitors. Several pyrazolopyrimidine derivatives effectively inhibit Mer kinase activity at sub-nanomolar concentrations. Furthermore, the lead compound shows a promising selectivity profile against a panel of 72 kinases and has excellent pharmacokinetic properties. We also describe the crystal structure of the complex between the lead compound and Mer, opening new opportunities for further optimization and new template design.

16.
Structure ; 20(6): 1086-96, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22632833

ABSTRACT

Protein design tests our understanding of protein stability and structure. Successful design methods should allow the exploration of sequence space not found in nature. However, when redesigning naturally occurring protein structures, most fixed backbone design algorithms return amino acid sequences that share strong sequence identity with wild-type sequences, especially in the protein core. This behavior places a restriction on functional space that can be explored and is not consistent with observations from nature, where sequences of low identity have similar structures. Here, we allow backbone flexibility during design to mutate every position in the core (38 residues) of a four-helix bundle protein. Only small perturbations to the backbone, 1-2 Å, were needed to entirely mutate the core. The redesigned protein, DRNN, is exceptionally stable (melting point >140°C). An NMR and X-ray crystal structure show that the side chains and backbone were accurately modeled (all-atom RMSD = 1.3 Å).


Subject(s)
Bacterial Proteins/chemistry , Protein Engineering , Algorithms , Amino Acid Sequence , Crystallography, X-Ray , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Monte Carlo Method , Nuclear Magnetic Resonance, Biomolecular , Protein Stability , Protein Structure, Secondary , Thermotoga maritima , Transition Temperature
17.
J Am Chem Soc ; 134(1): 375-85, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22092237

ABSTRACT

Computationally designing protein-protein interactions with high affinity and desired orientation is a challenging task. Incorporating metal-binding sites at the target interface may be one approach for increasing affinity and specifying the binding mode, thereby improving robustness of designed interactions for use as tools in basic research as well as in applications from biotechnology to medicine. Here we describe a Rosetta-based approach for the rational design of a protein monomer to form a zinc-mediated, symmetric homodimer. Our metal interface design, named MID1 (NESG target ID OR37), forms a tight dimer in the presence of zinc (MID1-zinc) with a dissociation constant <30 nM. Without zinc the dissociation constant is 4 µM. The crystal structure of MID1-zinc shows good overall agreement with the computational model, but only three out of four designed histidines coordinate zinc. However, a histidine-to-glutamate point mutation resulted in four-coordination of zinc, and the resulting metal binding site and dimer orientation closely matches the computational model (Cα rmsd = 1.4 Å).


Subject(s)
Drug Design , Protein Multimerization , Proteins/chemistry , Zinc , Models, Molecular , Protein Structure, Quaternary , Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 108(51): 20562-7, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143762

ABSTRACT

Computational design of novel protein-protein interfaces is a test of our understanding of protein interactions and has the potential to allow modification of cellular physiology. Methods for designing high-affinity interactions that adopt a predetermined binding mode have proved elusive, suggesting the need for new strategies that simplify the design process. A solvent-exposed backbone on a ß-strand is thought of as "sticky" and ß-strand pairing stabilizes many naturally occurring protein complexes. Here, we computationally redesign a monomeric protein to form a symmetric homodimer by pairing exposed ß-strands to form an intermolecular ß-sheet. A crystal structure of the designed complex closely matches the computational model (rmsd = 1.0 Å). This work demonstrates that ß-strand pairing can be used to computationally design new interactions with high accuracy.


Subject(s)
Proteins/chemistry , Crystallography, X-Ray/methods , DNA/chemistry , Dimerization , Escherichia coli/metabolism , Light , Molecular Conformation , Protein Conformation , Protein Engineering/methods , Protein Structure, Secondary , Scattering, Radiation , Software , Solvents , Surface Properties , Thermodynamics , Ultracentrifugation
19.
J Biol Chem ; 286(14): 12670-82, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21288910

ABSTRACT

TraI, a bifunctional enzyme containing relaxase and helicase activities, initiates and drives the conjugative transfer of the Escherichia coli F plasmid. Here, we examined the structure and function of the TraI helicase. We show that TraI binds to single-stranded DNA (ssDNA) with a site size of ∼25 nucleotides, which is significantly longer than the site size of other known superfamily I helicases. Low cooperativity was observed with the binding of TraI to ssDNA, and a double-stranded DNA-binding site was identified within the N-terminal region of TraI 1-858, outside the core helicase motifs of TraI. We have revealed that the affinity of TraI for DNA is negatively correlated with the ionic strength of the solution. The binding of AMPPNP or ADP results in a 3-fold increase in the affinity of TraI for ssDNA. Moreover, TraI prefers to bind ssDNA oligomers containing a single type of base. Finally, we elucidated the solution structure of TraI using small angle x-ray scattering. TraI exhibits an ellipsoidal shape in solution with four domains aligning along one axis. Taken together, these data result in the assembly of a model for the multidomain helicase activity of TraI.


Subject(s)
DNA Helicases/metabolism , Escherichia coli Proteins/metabolism , F Factor , Adenosine Diphosphate/metabolism , Adenylyl Imidodiphosphate/metabolism , Binding Sites , Contraindications , DNA/metabolism , DNA Helicases/genetics , DNA, Single-Stranded/metabolism , Escherichia coli Proteins/genetics , Fluorescence Polarization , Protein Binding , Scattering, Small Angle , X-Ray Diffraction
20.
J Biol Chem ; 286(5): 3351-8, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21115486

ABSTRACT

GoLoco motif proteins bind to the inhibitory G(i) subclass of G-protein α subunits and slow the release of bound GDP; this interaction is considered critical to asymmetric cell division and neuro-epithelium and epithelial progenitor differentiation. To provide protein tools for interrogating the precise cellular role(s) of GoLoco motif/Gα(i) complexes, we have employed structure-based protein design strategies to predict gain-of-function mutations that increase GoLoco motif binding affinity. Here, we describe fluorescence polarization and isothermal titration calorimetry measurements showing three predicted Gα(i1) point mutations, E116L, Q147L, and E245L; each increases affinity for multiple GoLoco motifs. A component of this affinity enhancement results from a decreased rate of dissociation between the Gα mutants and GoLoco motifs. For Gα(i1)(Q147L), affinity enhancement was seen to be driven by favorable changes in binding enthalpy, despite reduced contributions from binding entropy. The crystal structure of Gα(i1)(Q147L) bound to the RGS14 GoLoco motif revealed disorder among three peptide residues surrounding a well defined Leu-147 side chain. Monte Carlo simulations of the peptide in this region showed a sampling of multiple backbone conformations in contrast to the wild-type complex. We conclude that mutation of Glu-147 to leucine creates a hydrophobic surface favorably buried upon GoLoco peptide binding, yet the hydrophobic Leu-147 also promotes flexibility among residues 511-513 of the RGS14 GoLoco peptide.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Molecular Dynamics Simulation , Peptides/metabolism , Amino Acid Motifs , Amino Acid Sequence , Computer Simulation , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , Humans , Peptides/chemical synthesis , Protein Binding/genetics , Protein Conformation , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL