Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
MAbs ; 15(1): 2253788, 2023.
Article in English | MEDLINE | ID: mdl-37675979

ABSTRACT

The clinical successes of immune checkpoint blockade have invigorated efforts to activate T cell-mediated responses against cancer. Targeting members of the PVR family, consisting of inhibitory receptors TIGIT, CD96, and CD112R, has been an active area of clinical investigation. In this study, the binding interactions and molecular assemblies of the PVR family receptors and ligands have been assessed in vitro. Furthermore, the anti-TIGIT monoclonal antibody BMS-986207 crystal structure in complex with TIGIT was determined and shows that the antibody binds an epitope that is commonly targeted by the CD155 ligand as well as other clinical anti-TIGIT antibodies. In contrast to previously proposed models, where TIGIT outcompetes costimulatory receptor CD226 for binding to CD155 due to much higher affinity (nanomolar range), our data rather suggest that PVR family members all engage in interactions with relatively weak affinity (micromolar range), including TIGIT and CD155 interactions. Thus, TIGIT and other PVR inhibitory receptors likely elicit immune suppression via increased surface expression rather than inherent differences in affinity. This work provides an improved foundational understanding of the PVR family network and mechanistic insight into therapeutic antibody intervention.


Subject(s)
Neoplasms , Receptors, Immunologic , Humans , T-Lymphocytes/metabolism , Antibodies, Monoclonal/therapeutic use , Ligands
2.
Anal Chem ; 95(8): 3922-3931, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36791402

ABSTRACT

Characterization of antibody binding epitopes is an important factor in therapeutic drug discovery, as the binding site determines and drives antibody pharmacology and pharmacokinetics. Here, we present a novel application of carbene chemical footprinting with mass spectrometry for identification of antibody binding epitopes at the single-residue level. Two different photoactivated diazirine reagents provide complementary labeling information allowing structural refinement of the antibody binding interface. We applied this technique to map the epitopes of multiple MICA and CTLA-4 antibodies and validated the findings with X-ray crystallography and yeast surface display epitope mapping. The characterized epitopes were used to understand biolayer interferometry-derived competitive binding results at the structural level. We show that carbene footprinting provides fast and high-resolution epitope information critical in the antibody selection process and enables mechanistic understanding of function to accelerate the drug discovery process.


Subject(s)
Antibodies , Methane , Epitopes/chemistry , Epitope Mapping/methods
3.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35316223

ABSTRACT

T cell immunoglobulin mucin domain-containing protein 3 (Tim-3) negatively regulates innate and adaptive immunity in cancer. To identify the mechanisms of Tim-3 in cancer immunity, we evaluated the effects of Tim-3 blockade in human and mouse melanoma. Here, we show that human programmed cell death 1-positive (PD-1+) Tim-3+CD8+ tumor-infiltrating lymphocytes (TILs) upregulate phosphatidylserine (PS), a receptor for Tim-3, and acquire cell surface myeloid markers from antigen-presenting cells (APCs) through transfer of membrane fragments called trogocytosis. Tim-3 blockade acted on Tim-3+ APCs in a PS-dependent fashion to disrupt the trogocytosis of activated tumor antigen-specific CD8+ T cells and PD-1+Tim-3+ CD8+ TILs isolated from patients with melanoma. Tim-3 and PD-1 blockades cooperated to disrupt trogocytosis of CD8+ TILs in 2 melanoma mouse models, decreasing tumor burden and prolonging survival. Deleting Tim-3 in dendritic cells but not in CD8+ T cells impeded the trogocytosis of CD8+ TILs in vivo. Trogocytosed CD8+ T cells presented tumor peptide-major histocompatibility complexes and became the target of fratricide T cell killing, which was reversed by Tim-3 blockade. Our findings have uncovered a mechanism Tim-3 uses to limit antitumor immunity.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Melanoma , Animals , CD8-Positive T-Lymphocytes , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Lymphocytes, Tumor-Infiltrating , Melanoma/pathology , Mice , Programmed Cell Death 1 Receptor , Trogocytosis
4.
MAbs ; 14(1): 2024642, 2022.
Article in English | MEDLINE | ID: mdl-35192429

ABSTRACT

Although therapeutically efficacious, ipilimumab can exhibit dose-limiting toxicity that prevents maximal efficacious clinical outcomes and can lead to discontinuation of treatment. We hypothesized that an acidic pH-selective ipilimumab (pH Ipi), which preferentially and reversibly targets the acidic tumor microenvironment over the neutral periphery, may have a more favorable therapeutic index. While ipilimumab has pH-independent CTLA-4 affinity, pH Ipi variants have been engineered to have up to 50-fold enhanced affinity to CTLA-4 at pH 6.0 compared to pH 7.4. In hCTLA-4 knock-in mice, these variants have maintained anti-tumor activity and reduced peripheral activation, a surrogate marker for toxicity. pH-sensitive therapeutic antibodies may be a differentiating paradigm and a novel modality for enhanced tumor targeting and improved safety profiles.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Hydrogen-Ion Concentration , Ipilimumab/therapeutic use , Mice , Therapeutic Index
5.
Anal Chem ; 93(49): 16474-16480, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34854675

ABSTRACT

Identification of antibodies targeting diverse functional epitopes on an antigen is highly crucial for discovering effective therapeutic candidates. Employing a traditional stepwise antibody "screening funnel" as well as prioritizing affinity-based selections over epitope-based selections, result in lead antibody panels lacking epitope diversity. In the present study, we employed an array-based surface plasmon resonance (SPR) platform to perform high-throughput epitope binning analysis on a large number of monoclonal antibodies (mAbs) generated in the early drug discovery process. The mAb panel contained clones from different antibody generation techniques and diverse transgenic mouse strains. The epitope binning results were analyzed in unique ways using various visualizations in the form of dendrograms and network plots, which assisted in determining diversity and redundancy in the mAb sample set. The binning data were further integrated with affinity information to evaluate the performance of seven different transgenic mouse strains. The combination of epitope binning results with binding kinetics and sequence analysis provided an effective and efficient way of selecting high affinity antibodies representing a diverse set of sequence families and epitopes.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents, Immunological , Animals , Epitopes , Mice , Surface Plasmon Resonance
6.
MAbs ; 13(1): 1979800, 2021.
Article in English | MEDLINE | ID: mdl-34595996

ABSTRACT

The molecular interactions of mouse CD96 to CD155 ligand and to two surrogate antibodies have been investigated. Biophysical and structural studies demonstrate that CD96 forms a homodimer but assembles as 1:1 heterodimeric complexes with CD155 or with one of the surrogate antibodies, which compete for the same binding interface. In comparison, the other surrogate antibody binds across the mouse CD96 dimer and recognizes a quaternary epitope spanning both protomers to block exposure of the ligand-binding site. This study reveals different blocking mechanisms and modalities of these two antibodies and may provide insight into the functional effects of antibodies against CD96.


Subject(s)
Antigens, CD , Immunoglobulins , Animals , Antibodies, Blocking , Binding Sites , Mice , Protein Domains
7.
Nat Commun ; 12(1): 1378, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33654081

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) and GITR ligand (GITRL) are members of the tumor necrosis superfamily that play a role in immune cell signaling, activation, and survival. GITR is a therapeutic target for directly activating effector CD4 and CD8 T cells, or depleting GITR-expressing regulatory T cells (Tregs), thereby promoting anti-tumor immune responses. GITR activation through its native ligand is important for understanding immune signaling, but GITR structure has not been reported. Here we present structures of human and mouse GITR receptors bound to their cognate ligands. Both species share a receptor-ligand interface and receptor-receptor interface; the unique C-terminal receptor-receptor enables higher order structures on the membrane. Human GITR-GITRL has potential to form a hexameric network of membrane complexes, while murine GITR-GITRL complex forms a linear chain due to dimeric interactions. Mutations at the receptor-receptor interface in human GITR reduce cell signaling with in vitro ligand binding assays and minimize higher order membrane structures when bound by fluorescently labeled ligand in cell imaging experiments.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/chemistry , Tumor Necrosis Factors/metabolism , Animals , Biophysical Phenomena , Cell Line , Cell Membrane/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Mice , Models, Molecular , Protein Binding , Reproducibility of Results , Tumor Necrosis Factors/chemistry
8.
J Am Soc Mass Spectrom ; 32(7): 1567-1574, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-33415981

ABSTRACT

NK group 2 member A (NKG2A), an immune checkpoint inhibitor, is an emerging therapeutic target in immuno-oncology. NKG2A forms a heterodimer with CD94 on the cell surface of NK and a subset of T cells and recognizes the nonclassical human leukocyte antigen (HLA-E) in humans. Therapeutic blocking antibodies that block the ligation between HLA-E and NKG2A/CD94 have been shown to enhance antitumor immunity in mice and humans. In this study, we illustrate the practical utilities of mass spectrometry (MS)-based protein footprinting in areas from reagent characterization to antibody epitope mapping. Hydrogen/deuterium exchange mass spectrometry (HDX-MS) in the higher-order structure characterization of NKG2A in complex with CD94 provides novel insights into the conformational dynamics of NKG2A/CD94 heterodimer. To fully understand antibody/target interactions, we employed complementary protein footprinting methods, including HDX-MS and fast photochemical oxidation of proteins (FPOP)-MS, to determine the binding epitopes of therapeutic monoclonal antibodies targeting NKG2A. Such a combination approach provides molecular insights into the binding mechanisms of antibodies to NKG2A with high specificity, demonstrating the blockade of NKG2A/HLA-E interaction.


Subject(s)
Antibodies , Hydrogen Deuterium Exchange-Mass Spectrometry/methods , NK Cell Lectin-Like Receptor Subfamily C , NK Cell Lectin-Like Receptor Subfamily D , Protein Footprinting/methods , Antibodies/chemistry , Antibodies/metabolism , Epitope Mapping , Epitopes , Humans , NK Cell Lectin-Like Receptor Subfamily C/chemistry , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/chemistry , NK Cell Lectin-Like Receptor Subfamily D/metabolism
9.
MAbs ; 12(1): 1685350, 2020.
Article in English | MEDLINE | ID: mdl-31856660

ABSTRACT

The development of antibody therapeutics relies on animal models that accurately recapitulate disease biology. Syngeneic mouse models are increasingly used with new molecules to capture the biology of complex cancers and disease states, and to provide insight into the role of the immune system. The establishment of syngeneic mouse models requires the ability to generate surrogate mouse counterparts to antibodies designed for humans. In the field of bispecific antibodies, there remains a dearth of technologies available to generate native IgG-like mouse bispecific antibodies. Thus, we engineered a simple co-expression system for one-step purification of intact mouse IgG1 and IgG2a bispecific antibodies from any antibody pair. We demonstrated proof of concept with CD3/CD20 bispecific antibodies, which highlighted both the quality and efficacy of materials generated by this technology.


Subject(s)
Antibodies, Bispecific/genetics , Immunoglobulin G/genetics , Protein Engineering/methods , Rituximab/metabolism , T-Lymphocytes/metabolism , Animals , Antibodies, Bispecific/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , CHO Cells , Cloning, Molecular , Cricetulus , Disease Models, Animal , Immunoglobulin G/metabolism , Mice , Protein Binding , Protein Conformation , T-Lymphocytes/immunology , Transplantation, Isogeneic
11.
Oncoimmunology ; 6(9): e1339853, 2017.
Article in English | MEDLINE | ID: mdl-28932638

ABSTRACT

Elotuzumab is a humanized therapeutic monoclonal antibody directed to the surface glycoprotein SLAMF7 (CS1, CRACC, CD319), which is highly expressed on multiple myeloma (MM) tumor cells. Improved clinical outcomes have been observed following treatment of MM patients with elotuzumab in combination with lenalidomide or bortezomib. Previous work showed that elotuzumab stimulates NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC), via Fc-domain engagement with FcγRIIIa (CD16). SLAMF7 is also expressed on NK cells, where it can transmit stimulatory signals. We tested whether elotuzumab can directly activate NK cells via ligation with SLAMF7 on NK cells in addition to targeting ADCC through CD16. We show that elotuzumab strongly promoted degranulation and activation of NK cells in a CD16-dependent manner, and a non-fucosylated form of elotuzumab with higher affinity to CD16 exhibited enhanced potency. Using F(ab')2 or Fc-mutant forms of the antibody, the direct binding of elotuzumab to SLAMF7 alone could not stimulate measurable CD69 expression or degranulation of NK cells. However, the addition of soluble elotuzumab could costimulate calcium signaling responses triggered by multimeric engagement of NKp46 and NKG2D in a CD16-independent manner. Thus, while elotuzumab primarily stimulates NK cells through CD16, it can also transduce effective "trans"-costimulatory signals upon direct engagement with SLAMF7, since these responses did not require direct co-engagement with the activating receptors. Trans-costimulation by elotuzumab has potential to reduce activation thresholds of other NK cell receptors engaging with their ligands on myeloma target cell surfaces, thereby potentially further increasing NK cell responsiveness in patients.

12.
Bioconjug Chem ; 28(4): 1102-1114, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28151644

ABSTRACT

Antibody drug conjugates (ADCs) provide an efficacious and relatively safe means by which chemotherapeutic agents can be specifically targeted to cancer cells. In addition to the selection of antibody targets, ADCs offer a modular design that allows selection of ADC characteristics through the choice of linker chemistries, toxins, and conjugation sites. Many studies have indicated that release of toxins bound to antibodies via noncleavable linker chemistries relies on the internalization and intracellular trafficking of the ADC. While this can make noncleavable ADCs more stable in the serum, it can also result in lower efficacy when their respective targets are not internalized efficiently or are recycled back to the cell surface following internalization. Here, we show that a lysosomally targeted ADC against the protein APLP2 mediates cell killing, both in vitro and in vivo, more effectively than an ADC against Trop2, a protein with less efficient lysosomal targeting. We also engineered a bispecific ADC with one arm targeting HER2 for the purpose of directing the ADC to tumors, and the other arm targeting APLP2, whose purpose is to direct the ADC to lysosomes for toxin release. This proof-of-concept bispecific ADC demonstrates that this technology can be used to shift the intracellular trafficking of a constitutively recycled target by directing one arm of the antibody against a lysosomally delivered protein. Our data also show limitations of this approach and potential future directions for development.


Subject(s)
Drug Delivery Systems , Immunoconjugates/pharmacology , Lysosomes/metabolism , Transcytosis , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/metabolism , Mice, Nude , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/therapeutic use , Receptor, ErbB-2/immunology , Receptor, ErbB-2/therapeutic use
13.
J Mol Biol ; 427(6 Pt B): 1513-1534, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25284753

ABSTRACT

The ability of antibodies to bind an antigen with a high degree of affinity and specificity has led them to become the largest and fastest growing class of therapeutic proteins. Clearly identifying the epitope at which they bind their cognate antigen provides insight into their mechanism of action and helps differentiate antibodies that bind the same antigen. Here, we describe a method to precisely and efficiently map the epitopes of a panel of antibodies in parallel over the course of several weeks. This method relies on the combination of rational library design, quantitative yeast surface display and next-generation DNA sequencing and was demonstrated by mapping the epitopes of several antibodies that neutralize alpha toxin from Staphylococcus aureus. The accuracy of this method was confirmed by comparing the results to the co-crystal structure of one antibody and alpha toxin and was further refined by the inclusion of a lower-affinity variant of the antibody. In addition, this method produced quantitative insight into the epitope residues most critical for the antibody-antigen interaction and enabled the relative affinities of each antibody toward alpha toxin variants to be estimated. This affinity estimate serves as a predictor of neutralizing antibody potency and was used to anticipate the ability of each antibody to effectively bind and neutralize naturally occurring alpha toxin variants secreted by strains of S. aureus, including clinically relevant strains. Ultimately this type information can be used to help select the best clinical candidate among a set of antibodies against a given antigen.


Subject(s)
Antibodies, Monoclonal/immunology , Bacterial Toxins/immunology , Epitopes/analysis , Hemolysin Proteins/immunology , High-Throughput Nucleotide Sequencing , Peptide Library , Saccharomyces cerevisiae/immunology , Staphylococcal Infections/prevention & control , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Bacterial Toxins/genetics , Epitope Mapping/methods , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Flow Cytometry , Hemolysin Proteins/genetics , Humans , Molecular Sequence Data , Protein Conformation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology
14.
PLoS One ; 8(11): e80501, 2013.
Article in English | MEDLINE | ID: mdl-24223227

ABSTRACT

Monoclonal antibodies (mAbs) are a growing segment of therapeutics, yet their in vitro characterization remains challenging. While it is essential that a therapeutic mAb recognizes the native, physiologically occurring epitope, the generation and selection of mAbs often rely on the use of purified recombinant versions of the antigen that may display non-native epitopes. Here, we present a method to measure both, the binding affinity of a therapeutic mAb towards its native unpurified antigen in human serum, and the antigen's endogenous concentration, by combining the kinetic exclusion assay and Biacore's calibration free concentration analysis. To illustrate the broad utility of our method, we studied a panel of mAbs raised against three disparate soluble antigens that are abundant in the serum of healthy donors: proprotein convertase subtilisin/kexin type 9 (PCSK9), progranulin (PGRN), and fatty acid binding protein (FABP4). We also determined the affinity of each mAb towards its purified recombinant antigen and assessed whether the interactions were pH-dependent. Of the six mAbs studied, three did not appear to discriminate between the serum and recombinant forms of the antigen; one mAb bound serum antigen with a higher affinity than recombinant antigen; and two mAbs displayed a different affinity for serum antigen that could be explained by a pH-dependent interaction. Our results highlight the importance of taking pH into account when measuring the affinities of mAbs towards their serum antigens, since the pH of serum samples becomes increasingly alkaline upon aerobic handling.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens/blood , Antigens/metabolism , Cell Line , Fatty Acid-Binding Proteins/blood , Fatty Acid-Binding Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/metabolism , Progranulins , Proprotein Convertase 9 , Proprotein Convertases/blood , Proprotein Convertases/metabolism , Serine Endopeptidases/blood , Serine Endopeptidases/metabolism
15.
J Mol Biol ; 425(10): 1641-54, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23416200

ABSTRACT

The emergence and spread of multi-drug-resistant strains of Staphylococcus aureus in hospitals and in the community emphasize the urgency for the development of novel therapeutic interventions. Our approach was to evaluate the potential of harnessing the human immune system to guide the development of novel therapeutics. We explored the role of preexisting antibodies against S. aureus α-hemolysin in the serum of human individuals by isolating and characterizing one antibody with a remarkably high affinity to α-hemolysin. The antibody provided protection in S. aureus pneumonia, skin, and bacteremia mouse models of infection and also showed therapeutic efficacy when dosed up to 18 h post-infection in the pneumonia model. Additionally, in pneumonia and bacteremia animal models, the therapeutic efficacy of the α-hemolysin antibody appeared additive to the antibiotic linezolid. To better understand the mechanism of action of this isolated antibody, we solved the crystal structure of the α-hemolysin:antibody complex. To our knowledge, this is the first report of the crystal structure of the α-hemolysin monomer. The structure of the complex shows that the antibody binds α-hemolysin between the cap and the rim domains. In combination with biochemical data, the structure suggests that the antibody neutralizes the activity of the toxin by preventing binding to the plasma membrane of susceptible host cells. The data presented here suggest that protective antibodies directed against S. aureus molecules exist in some individuals and that such antibodies have a therapeutic potential either alone or in combination with antibiotics.


Subject(s)
Antibodies, Bacterial/administration & dosage , Antibodies, Bacterial/therapeutic use , Antigen-Antibody Complex/administration & dosage , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/pharmacology , Antibodies, Blocking/blood , Antibodies, Blocking/chemistry , Antibodies, Blocking/therapeutic use , Antigen-Antibody Complex/chemistry , Antigen-Antibody Reactions/immunology , Bacterial Toxins/chemistry , Crystallography, X-Ray , Disease Models, Animal , Female , Hemolysin Proteins/chemistry , Humans , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Staphylococcal Infections/blood , Staphylococcus aureus/pathogenicity
16.
PLoS One ; 7(4): e36261, 2012.
Article in English | MEDLINE | ID: mdl-22558410

ABSTRACT

Therapeutic antibodies are often engineered or selected to have high on-target binding affinities that can be challenging to determine precisely by most biophysical methods. Here, we explore the dynamic range of the kinetic exclusion assay (KinExA) by exploiting the interactions of an anti-DKK antibody with a panel of DKK antigens as a model system. By tailoring the KinExA to each studied antigen, we obtained apparent equilibrium dissociation constants (K(D) values) spanning six orders of magnitude, from approximately 100 fM to 100 nM. Using a previously calibrated antibody concentration and working in a suitable concentration range, we show that a single experiment can yield accurate and precise values for both the apparent K(D) and the apparent active concentration of the antigen, thereby increasing the information content of an assay and decreasing sample consumption. Orthogonal measurements obtained on Biacore and Octet label-free biosensor platforms further validated our KinExA-derived affinity and active concentration determinations. We obtained excellent agreement in the apparent affinities obtained across platforms and within the KinExA method irrespective of the assay orientation employed or the purity of the recombinant or native antigens.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity , Biological Assay/methods , Intercellular Signaling Peptides and Proteins/immunology , Animals , Biosensing Techniques , Calibration , Cell Line, Tumor , Humans , Mice , Rats
17.
J Biol Chem ; 285(21): 16258-66, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20339001

ABSTRACT

NORE1A is a Ras-binding protein that belongs to a group of tumor suppressors known as the Ras association domain family. Their growth- and tumor-suppressive function is assumed to be dependent on association with the microtubule cytoskeleton. However, a detailed understanding of this interplay is still missing. Here, we show that NORE1A directly interacts with tubulin and is capable of nucleating microtubules. Strikingly, the ability to stimulate nucleation is regulated in a dual specific way either via phosphorylation of NORE1A within the Ras-binding domain by Aurora A kinase or via binding to activated Ras. We also demonstrate that NORE1A mediates a negative effect of activated Ras on microtubule nucleation. On the basis of our results, we propose a novel regulatory network composed of the tumor suppressor NORE1A, the mitotic kinase Aurora A, the small GTPase Ras, and the microtubule cytoskeleton.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Microtubules/metabolism , Monomeric GTP-Binding Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Aurora Kinase A , Aurora Kinases , Cell Line , Humans , Mice , Microtubules/genetics , Monomeric GTP-Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tubulin/genetics , Tubulin/metabolism , Tumor Suppressor Proteins/genetics , ras Proteins/genetics
18.
Bioconjug Chem ; 19(9): 1938-44, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18712896

ABSTRACT

Many biological processes take place in close proximity to lipid membranes. For a detailed understanding of the underlying mechanisms, tools are needed for the quantitative characterization of such biomolecular interactions. In this work, we describe the development of methods addressing the dynamics and affinities of protein complexes attached to an artificial membrane system. A semisynthetic approach provides the Ras protein with palmitoyl anchors, which allow stable membrane insertion, as a paradigm for membrane associated proteins that interact with multiple effectors. An artificial membrane system is constituted by nanoparticles covered with a lipid bilayer. Such a stable suspension allows for the characterization of the interaction between membrane-bound Ras and effector proteins using conventional fluorescence-based methods.


Subject(s)
Biosensing Techniques/methods , Lipid Bilayers/chemistry , Membranes, Artificial , Nanoparticles/chemistry , ras Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Phosphatidylglycerols/chemistry , Scattering, Radiation , Spectrometry, Fluorescence/methods , ras Proteins/chemistry
19.
EMBO J ; 27(14): 1995-2005, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18596699

ABSTRACT

A class of putative Ras effectors called Ras association domain family (RASSF) represents non-enzymatic adaptors that were shown to be important in tumour suppression. RASSF5, a member of this family, exists in two splice variants known as NORE1A and RAPL. Both of them are involved in distinct cellular pathways triggered by Ras and Rap, respectively. Here we describe the crystal structure of Ras in complex with the Ras binding domain (RBD) of NORE1A/RAPL. All Ras effectors share a common topology in their RBD creating an interface with the switch I region of Ras, whereas NORE1A/RAPL RBD reveals additional structural elements forming a unique Ras switch II binding site. Consequently, the contact area of NORE1A is extended as compared with other Ras effectors. We demonstrate that the enlarged interface provides a rationale for an exceptionally long lifetime of the complex. This is a specific attribute characterizing the effector function of NORE1A/RAPL as adaptors, in contrast to classical enzymatic effectors such as Raf, RalGDS or PI3K, which are known to form highly dynamic short-lived complexes with Ras.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Molecular Sequence Data , Monomeric GTP-Binding Proteins/genetics , Mutagenesis , Protein Interaction Domains and Motifs , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...