19 results on '"Dora Toledo Warshaviak"'
Search Results
2. The integration of pharmacophore-based 3D QSAR modeling and virtual screening in safety profiling: A case study to identify antagonistic activities against adenosine receptor, A2A, using 1,897 known drugs.
- Author
-
Fan Fan, Dora Toledo Warshaviak, Hisham K Hamadeh, and Robert T Dunn
- Subjects
Medicine ,Science - Abstract
Safety pharmacology screening against a wide range of unintended vital targets using in vitro assays is crucial to understand off-target interactions with drug candidates. With the increasing demand for in vitro assays, ligand- and structure-based virtual screening approaches have been evaluated for potential utilization in safety profiling. Although ligand based approaches have been actively applied in retrospective analysis or prospectively within well-defined chemical space during the early discovery stage (i.e., HTS screening and lead optimization), virtual screening is rarely implemented in later stage of drug discovery (i.e., safety). Here we present a case study to evaluate ligand-based 3D QSAR models built based on in vitro antagonistic activity data against adenosine receptor 2A (A2A). The resulting models, obtained from 268 chemically diverse compounds, were used to test a set of 1,897 chemically distinct drugs, simulating the real-world challenge of safety screening when presented with novel chemistry and a limited training set. Due to the unique requirements of safety screening versus discovery screening, the limitations of 3D QSAR methods (i.e., chemotypes, dependence on large training set, and prone to false positives) are less critical than early discovery screen. We demonstrated that 3D QSAR modeling can be effectively applied in safety assessment prior to in vitro assays, even with chemotypes that are drastically different from training compounds. It is also worth noting that our model is able to adequately make the mechanistic distinction between agonists and antagonists, which is important to inform subsequent in vivo studies. Overall, we present an in-depth analysis of the appropriate utilization and interpretation of pharmacophore-based 3D QSAR models for safety screening.
- Published
- 2019
- Full Text
- View/download PDF
3. HLA-A∗02-gated safety switch for cancer therapy has exquisite specificity for its allelic target antigen
- Author
-
Jee-Young Mock, Aaron Winters, Timothy P. Riley, Richele Bruno, Martin S. Naradikian, Shruti Sharma, Claudia A. Jette, Ryan Elshimali, Casey Gahrs, Dora Toledo-Warshaviak, Anthony P. West, Jr., Alexander Kamb, and Agnes E. Hamburger
- Subjects
Tmod ,logic gate ,selectivity ,CAR ,cell therapy ,structure ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Innovative cell-based therapies are important new weapons in the fight against difficult-to-treat cancers. One promising strategy involves cell therapies equipped with multiple receptors to integrate signals from more than one antigen. We developed a specific embodiment of this approach called Tmod, a two-receptor system that combines activating and inhibitory inputs to distinguish between tumor and normal cells. The selectivity of Tmod is enforced by the inhibitory receptor (blocker) that recognizes an antigen, such as an HLA allele, whose expression is absent from tumors because of loss of heterozygosity. Although unwanted cross-reactivity of the blocker likely reduces efficacy rather than safety, it is important to verify the blocker’s specificity. We have tested an A∗02-directed blocker derived from the PA2.1 mouse antibody as a safety mechanism paired with a mesothelin-specific activating CAR in our Tmod construct. We solved the crystal structure of humanized PA2.1 Fab in complex with HLA-A∗02 to determine its binding epitope, which was used to bioinformatically select specific class I HLA alleles to test the blocker’s functional specificity in vitro. We found that this A∗02-directed blocker is highly specific for its cognate antigen, with only one cross-reactive allele (A∗69) capable of triggering comparable function.
- Published
- 2022
- Full Text
- View/download PDF
4. Supplementary Data from Chimeric Antigen Receptors Directed at Mutant KRAS Exhibit an Inverse Relationship Between Functional Potency and Neoantigen Selectivity
- Author
-
Agnes E. Hamburger, Alexander Kamb, Dora Toledo Warshaviak, Aaron D. Martin, Mark E. Daris, Jee-Young Mock, Martin S. Naradikian, Grace E. Asuelime, and Talar Tokatlian
- Abstract
S.Figures 1-5 and S.Table 1
- Published
- 2023
5. Chimeric Antigen Receptors Directed at Mutant KRAS Exhibit an Inverse Relationship Between Functional Potency and Neoantigen Selectivity
- Author
-
Talar Tokatlian, Grace E. Asuelime, Martin S. Naradikian, Jee-Young Mock, Mark E. Daris, Aaron D. Martin, Dora Toledo Warshaviak, Alexander Kamb, and Agnes E. Hamburger
- Abstract
Neoantigens are among the most intriguing potential immuno-oncology targets because, unlike many cancer targets that are expressed on normal tissues, they are by definition restricted to cancer cells. Medicines directed at common neoantigens such as mutant KRAS are especially interesting because they may offer the convenience and cost of an off-the-shelf therapy. However, all common KRAS mutations produce proteins that differ from the wild type at a single amino acid, creating challenges for molecular discrimination. We have undertaken an effort to optimize single-chain variable fragments (scFv) against peptide/major histocompatibility antigen complexes composed of HLA-A*11 and either G12V- or G12D-mutant KRAS peptides. These scFvs could in principle be used in chimeric antigen receptor (CAR) T-cell therapies for selected patients whose tumors bear either of these mutations. Here we show that optimization of such CARs involves a trade-off between potency and selectivity. We further show that targeting this family without high selectivity engenders risks of cross-reactivity against other members of the G-protein family to which KRAS belongs.Significance:We report an effort to generate high potency, selective CARs directed at mutant KRAS peptides. Although the heavily optimized CARs maintain high selectivity against wild-type KRAS, they lose selectivity against other KRAS-related peptides derived from human proteins. To our knowledge, this work is the first to examine the trade-off between potency and selectivity with regard to KRAS pMHC-directed CARs, illustrating the challenge to achieve both sufficient potency and high selectivity.
- Published
- 2022
6. Extensive functional comparisons between chimeric antigen receptors and T cell receptors highlight fundamental similarities
- Author
-
Mark L. Sandberg, Yuta Ando, Alexander Kamb, Ming-Lun Wu, Kathleen R Negri, Wen-Hua Lee, Julyun Oh, Xueyin Wang, Michele McElvain, Grant B Gabrelow, Aaron D. Martin, Han Xu, and Dora Toledo Warshaviak
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,T cell ,Immunology ,T-cell receptor ,Receptors, Antigen, T-Cell ,hemic and immune systems ,chemical and pharmacologic phenomena ,Context (language use) ,Biology ,Lymphocyte Activation ,Chimeric antigen receptor ,Immune system ,medicine.anatomical_structure ,medicine ,Humans ,Receptor ,Molecular Biology ,Neuroscience ,CD80 ,Function (biology) - Abstract
Though TCRs have been subject to limited engineering in the context of therapeutic design and optimization, they are used largely as found in nature. On the other hand, CARs are artificial, composed of different segments of proteins that function in the immune system. This characteristic raises the possibility of altered response to immune regulatory stimuli. Here we describe a large-scale, systematic comparison of CARs and TCRs across 5 different pMHC targets, with a total of 19 constructs examined in vitro. These functional measurements include CAR- and TCR-mediated activation, proliferation, and cytotoxicity in both acute and chronic settings. Surprisingly, we find no consistent difference between CARs and TCRs as receptor classes with respect to their relative sensitivity to major regulators of T cell activation: PD-L1, CD80/86 and IL-2. Though TCRs often emerge from human blood directly as potent, selective receptors, CARs must be heavily optimized to attain these properties for pMHC targets. Nonetheless, when iteratively improved and compared head to head in functional tests, CARs appear remarkably similar to TCRs with respect to immune modulation.
- Published
- 2021
7. Potent, Selective CARs as Potential T-Cell Therapeutics for HPV-positive Cancers
- Author
-
Kathleen R Negri, Xueyin Wang, Han Xu, Mark E. Daris, Aaron D. Martin, Craig Pigott, Falene Chai, Christine Yao, Ming-Lun Wu, James Furney, Dora Toledo Warshaviak, Grant B Gabrelow, Daniel P Nampe, Mark L. Sandberg, Alexander Kamb, Wen-Hua Lee, Julyun Oh, and Michele McElvain
- Subjects
Cancer Research ,Papillomavirus E7 Proteins ,T cell ,Green Fluorescent Proteins ,Immunology ,Basic Studies ,Context (language use) ,Immunotherapy, Adoptive ,Cell Line ,head and neck ,Interferon-gamma ,Luciferases, Firefly ,Neoplasms ,HLA-A2 Antigen ,medicine ,Humans ,Immunology and Allergy ,Avidity ,Receptor ,Pharmacology ,Receptors, Chimeric Antigen ,biology ,Jurkat ,Chemistry ,Papillomavirus Infections ,Oncogene Proteins, Viral ,Chimeric antigen receptor ,In vitro ,Repressor Proteins ,bifunctional ,medicine.anatomical_structure ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,biology.protein ,Cancer research ,cytotoxicity ,cell therapy ,Antibody ,Peptides ,optimization ,TCR ,Function (biology) ,Single-Chain Antibodies - Abstract
Supplemental Digital Content is available in the text., Next-generation T-cell therapies will likely continue to utilize T-cell receptors (TCRs) and chimeric antigen receptors (CARs) because each receptor type has advantages. TCRs often possess exceptional properties even when tested unmodified from patients’ T cells. CARs are generally less sensitive, possibly because their ligand-binding domains are grafted from antibodies selected for binding affinity or avidity and not broadly optimized for a functional response. Because of the disconnect between binding and function among these receptor types, the ultimate potential of CARs optimized for sensitivity and selectivity is not clear. Here, we focus on a thoroughly studied immuno-oncology target, the HLA-A*02/HPV-E629–38 complex, and show that CARs can be optimized by a combination of high-throughput binding screens and low-throughput functional assays to have comparable activity to clinical TCRs in acute assays in vitro. These results provide a case study for the challenges and opportunities of optimizing high-performing CARs, especially in the context of targets utilized naturally by TCRs.
- Published
- 2021
8. Mesothelin-specific CAR-T cell therapy that incorporates an HLA-gated safety mechanism selectively kills tumor cells
- Author
-
Talar Tokatlian, Grace E Asuelime, Jee-Young Mock, Breanna DiAndreth, Shruti Sharma, Dora Toledo Warshaviak, Mark E Daris, Kristian Bolanos, Breanna L Luna, Martin S Naradikian, Kiran Deshmukh, Agnes E Hamburger, and Alexander Kamb
- Subjects
Cancer Research ,T-Lymphocytes ,Immunology ,receptors ,Loss of Heterozygosity ,cell engineering ,Immunotherapy, Adoptive ,Mice ,Cell Line, Tumor ,Neoplasms ,HLA-A2 Antigen ,Immunology and Allergy ,Animals ,Humans ,immunologic ,RC254-282 ,Pharmacology ,Receptors, Chimeric Antigen ,Immune Cell Therapies and Immune Cell Engineering ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xenograft Model Antitumor Assays ,Oncology ,Mesothelin ,Molecular Medicine ,Female ,immunotherapy - Abstract
BackgroundMesothelin (MSLN) is a classic tumor-associated antigen that is expressed in lung cancer and many other solid tumors. However, MSLN is also expressed in normal mesothelium which creates a significant risk of serious inflammation for MSLN-directed therapeutics. We have developed a dual-receptor (Tmod™) system that exploits the difference between tumor and normal tissue in a subset of patients with defined heterozygous gene loss (LOH) in their tumors.MethodsT cells engineered with the MSLN CAR Tmod construct described here contain (1) a novel MSLN-activated CAR and (2) an HLA-A*02-gated inhibitory receptor (blocker). A*02 binding is intended to override T-cell cytotoxicity, even in the presence of MSLN. The Tmod system is designed to treat heterozygous HLA class I patients, selected for HLA LOH. When A*02 is absent from tumors selected for LOH, the MSLN Tmod cells are predicted to mediate potent killing of the MSLN(+)A*02(−) malignant cells.ResultsThe sensitivity of the MSLN Tmod cells is comparable with a benchmark MSLN CAR-T that was active but toxic in the clinic. Unlike MSLN CAR-T cells, the Tmod system robustly protects surrogate “normal” cells even in mixed-cell populations in vitro and in a xenograft model. The MSLN CAR can also be paired with other HLA class I blockers, supporting extension of the approach to patients beyond A*02 heterozygotes.ConclusionsThe Tmod mechanism exemplified by the MSLN CAR Tmod construct provides an alternative route to leverage solid-tumor antigens such as MSLN in safer, more effective ways than previously possible.
- Published
- 2022
9. 125 Reexamination of MAGE-A3 as a T-cell Therapeutic Target
- Author
-
Aaron D. Martin, Mark L. Sandberg, Grant B Gabrelow, Alexander Kamb, Xueyin Wang, Bella Lee, Ming Wu, Mark E. Daris, Han Xu, Michele McElvain, Dora Toledo Warshaviak, and Kathleen R Negri
- Subjects
0301 basic medicine ,endocrine system ,Genetic enhancement ,T cell ,Cell ,T-cell receptor ,Cancer ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,030220 oncology & carcinogenesis ,Toxicity ,medicine ,Cancer research ,Significant risk ,neoplasms - Abstract
Background Recurrent cancer-specific targets are rare. Given the pace of genomic research over the past three decades, few are likely to lie yet undiscovered. In 2013 an innovative MAGE-A3-directed cancer therapeutic of great potential value was terminated in the clinic because of neurotoxicity.1 The safety problems were hypothesized to originate from off-target TCR activity against a closely related MAGE-A12 peptide. Methods A combination of published and new data led us to test this hypothesis with current technology, including RNA hybridization in situ and further analysis of the clinical TCR’s specificity to MAGE-A12 and other antigens. Results We find that a key prediction of the MAGE-A12 toxicity hypothesis, the existence of rare, high-MAGE-A12-expressing cells in the brain, is not supported by the data. Our results imply that an alternative related peptide from the EPS8L2 protein is more likely responsible for the toxicity. Therefore, it may be valuable to reconsider MAGE-A3 as a cancer target using HLA-A*02-restricted-TCRs or CARs. As a step in this direction, we isolated MAGE-A3 pMHC-directed CARs, targeting the same peptide as the clinical TCR. These CARs have high selectivity, and avoid cross-reaction with the EPS8L2 peptide that represents a significant risk for MAGE-A3-targeted therapeutics. Conclusions Given the qualities of MAGE-A3 as an onco-testis antigen widely expressed in tumors and largely absent from normal adult tissues, our findings suggest that MAGE-A3 may deserve further consideration as a cancer target. We have identified CARs with selectivity profiles consistent with a cell therapeutic directed against HLA-A*02-positive, MAGE-A3-expressing cancers. The relative merits of TCRs and CARs for this target will be discussed. Reference Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, Dudley ME, Feldman SA, Yang JC, Sherry RM, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 2013;36:133–151, doi:10.1097/CJI.0b013e3182829903.
- Published
- 2020
10. A rational approach to assess off-target reactivity of a dual-signal integrator for T cell therapy
- Author
-
Xueyin, Wang, Lu Min, Wong, Michele E, McElvain, Sara, Martire, Wen-Hua, Lee, Chuck Z, Li, Fernando A, Fisher, Ruchika L, Maheshwari, Ming Lun, Wu, Maria C, Imun, Rabi, Murad, Dora, Toledo Warshaviak, Jun, Yin, Alexander, Kamb, and Han, Xu
- Subjects
Pharmacology ,Gene Expression Regulation ,Cell Line, Tumor ,T-Lymphocytes ,Antigens, CD19 ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,Computational Biology ,Humans ,RNA, Messenger ,Toxicology ,Gene Deletion - Abstract
Cell therapy is an emerging therapeutic modality with the power to exploit new cancer targets and potentially achieve positive outcomes for patients with few other options. Like all synthetic treatments, cell therapy has the risk of toxicity via unpredicted off-target behavior. We describe an empirical method to model off-tumor, off-target reactivity of receptors used for investigational T cell therapies. This approach utilizes an optimal panel of diverse human cell-lines to capture the large majority of protein-coding gene expression in adult human tissues. We apply this cell-line set to test Jurkat and primary T cells engineered with a dual-signal integrator, called Tmod
- Published
- 2022
11. 122 A powerful, precise targeting system controlled by tumor deletions transforms CEA and MSLN CAR-T cells into tumor-selective agents
- Author
-
Maria Imun, Mark L. Sandberg, Yuta Ando, Wen-Hua Lee, Xueyin Wang, Fernando Fisher, Michele McElvain, Kiran Deshmukh, Agnes E. Hamburger, Han Xu, Lu-Min Wong, Chuck Li, Breanna DiAndreth, Alexander Kamb, David M.C. Ju, Grant B Gabrelow, Dora Toledo-Warshaviak, Sanam Shafaattalab, Daniel P Nampe, Talar Tokatlian, Mark E. Daris, Shruti Sharma, Edwin Liu, Jee-Young Mock, Martin S. Naradikian, Kristian Bolanos-Ibarra, Aaron D. Martin, and Grace E. Asuelime
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Chemistry ,Immunology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Car t cells - Abstract
BackgroundMesothelin (MSLN) and carcinoembryonic antigen (CEA) are classic tumor-associated antigens that are expressed in many solid tumors including the majority of lung, colorectal and pancreatic cancers. However, both MSLN and CEA are also expressed in vital normal organs. This normal expression creates risk of serious inflammation for CEA- or MSLN-directed therapeutics. To date all active CEA- or MSLN-targeted investigational therapeutics have been toxic when administered systemically.MethodsWe have developed a safety mechanism to protect normal tissues without abrogating sensitivity of cytotoxic T cells directed at MLSN(+) or CEA(+) tumors in a subset of patients with defined loss of heterozygosity (LOH) in their tumors (figure 1). This dual-receptor (Tmod< sup >TM) system exploits common LOH at the HLA locus in cancer cells, allowing T cells to recognize the difference between tumor and normal tissue.1 2 T cells engineered with specific Tmod constructs contain: (i) a MSLN- or CEA-activated CAR; and, (ii) an inhibitory receptor gated by HLA-A*02. HLA-A*02 binding blocks T cell cytotoxicity, even in the presence of MSLN or CEA. The Tmod system is designed to treat heterozygous HLA class I patients, selected for HLA LOH. When HLA-A*02 is absent from tumors selected for LOH, the CARs are predicted to mediate potent killing of the A*02(-) malignant cells.ResultsThe Tmod system robustly protects surrogate normal cells even in mixed-cell populations in vitro while mediating robust cytotoxicity of tumor cells in xenograft models (see example in figure 2). The MSLN CAR can also be paired with other blockers, supporting scalability of the approach to patients beyond HLA-A*02 heterozygotes.Abstract 122 Figure 1Illustration of the Tmod T cell engaging with tumor cells with somatic loss of HLA-A*02 and with normal cells.Abstract 122 Figure 2Bioluminescence measurements show the average difference between the size of the MSLN(+)A*02(+) ‘normal’ graft compared to the MSLN(+)A*02(-) tumor graft on the two flanks of mice after T cell infusion. Both tumor and normal grafts are destroyed by CAR-Ts (CAR-3 and M5 benchmark) while the MSLN Tmod cells kill the tumor but not the normal graft.ConclusionsThe Tmod mechanism may provide an alternative route to leverage solid-tumor antigens such as MSLN and CEA in safer, more effective ways than previously possible.ReferencesHamburger AE, DiAndreth B, Cui J, et al. Engineered T cells directed at tumors with defined allelic loss. Mol Immunol 2020;128:298–310.Hwang MS, Mog BJ, Douglass J, et al. Targeting loss of heterozygosity for cancer-specific immunotherapy. Proc Natl Acad Sci U S A 2021;118(12):e2022410118.
- Published
- 2021
12. Single variable domains from the T cell receptor β chain function as mono- and bifunctional CARs and TCRs
- Author
-
Jaspal Singh Kang, Abby Lin, Julyun Oh, Alexander Kamb, Craig Pigott, Falene Chai, Michael Gallo, Melanie L. Munguia, Mikayel Mkrtichyan, and Dora Toledo Warshaviak
- Subjects
0301 basic medicine ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,T cell ,Primary Cell Culture ,Immunoglobulin Variable Region ,lcsh:Medicine ,chemical and pharmacologic phenomena ,Cancer immunotherapy ,Computational biology ,Gene delivery ,Ligands ,Transfection ,Immunotherapy, Adoptive ,Jurkat cells ,Article ,Jurkat Cells ,03 medical and health sciences ,Applied immunology ,0302 clinical medicine ,Antigen ,Neoplasms ,medicine ,Humans ,lcsh:Science ,Receptor ,Cell Engineering ,Receptors, Chimeric Antigen ,Multidisciplinary ,Chemistry ,lcsh:R ,HEK 293 cells ,T-cell receptor ,hemic and immune systems ,Recombinant Proteins ,Chimeric antigen receptor ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,lcsh:Q ,Tumor Escape - Abstract
Cell therapy using T cell receptors (TCRs) and chimeric antigen receptors (CARs) represents a new wave of immunotherapies garnering considerable attention and investment. Further progress in this area of medicine depends in part on improving the functional capabilities of the engineered components, while maintaining the overall size of recombinant constructs to ensure their compatibility with existing gene delivery vehicles. We describe a single-variable-domain TCR (svd TCR) that utilizes only the variable domain of the β chain (Vβ). This Vβ module not only works in TCR and CAR formats, but also can be used to create single-chain bispecific CARs and TCRs. Comparison of individual ligand-binding Vβ domains in different formats suggests that the lone Vβ sequence controls the sensitivity and a major part of the specificity of the CAR or TCR construct, regardless of signaling format, in Jurkat and primary T cells.
- Published
- 2019
13. Structure and dynamics of an imidazoline nitroxide side chain with strongly hindered internal motion in proteins
- Author
-
Wayne L. Hubbell, Duilio Cascio, Dora Toledo Warshaviak, Valery V. Khramtsov, and Christian Altenbach
- Subjects
Models, Molecular ,Nuclear and High Energy Physics ,Nitroxide mediated radical polymerization ,Protein Conformation ,Stereochemistry ,Biophysics ,Crystal structure ,Crystallography, X-Ray ,Biochemistry ,Protein Structure, Secondary ,Article ,law.invention ,Motion ,Protein structure ,law ,Side chain ,Imidazolines ,Electron paramagnetic resonance ,Conformational isomerism ,Quantitative Biology::Biomolecules ,Chemistry ,Electron Spin Resonance Spectroscopy ,Proteins ,Site-directed spin labeling ,Condensed Matter Physics ,Crystallography ,Muramidase ,Spin Labels ,Alpha helix - Abstract
A disulfide-linked imidazoline nitroxide side chain (V1) has a similar and highly constrained internal motion at diverse topological sites in a protein, unlike that for the disulfide-linked pyrroline nitroxide side chain (R1) widely used in site directed spin labeling EPR. Crystal structures of V1 at two positions in a helix of T4 Lysozyme and quantum mechanical calculations suggest the source of the constraints as intra-side chain interactions of the disulfide sulfur atoms with both the protein backbone and the 3-nitrogen in the imidazoline ring. These interactions apparently limit the conformation of the side chain to one of only three possible rotamers, two of which are observed in the crystal structure. An inter-spin distance measurement in frozen solution using double electron–electron resonance (DEER) gives a value essentially identical to that determined from the crystal structure of the protein containing two copies of V1, indicating that lattice forces do not dictate the rotamers observed. Collectively, the results suggest the possibility of predetermining a unique rotamer of V1 in helical structures. In general, the reduced rotameric space of V1 compared to R1 should simplify interpretation of inter-spin distance information in terms of protein structure, while the highly constrained internal motion is expected to extend the dynamic range for characterizing large amplitude nanosecond backbone fluctuations.
- Published
- 2013
14. Effect of membrane tension on the physical properties of DOPC lipid bilayer membrane
- Author
-
A. Srinivas Reddy, Dora Toledo Warshaviak, and Mirianas Chachisvilis
- Subjects
Models, Molecular ,Anomalous diffusion ,Diffusion ,Lipid Bilayers ,Molecular Conformation ,Analytical chemistry ,Biophysics ,02 engineering and technology ,Molecular Dynamics Simulation ,Lipid diffusion ,Biochemistry ,Article ,03 medical and health sciences ,GPCR ,Mechanosensing ,Pressure ,Membrane fluidity ,Lipid bilayer phase behavior ,Lipid bilayer ,030304 developmental biology ,0303 health sciences ,Membranes ,Models, Statistical ,Shear stress ,Chemistry ,Bilayer ,Lipid bilayer mechanics ,Cell Biology ,Membrane tension ,021001 nanoscience & nanotechnology ,Lipids ,Membrane ,Models, Chemical ,Phosphatidylcholines ,lipids (amino acids, peptides, and proteins) ,0210 nano-technology ,Software - Abstract
Molecular dynamics simulations of a dioleoylphosphocholine (DOPC) lipid bilayer were performed to explore its mechanosensitivity. Variations in the bilayer properties, such as area per lipid, volume, thickness, hydration depth (HD), hydration thickness (HT), lateral diffusion coefficient, and changes in lipid structural order were computed in the membrane tension range 0 to 15 dyn/cm. We determined that an increase in membrane tension results in a decrease in the bilayer thickness and HD of ~ 5% and ~ 5.7% respectively, whereas area per lipid, volume, and HT/HD increased by 6.8%, 2.4%, and 5% respectively. The changes in lipid conformation and orientation were characterized using orientational (S2) and deuterium (SCD) order parameters. Upon increase of membrane tension both order parameters indicated an increase in lipid disorder by 10–20%, mostly in the tail end region of the hydrophobic chains. The effect of membrane tension on lipid lateral diffusion in the DOPC bilayer was analyzed on three different time scales corresponding to inertial motion, anomalous diffusion and normal diffusion. The results showed that lateral diffusion of lipid molecules is anomalous in nature due to the non-exponential distribution of waiting times. The anomalous and normal diffusion coefficients increased by 20% and 52% when the membrane tension changed from 0 to 15 dyn/cm, respectively. In conclusion, our studies showed that membrane tension causes relatively significant changes in the area per lipid, volume, polarity, membrane thickness, and fluidity of the membrane suggesting multiple mechanisms by which mechanical perturbation of the membrane could trigger mechanosensitive response in cells.
- Published
- 2012
- Full Text
- View/download PDF
15. Conformational Analysis of a Nitroxide Side Chain in an α-Helix with Density Functional Theory
- Author
-
Kendall N. Houk, Wayne L. Hubbell, Laura Serbulea, and Dora Toledo Warshaviak
- Subjects
Models, Molecular ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Protein Structure, Secondary ,Article ,law.invention ,Molecular dynamics ,Protein structure ,law ,Materials Chemistry ,Side chain ,Disulfides ,Physical and Theoretical Chemistry ,Electron paramagnetic resonance ,Conformational isomerism ,Chemistry ,Electron Spin Resonance Spectroscopy ,Proteins ,Stereoisomerism ,Site-directed spin labeling ,Protein Structure, Tertiary ,Surfaces, Coatings and Films ,Solutions ,Crystallography ,Helix ,Mutagenesis, Site-Directed ,Solvents ,Thermodynamics ,Spin Labels ,Hydrophobic and Hydrophilic Interactions ,Alpha helix - Abstract
In site directed spin labeling, a nitroxide side chain is introduced at a selected site in a protein; the most commonly used is a disulfide-linked side chain designated R1. The electron paramagnetic resonance (EPR) spectra of R1, and the interspin distance between pairs of R1 residues as determined by dipolar EPR spectroscopy, encode a wealth of information on the protein structure and dynamics. However, extracting this information requires structural and dynamical models of the R1 side chain, that is, the favored rotamers, the intraresidue interactions that stabilize them, and the internal modes of motion. X-ray crystal structures of R1 in proteins have revealed a set of preferred rotamers in the crystal lattice. To identify the intraresidue interactions that stabilize particular rotamers of R1 in the absence of interactions with nearby side chains in a helix, and to evaluate models for the internal motion of the side chain, quantum mechanical calculations were performed on a relevant fragment of R1 in a 10-residue α-helix. Relative rotamer energies were determined in the gas phase, and solvation energies were estimated from a continuum solvent model that includes both electrostatic and hydrophobic contributions. The results identified preferred rotamers that are in agreement with the X-ray crystallographic studies. The rotamers are apparently stabilized by intraresidue sulfur-backbone interactions, suggesting that the preferred rotamers may be the same at all solvent-exposed helix sites.
- Published
- 2010
16. Hydrophobic Loop Dynamics and Actin Filament Stability
- Author
-
Alexander Shvetsov, John D. Stamm, Peter A. Rubenstein, Dora Toledo-Warshaviak, Martin L. Phillips, Wayne L. Hubbell, Emil Reisler, Christian Altenbach, and Damon Scoville
- Subjects
Light ,Phalloidine ,Protein Conformation ,Phalloidin ,Arp2/3 complex ,Saccharomyces cerevisiae ,macromolecular substances ,Biochemistry ,Protein filament ,chemistry.chemical_compound ,Protein structure ,Scattering, Radiation ,Actin-binding protein ,biology ,Chemistry ,Electron Spin Resonance Spectroscopy ,Actin remodeling ,Site-directed spin labeling ,Actin Cytoskeleton ,Microscopy, Electron ,Crystallography ,Treadmilling ,Amino Acid Substitution ,biology.protein ,Spin Labels ,Hydrophobic and Hydrophilic Interactions - Abstract
It has been postulated that the hydrophobic loop of actin (residues 262-274) swings out and inserts into the opposite strand in the filament, stabilizing the filament structure. Here, we analyzed the hydrophobic loop dynamics utilizing four mutants that have cysteine residues introduced at a single location along the yeast actin loop. Lateral, copper-catalyzed disulfide cross-linking of the mutant cysteine residues to the native C374 in the neighboring strand within the filament was fastest for S265C, followed by V266C, L267C, and then L269C. Site-directed spin labeling (SDSL) studies revealed that C265 lies closest to C374 within the filament, followed by C266, C267, and then C269. These results are not predicted by the Holmes extended loop model of F-actin. Furthermore, we find that disulfide cross-linking destroys L267C and L269C filaments; only small filaments are observed via electron microscopy. Conversely, phalloidin protects the L267C and L269C filaments and inhibits their disulfide cross-linking. Combined, our data indicate that, in solution, the loop resides predominantly in a "parked" position within the filament but is able to dynamically populate other conformational states which stabilize or destabilize the filament. Such states may be exploited within a cell by filament-stabilizing and -destabilizing factors.
- Published
- 2006
17. Conformational Dynamics of Loop 262−274 in G- and F-actin
- Author
-
John D. Stamm, Christian Altenbach, Peter A. Rubenstein, Martin L. Phillips, Wayne L. Hubbell, Dora Toledo Warshaviak, Alexander Shvetsov, Kálmán Hideg, and Emil Reisler
- Subjects
Nitroxide mediated radical polymerization ,Phalloidine ,Protein Conformation ,Saccharomyces cerevisiae ,Tropomyosin ,macromolecular substances ,Biochemistry ,law.invention ,Protein filament ,Bridged Bicyclo Compounds ,Protein structure ,law ,Myosin ,Cysteine ,Disulfides ,Electron paramagnetic resonance ,Actin ,Mesylates ,Chemistry ,Electron Spin Resonance Spectroscopy ,Myosin Subfragments ,Actins ,Actin Cytoskeleton ,Crystallography ,Cross-Linking Reagents ,Polymerization ,Hydrophobic and Hydrophilic Interactions - Abstract
According to the original Holmes model of F-actin structure, the hydrophobic loop 262-274 stabilizes the actin filament by inserting into a pocket formed at the interface between two protomers on the opposing strand. Using a yeast actin triple mutant, L180C/L269C/C374A [(LC)(2)CA], we showed previously that locking the hydrophobic loop to the G-actin surface by a disulfide bridge prevents filament formation. We report here that the hydrophobic loop is mobile in F- as well as in G-actin, fluctuating between the extended and parked conformations. Copper-catalyzed, brief air oxidation of (LC)(2)CA F-actin on electron microscopy grids resulted in the severing of thin filaments and their conversion to amorphous aggregates. Disulfide, bis(methanethiosulfonate) (MTS), and dibromobimane (DBB) cross-linking reactions proceeded in solution at a faster rate with G- than with F-actin. Cross-linking of C180 to C269 by DBB (4.4 A) in either G- or F-actin resulted in shorter and less stable filaments. The cross-linking with a longer MTS-6 reagent (9.6 A) did not impair actin polymerization or filament structure. Myosin subfragment 1 (S1) and tropomyosin inhibited the disulfide cross-linking of phalloidin-stabilized F-actin. Electron paramagnetic resonance measurements with nitroxide spin-labeled actin revealed strong spin-spin coupling and a similar mean interspin distance ( approximately 10 A) in G- and in F-actin, with a broader distance distribution in G-actin. These results show loop 262-274 fluctuations in G- and F-actin and correlate loop dynamics with actin filament formation and stability.
- Published
- 2006
18. Effect of Membrane Tension on the Electric Field and Dipole Potential of Lipid Bilayer Membrane
- Author
-
Mirianas Chachisvilis, Dora Toledo Warshaviak, and Michael J. Muellner
- Subjects
Lipid Bilayers ,Static Electricity ,Analytical chemistry ,Biophysics ,02 engineering and technology ,Model lipid bilayer ,Molecular Dynamics Simulation ,Biochemistry ,Article ,Lipid bilayer ,03 medical and health sciences ,GPCR ,Mechanosensing ,Membrane fluidity ,Lipid bilayer phase behavior ,030304 developmental biology ,0303 health sciences ,Chemistry ,Vesicle ,Bilayer ,Fluid shear stress ,Membranes, Artificial ,Lipid bilayer mechanics ,Cell Biology ,Membrane tension ,021001 nanoscience & nanotechnology ,Dipole potential ,Molecular Probes ,Phosphatidylcholines ,lipids (amino acids, peptides, and proteins) ,0210 nano-technology ,Elasticity of cell membranes - Abstract
The dipole potential of lipid bilayer membrane controls the difference in permeability of the membrane to oppositely charged ions. We have combined molecular dynamics (MD) simulations and experimental studies to determine changes in electric field and electrostatic potential of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) lipid bilayer in response to applied membrane tension. MD simulations based on CHARMM36 force field showed that electrostatic potential of DOPC bilayer decreases by ~ 45 mV in the physiologically relevant range of membrane tension values (0 to 15 dyn/cm). The electrostatic field exhibits a peak (~ 0.8 × 109 V/m) near the water/lipid interface which shifts by 0.9 A towards the bilayer center at 15 dyn/cm. Maximum membrane tension of 15 dyn/cm caused 6.4% increase in area per lipid, 4.7% decrease in bilayer thickness and 1.4% increase in the volume of the bilayer. Dipole-potential sensitive fluorescent probes were used to detect membrane tension induced changes in DOPC vesicles exposed to osmotic stress. Experiments confirmed that dipole potential of DOPC bilayer decreases at higher membrane tensions. These results are suggestive of a potentially new mechanosensing mechanism by which mechanically induced structural changes in the lipid bilayer membrane could modulate the function of membrane proteins by altering electrostatic interactions and energetics of protein conformational states.
- Published
- 2011
19. Mapping the cofilin binding site on yeast G-actin by chemical cross-linking
- Author
-
Rachel R. Ogorzalek Loo, David Sept, Pinmanee Boontheung, Joseph A. Loo, Emil Reisler, Kym F. Faull, Julian P. Whitelegge, Elena E. Grintsevich, Dora Toledo Warshaviak, Frédéric Halgand, Sabrina A. Benchaar, Institut de Chimie des Substances Naturelles (ICSN), and Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC)
- Subjects
Models, Molecular ,MESH: Protein Structure, Quaternary ,Plasma protein binding ,MESH: Amino Acid Sequence ,Microfilament ,environment and public health ,MESH: Protein Structure, Tertiary ,0302 clinical medicine ,Protein structure ,Structural Biology ,Cytoskeleton ,MESH: Structural Homology, Protein ,0303 health sciences ,biology ,Cofilin ,MESH: Saccharomyces cerevisiae ,Cross-Linking Reagents ,Biochemistry ,Actin Depolymerizing Factors ,Ethylmaleimide ,MESH: Ethylmaleimide ,MESH: Models, Molecular ,Protein Binding ,MESH: Mutation ,MESH: Cross-Linking Reagents ,Molecular Sequence Data ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,macromolecular substances ,Saccharomyces cerevisiae ,MESH: Actins ,Article ,03 medical and health sciences ,MESH: Actin Depolymerizing Factors ,MESH: Protein Binding ,Actin-binding protein ,Amino Acid Sequence ,Binding site ,Protein Structure, Quaternary ,Molecular Biology ,Actin ,030304 developmental biology ,MESH: Molecular Sequence Data ,Binding Sites ,Actins ,Protein Structure, Tertiary ,MESH: Binding Sites ,Structural Homology, Protein ,Mutation ,biology.protein ,Biophysics ,030217 neurology & neurosurgery - Abstract
International audience; Cofilin is a major cytoskeletal protein that binds to both monomeric actin (G-actin) and polymeric actin (F-actin) and is involved in microfilament dynamics. Although an atomic structure of the G-actin-cofilin complex does not exist, models of the complex have been built using molecular dynamics simulations, structural homology considerations, and synchrotron radiolytic footprinting data. The hydrophobic cleft between actin subdomains 1 and 3 and, alternatively, the cleft between actin subdomains 1 and 2 have been proposed as possible high-affinity cofilin binding sites. In this study, the proposed binding of cofilin to the subdomain 1/subdomain 3 region on G-actin has been probed using site-directed mutagenesis, fluorescence labeling, and chemical cross-linking, with yeast actin mutants containing single reactive cysteines in the actin hydrophobic cleft and with cofilin mutants carrying reactive cysteines in the regions predicted to bind to G-actin. Mass spectrometry analysis of the cross-linked complex revealed that cysteine 345 in subdomain 1 of mutant G-actin was cross-linked to native cysteine 62 on cofilin. A cofilin mutant that carried a cysteine substitution in the alpha 3-helix (residue 95) formed a cross-link with residue 144 in actin subdomain 3. Distance constraints imposed by these cross-links provide experimental evidence for cofilin binding between actin subdomains 1 and 3 and fit a corresponding docking-based structure of the complex. The cross-linking of the N-terminal region of recombinant yeast cofilin to actin residues 346 and 374 with dithio-bis-maleimidoethane (12.4 A) and via disulfide bond formation was also documented. This set of cross-linking data confirms the important role of the N-terminal segment of cofilin in interactions with G-actin.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.