1
|
Encinas-Basurto D, Muralidharan P, Saiful Islam MD, Vallorz EL, Black SM, Kraft M, Ledford JG, Mansour HM. Comprehensive physicochemical, biophysical, and in vitro characterization of lung surfactant SP-A peptidomimetics. RSC PHARMACEUTICS 2025:d4pm00265b. [PMID: 40342402 PMCID: PMC12053052 DOI: 10.1039/d4pm00265b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Surfactant protein-A (SP-A) is an endogenous and essential lung surfactant-specific protein that is integral to pulmonary immunity, including inhibition of asthma exacerbations. This study aims to comprehensively characterize two peptides (10-AA and 20-AA) of SP-A which confer activity similar to the full-length oligomeric SP-A protein. Spectroscopic and chromatographic analyses revealed that the phosphate (PS) and acetate (AC) salts exhibited distinct solubility and log P partitioning behavior, impacting their physicochemical properties. MD simulations and circular dichroism showed that SP-A 10-AA initially adopts an α-helical structure but loses helicity over time, while SP-A 20-AA remains disordered. Differential scanning calorimetry confirmed variations in thermal stability between salt forms and zeta potential measurements showed that PS salts had a more negative surface charge, potentially influencing membrane interactions. In vitro studies showed high cell viability (>90%) and stable TEER values at the air-liquid interface, confirming biocompatibility and potential epithelial permeability. These findings provide crucial insights into the structural and functional properties of SP-A peptides, supporting their potential as therapeutic agents for pulmonary diseases.
Collapse
Affiliation(s)
- David Encinas-Basurto
- The University of Arizona College of Pharmacy Tucson AZ USA
- University of Sonora Mexico
| | | | | | | | - Stephen M Black
- The University of Arizona College of Medicine Tucson AZ USA
- Florida International University, Center for Translational Science Port St. Lucie FL USA
| | - Monica Kraft
- The University of Arizona College of Medicine Tucson AZ USA
- The University of Arizona Health Sciences Asthma and Airway Disease Research Center Tucson AZ USA
- The University of Arizona, the BIO5 Research Institute Tucson AZ USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System New York NY USA
| | - Julie G Ledford
- The University of Arizona College of Medicine Tucson AZ USA
- The University of Arizona Health Sciences Asthma and Airway Disease Research Center Tucson AZ USA
- The University of Arizona, the BIO5 Research Institute Tucson AZ USA
| | - Heidi M Mansour
- The University of Arizona College of Pharmacy Tucson AZ USA
- The University of Arizona College of Medicine Tucson AZ USA
- Florida International University, Center for Translational Science Port St. Lucie FL USA
- The University of Arizona, the BIO5 Research Institute Tucson AZ USA
| |
Collapse
|
2
|
Grewal S, Iyamu U, Vinals D, Mitran C, Hegde N, Yanow S. A machine learning framework to identify complex physicochemical features of B cell epitopes. RESEARCH SQUARE 2025:rs.3.rs-6255613. [PMID: 40321766 PMCID: PMC12047986 DOI: 10.21203/rs.3.rs-6255613/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
During infection with Plasmodium falciparum in pregnancy, parasites express a unique virulence factor, VAR2CSA, that mediates binding of infected red blood cells to the placenta. A major goal in designing vaccines to protect pregnant women from malaria is to elicit antibodies to VAR2CSA. The challenge is that VAR2CSA is highly polymorphic and identifying conserved epitopes is essential to elicit strain-transcending immunity. Unexpectedly, a mouse monoclonal antibody, 3D10, raised against the unrelated Duffy binding protein from P. vivax (DBPII) cross-reacts with diverse alleles of VAR2CSA in vitro. To identify these potentially conserved epitopes in VAR2CSA, we designed a machine learning framework to analyse 3D10 reactivity to peptides derived from two alleles of VAR2CSA, DBPII, and PvEBP2 (negative control). We used decision trees and a panel of 430 features to extract features correlated to 3D10 binding. We analysed patterns of these features in the dataset and designed mutant peptides to test complex sequence motifs. Features associated with 3D10 reactivity were mapped onto predicted 3D structures of Plasmodium proteins and validated based on 3D10 reactivity to the recombinant antigens. While the array data identified certain linear epitopes, the framework predicted other epitopes that are conformational. With this approach, peptide array data can be mined to extract physicochemical properties of epitopes recognized by polyreactive antibodies.
Collapse
|
3
|
McDonald EF, Kim M, Olson JA, Meiler J, Plate L. Proteostasis Landscapes of Cystic Fibrosis Variants Reveals Drug Response Vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.10.602964. [PMID: 39026768 PMCID: PMC11257600 DOI: 10.1101/2024.07.10.602964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cystic Fibrosis (CF) is a lethal genetic disorder caused by variants in CF transmembrane conductance regulator (CFTR). Many variants are treatable with correctors, which enhance the folding and trafficking of CFTR. However, approximately 3% of persons with CF harbor poorly responsive variants. Here, we used affinity purification mass spectrometry proteomics to profile the protein homeostasis (proteostasis) changes of CFTR variants during correction to assess modulated interactions with protein folding and maturation pathways. Responsive variant interactions converged on similar proteostasis pathways during correction. In contrast, poorly responsive variants subtly diverged, revealing a partial restoration of protein quality control surveillance and partial correction. Computational structural modeling showed that corrector VX-445 failed to confer enough NBD1 stability to poor responders. NBD1 secondary stabilizing mutations rescued poorly responsive variants, revealing structural vulnerabilities in NBD1 required for treating poor responders. Our study provides a framework for discerning the underlying protein quality control and structural defects of CFTR variants not reached with existing drugs to expand therapeutics to all susceptible CFTR variants. SIGNIFICANCE STATEMENT Cystic Fibrosis (CF) is a lethal genetic disease with variants leading to misfolding of an anion channel protein. Enhancing productive channel folding using a novel class of small molecules called correctors has emerged as the current CF treatment paradigm. However, correctors fail to reach all patient variants. Using high throughput interactomics, Rosetta simulations, and biochemical trafficking assays, this study demonstrates poorly responsive CF variants experience diverse misfolding pathways caused by structural defects in the core of a nucleotide-binding domain. Stabilizing secondary mutations in this domain rescues poorly responsive variants, paving the way for mechanistic-based therapeutic development for untreatable CF variants and future protein misfolding corrector drugs.
Collapse
Affiliation(s)
- Eli Fritz McDonald
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Minsoo Kim
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37240, USA
| | - John A. Olson
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240, USA
- Institute for Drug Discovery, Leipzig University, Leipzig, SAC 04103, Germany
| | - Lars Plate
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37240, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Penna BR, Gomes-Neto F, Anobom CD, Valente AP. Structural and dynamics characterization of the Zika virus NS2B using nuclear magnetic resonance and RosettaMP: A challenge for transmembrane protein studies. Int J Biol Macromol 2024; 280:136074. [PMID: 39341314 DOI: 10.1016/j.ijbiomac.2024.136074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Zika virus (ZIKV) is an emergent flavivirus that represents a global public health concern due to its association with severe neurological disorders. NS2B is a multifunctional viral membrane protein primarily used to regulate viral protease activity and is crucial for virus replication, making it an appealing target for antiviral drugs. This study presents the structural elucidation of full-length ZIKV NS2B in sodium dodecyl sulfate (SDS) micelles using solution nuclear magnetic resonance experimental data and RosettaMP. The protein structure has four transmembrane α-helices, two amphipathic α-helices, and a β-hairpin in the hydrophilic region. NS2B presented secondary and tertiary stability in different concentrations of SDS. Furthermore, we studied the dynamics of NS2B in SDS micelles through relaxation parameters and paramagnetic relaxation enhancement experiments. The findings were consistent with the structural calculations. Our work will be essential in understanding the role of NS2B in viral replication and screening for inhibitors against ZIKV.
Collapse
Affiliation(s)
- Beatriz R Penna
- Institute of Medical Biochemistry (IBqM), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Center of Nuclear Magnetic Resonance (CNRMN), Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Cristiane D Anobom
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- Institute of Medical Biochemistry (IBqM), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Center of Nuclear Magnetic Resonance (CNRMN), Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
5
|
Todorovic J, Swapna I, Suma A, Carnevale V, Zakon H. Dual mechanisms contribute to enhanced voltage dependence of an electric fish potassium channel. Biophys J 2024; 123:2097-2109. [PMID: 38429925 PMCID: PMC11309972 DOI: 10.1016/j.bpj.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
The voltage dependence of different voltage-gated potassium channels, described by the voltage at which half of the channels are open (V1/2), varies over a range of 80 mV and is influenced by factors such as the number of positive gating charges and the identity of the hydrophobic amino acids in the channel's voltage sensor (S4). Here we explore by experimental manipulations and molecular dynamics simulation the contributions of two derived features of an electric fish potassium channel (Kv1.7a) that is among the most voltage-sensitive Shaker family potassium channels known. These are a patch of four contiguous negatively charged glutamates in the S3-S4 extracellular loop and a glutamate in the S3b helix. We find that these negative charges affect V1/2 by separate, complementary mechanisms. In the closed state, the S3-S4 linker negative patch reduces the membrane surface charge biasing the channel to enter the open state while, upon opening, the negative amino acid in the S3b helix faces the second (R2) gating charge of the voltage sensor electrostatically biasing the channel to remain in the open state. This work highlights two evolutionary novelties that illustrate the potential influence of negatively charged amino acids in extracellular loops and adjacent helices to voltage dependence.
Collapse
Affiliation(s)
- Jelena Todorovic
- Department of Neuroscience, The University of Texas, Austin, Texas
| | - Immani Swapna
- Department of Neuroscience, The University of Texas, Austin, Texas
| | - Antonio Suma
- Institute for Computational Molecular Science, College of Science and Technology & Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology & Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania
| | - Harold Zakon
- Department of Neuroscience, The University of Texas, Austin, Texas; Department of Integrative Biology, The University of Texas, Austin, Texas.
| |
Collapse
|
6
|
Hermosilla AM, Berner C, Ovchinnikov S, Vorobieva AA. Validation of de novo designed water-soluble and transmembrane β-barrels by in silico folding and melting. Protein Sci 2024; 33:e5033. [PMID: 38864690 PMCID: PMC11168064 DOI: 10.1002/pro.5033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/14/2024] [Accepted: 05/08/2024] [Indexed: 06/13/2024]
Abstract
In silico validation of de novo designed proteins with deep learning (DL)-based structure prediction algorithms has become mainstream. However, formal evidence of the relationship between a high-quality predicted model and the chance of experimental success is lacking. We used experimentally characterized de novo water-soluble and transmembrane β-barrel designs to show that AlphaFold2 and ESMFold excel at different tasks. ESMFold can efficiently identify designs generated based on high-quality (designable) backbones. However, only AlphaFold2 can predict which sequences have the best chance of experimentally folding among similar designs. We show that ESMFold can generate high-quality structures from just a few predicted contacts and introduce a new approach based on incremental perturbation of the prediction ("in silico melting"), which can reveal differences in the presence of favorable contacts between designs. This study provides a new insight on DL-based structure prediction models explainability and on how they could be leveraged for the design of increasingly complex proteins; in particular membrane proteins which have historically lacked basic in silico validation tools.
Collapse
Affiliation(s)
- Alvaro Martin Hermosilla
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
| | - Carolin Berner
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
| | - Sergey Ovchinnikov
- John Harvard Distinguished Science Fellowship ProgramHarvard UniversityCambridgeMassachusettsUSA
- Present address:
Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Anastassia A. Vorobieva
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
- VIB Center for AI and Computational BiologyBelgium
| |
Collapse
|
7
|
Weinstein JJ, Saikia C, Karbat I, Goldenzweig A, Reuveny E, Fleishman SJ. One-shot design elevates functional expression levels of a voltage-gated potassium channel. Protein Sci 2024; 33:e4995. [PMID: 38747377 PMCID: PMC11094769 DOI: 10.1002/pro.4995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 05/19/2024]
Abstract
Membrane proteins play critical physiological roles as receptors, channels, pumps, and transporters. Despite their importance, however, low expression levels often hamper the experimental characterization of membrane proteins. We present an automated and web-accessible design algorithm called mPROSS (https://mPROSS.weizmann.ac.il), which uses phylogenetic analysis and an atomistic potential, including an empirical lipophilicity scale, to improve native-state energy. As a stringent test, we apply mPROSS to the Kv1.2-Kv2.1 paddle chimera voltage-gated potassium channel. Four designs, encoding 9-26 mutations relative to the parental channel, were functional and maintained potassium-selective permeation and voltage dependence in Xenopus oocytes with up to 14-fold increase in whole-cell current densities. Additionally, single-channel recordings reveal no significant change in the channel-opening probability nor in unitary conductance, indicating that functional expression levels increase without impacting the activity profile of individual channels. Our results suggest that the expression levels of other dynamic channels and receptors may be enhanced through one-shot design calculations.
Collapse
Affiliation(s)
- Jonathan Jacob Weinstein
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
- Present address:
Scala Biodesign LtdTel AvivIsrael
| | - Chandamita Saikia
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
- Present address:
Institute for BiochemistryUniversity of LübeckLübeckGermany
| | - Izhar Karbat
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | | | - Eitan Reuveny
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | | |
Collapse
|
8
|
Zhu H, Szymczyk A, Ghoufi A. Multiscale modelling of transport in polymer-based reverse-osmosis/nanofiltration membranes: present and future. DISCOVER NANO 2024; 19:91. [PMID: 38771417 PMCID: PMC11109084 DOI: 10.1186/s11671-024-04020-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/22/2024] [Indexed: 05/22/2024]
Abstract
Nanofiltration (NF) and reverse osmosis (RO) processes are physical separation technologies used to remove contaminants from liquid streams by employing dense polymer-based membranes with nanometric voids that confine fluids at the nanoscale. At this level, physical properties such as solvent and solute permeabilities are intricately linked to molecular interactions. Initially, numerous studies focused on developing macroscopic transport models to gain insights into separation properties at the nanometer scale. However, continuum-based models have limitations in nanoconfined situations that can be overcome by force field molecular simulations. Continuum-based models heavily rely on bulk properties, often neglecting critical factors like liquid structuring, pore geometry, and molecular/chemical specifics. Molecular/mesoscale simulations, while encompassing these details, often face limitations in time and spatial scales. Therefore, achieving a comprehensive understanding of transport requires a synergistic integration of both approaches through a multiscale approach that effectively combines and merges both scales. This review aims to provide a comprehensive overview of the state-of-the-art in multiscale modeling of transport through NF/RO membranes, spanning from the nanoscale to continuum media.
Collapse
Affiliation(s)
- Haochen Zhu
- State Key Laboratory of Pollution Control and Resources Reuse, Key Laboratory of Yangtze River Water Environment, College of Environmental Science and Engineering, Tongji University, 1239 Siping Rd., Shanghai, 200092, China.
| | - Anthony Szymczyk
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, Univ Rennes, 35000, Rennes, France.
| | - Aziz Ghoufi
- CNRS, ICMPE (Institut de Chimie et des Matériaux Paris-Est) - UMR 7182, Univ Paris-East Creteil, 94320, Thiais, France.
- CNRS, IPR (Institut de Physique de Rennes) - UMR 6251, Univ Rennes, 35000, Rennes, France.
| |
Collapse
|
9
|
Woods H, Leman JK, Meiler J. Modeling membrane geometries implicitly in Rosetta. Protein Sci 2024; 33:e4908. [PMID: 38358133 PMCID: PMC10868433 DOI: 10.1002/pro.4908] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
Interactions between membrane proteins (MPs) and lipid bilayers are critical for many cellular functions. In the Rosetta molecular modeling suite, the implicit membrane energy function is based on a "slab" model, which represent the membrane as a flat bilayer. However, in nature membranes often have a curvature that is important for function and/or stability. Even more prevalent, in structural biology research MPs are reconstituted in model membrane systems such as micelles, bicelles, nanodiscs, or liposomes. Thus, we have modified the existing membrane energy potentials within the RosettaMP framework to allow users to model MPs in different membrane geometries. We show that these modifications can be utilized in core applications within Rosetta such as structure refinement, protein-protein docking, and protein design. For MP structures found in curved membranes, refining these structures in curved, implicit membranes produces higher quality models with structures closer to experimentally determined structures. For MP systems embedded in multiple membranes, representing both membranes results in more favorable scores compared to only representing one of the membranes. Modeling MPs in geometries mimicking the membrane model system used in structure determination can improve model quality and model discrimination.
Collapse
Affiliation(s)
- Hope Woods
- Center of Structural Biology, Vanderbilt UniversityNashvilleTennesseeUSA
- Chemical and Physical Biology ProgramVanderbilt UniversityNashvilleTennesseeUSA
| | | | - Jens Meiler
- Center of Structural Biology, Vanderbilt UniversityNashvilleTennesseeUSA
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Institute for Drug Discovery, Leipzig University Medical SchoolLeipzigGermany
| |
Collapse
|
10
|
Samanta R, Gray JJ. Implicit model to capture electrostatic features of membrane environment. PLoS Comput Biol 2024; 20:e1011296. [PMID: 38252688 PMCID: PMC10833867 DOI: 10.1371/journal.pcbi.1011296] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 02/01/2024] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Membrane protein structure prediction and design are challenging due to the complexity of capturing the interactions in the lipid layer, such as those arising from electrostatics. Accurately capturing electrostatic energies in the low-dielectric membrane often requires expensive Poisson-Boltzmann calculations that are not scalable for membrane protein structure prediction and design. In this work, we have developed a fast-to-compute implicit energy function that considers the realistic characteristics of different lipid bilayers, making design calculations tractable. This method captures the impact of the lipid head group using a mean-field-based approach and uses a depth-dependent dielectric constant to characterize the membrane environment. This energy function Franklin2023 (F23) is built upon Franklin2019 (F19), which is based on experimentally derived hydrophobicity scales in the membrane bilayer. We evaluated the performance of F23 on five different tests probing (1) protein orientation in the bilayer, (2) stability, and (3) sequence recovery. Relative to F19, F23 has improved the calculation of the tilt angle of membrane proteins for 90% of WALP peptides, 15% of TM-peptides, and 25% of the adsorbed peptides. The performances for stability and design tests were equivalent for F19 and F23. The speed and calibration of the implicit model will help F23 access biophysical phenomena at long time and length scales and accelerate the membrane protein design pipeline.
Collapse
Affiliation(s)
- Rituparna Samanta
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jeffrey J. Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
11
|
Samanta R, Gray JJ. Implicit model to capture electrostatic features of membrane environment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546486. [PMID: 37425950 PMCID: PMC10327106 DOI: 10.1101/2023.06.26.546486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Membrane protein structure prediction and design are challenging due to the complexity of capturing the interactions in the lipid layer, such as those arising from electrostatics. Accurately capturing electrostatic energies in the low-dielectric membrane often requires expensive Poisson-Boltzmann calculations that are not scalable for membrane protein structure prediction and design. In this work, we have developed a fast-to-compute implicit energy function that considers the realistic characteristics of different lipid bilayers, making design calculations tractable. This method captures the impact of the lipid head group using a mean-field-based approach and uses a depth-dependent dielectric constant to characterize the membrane environment. This energy function Franklin2023 (F23) is built upon Franklin2019 (F19), which is based on experimentally derived hydrophobicity scales in the membrane bilayer. We evaluated the performance of F23 on five different tests probing (1) protein orientation in the bilayer, (2) stability, and (3) sequence recovery. Relative to F19, F23 has improved the calculation of the tilt angle of membrane proteins for 90% of WALP peptides, 15% of TM-peptides, and 25% of the adsorbed peptides. The performances for stability and design tests were equivalent for F19 and F23. The speed and calibration of the implicit model will help F23 access biophysical phenomena at long time and length scales and accelerate the membrane protein design pipeline.
Collapse
Affiliation(s)
- Rituparna Samanta
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
12
|
McKee AG, McDonald EF, Penn WD, Kuntz CP, Noguera K, Chamness LM, Roushar FJ, Meiler J, Oliver KE, Plate L, Schlebach JP. General trends in the effects of VX-661 and VX-445 on the plasma membrane expression of clinical CFTR variants. Cell Chem Biol 2023; 30:632-642.e5. [PMID: 37253358 PMCID: PMC10330547 DOI: 10.1016/j.chembiol.2023.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/17/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023]
Abstract
Cystic fibrosis (CF) is caused by mutations that compromise the expression and/or function of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. Most people with CF harbor a common misfolded variant (ΔF508) that can be partially rescued by therapeutic "correctors" that restore its expression. Nevertheless, many other CF variants are insensitive to correctors. Using deep mutational scanning, we quantitatively compare the effects of two correctors on the plasma membrane expression of 129 CF variants. Though structural calculations suggest corrector binding provides similar stabilization to most variants, it's those with intermediate expression and mutations near corrector binding pockets that exhibit the greatest response. Deviations in sensitivity appear to depend on the degree of variant destabilization and the timing of misassembly. Combining correctors appears to rescue more variants by doubling the binding energy and stabilizing distinct cotranslational folding transitions. These results provide an overview of rare CF variant expression and establish new tools for precision pharmacology.
Collapse
Affiliation(s)
- Andrew G McKee
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Eli F McDonald
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Wesley D Penn
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Charles P Kuntz
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Karen Noguera
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Laura M Chamness
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Francis J Roushar
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; Institute for Drug Development, Leipzig University, Leipzig, SAC 04109, Germany
| | - Kathryn E Oliver
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lars Plate
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
13
|
Gulsevin A, Han B, Porta JC, Mchaourab HS, Meiler J, Kenworthy AK. Template-free prediction of a new monotopic membrane protein fold and assembly by AlphaFold2. Biophys J 2023; 122:2041-2052. [PMID: 36352786 PMCID: PMC10257013 DOI: 10.1016/j.bpj.2022.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
AlphaFold2 (AF2) has revolutionized the field of protein structural prediction. Here, we test its ability to predict the tertiary and quaternary structure of a previously undescribed scaffold with new folds and unusual architecture, the monotopic membrane protein caveolin-1 (CAV1). CAV1 assembles into a disc-shaped oligomer composed of 11 symmetrically arranged protomers, each assuming an identical new fold, and contains the largest parallel β-barrel known to exist in nature. Remarkably, AF2 predicts both the fold of the protomers and the interfaces between them. It also assembles between seven and 15 copies of CAV1 into disc-shaped complexes. However, the predicted multimers are energetically strained, especially the parallel β-barrel. These findings highlight the ability of AF2 to correctly predict new protein folds and oligomeric assemblies at a granular level while missing some elements of higher-order complexes, thus positing a new direction for the continued development of deep-learning protein structure prediction approaches.
Collapse
Affiliation(s)
- Alican Gulsevin
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Bing Han
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Jason C Porta
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Hassane S Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee; Institute for Drug Discovery, Leipzig University, Leipzig, Germany.
| | - Anne K Kenworthy
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
14
|
Jefferson RE, Oggier A, Füglistaler A, Camviel N, Hijazi M, Villarreal AR, Arber C, Barth P. Computational design of dynamic receptor-peptide signaling complexes applied to chemotaxis. Nat Commun 2023; 14:2875. [PMID: 37208363 DOI: 10.1038/s41467-023-38491-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/04/2023] [Indexed: 05/21/2023] Open
Abstract
Engineering protein biosensors that sensitively respond to specific biomolecules by triggering precise cellular responses is a major goal of diagnostics and synthetic cell biology. Previous biosensor designs have largely relied on binding structurally well-defined molecules. In contrast, approaches that couple the sensing of flexible compounds to intended cellular responses would greatly expand potential biosensor applications. Here, to address these challenges, we develop a computational strategy for designing signaling complexes between conformationally dynamic proteins and peptides. To demonstrate the power of the approach, we create ultrasensitive chemotactic receptor-peptide pairs capable of eliciting potent signaling responses and strong chemotaxis in primary human T cells. Unlike traditional approaches that engineer static binding complexes, our dynamic structure design strategy optimizes contacts with multiple binding and allosteric sites accessible through dynamic conformational ensembles to achieve strongly enhanced signaling efficacy and potency. Our study suggests that a conformationally adaptable binding interface coupled to a robust allosteric transmission region is a key evolutionary determinant of peptidergic GPCR signaling systems. The approach lays a foundation for designing peptide-sensing receptors and signaling peptide ligands for basic and therapeutic applications.
Collapse
Affiliation(s)
- Robert E Jefferson
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Aurélien Oggier
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Andreas Füglistaler
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Nicolas Camviel
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Department of Oncology UNIL-CHUV, University Hospital Lausanne (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Mahdi Hijazi
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Ana Rico Villarreal
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Department of Oncology UNIL-CHUV, University Hospital Lausanne (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland.
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland.
| |
Collapse
|
15
|
Koehler Leman J, Künze G. Recent Advances in NMR Protein Structure Prediction with ROSETTA. Int J Mol Sci 2023; 24:ijms24097835. [PMID: 37175539 PMCID: PMC10178863 DOI: 10.3390/ijms24097835] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Nuclear magnetic resonance (NMR) spectroscopy is a powerful method for studying the structure and dynamics of proteins in their native state. For high-resolution NMR structure determination, the collection of a rich restraint dataset is necessary. This can be difficult to achieve for proteins with high molecular weight or a complex architecture. Computational modeling techniques can complement sparse NMR datasets (<1 restraint per residue) with additional structural information to elucidate protein structures in these difficult cases. The Rosetta software for protein structure modeling and design is used by structural biologists for structure determination tasks in which limited experimental data is available. This review gives an overview of the computational protocols available in the Rosetta framework for modeling protein structures from NMR data. We explain the computational algorithms used for the integration of different NMR data types in Rosetta. We also highlight new developments, including modeling tools for data from paramagnetic NMR and hydrogen-deuterium exchange, as well as chemical shifts in CS-Rosetta. Furthermore, strategies are discussed to complement and improve structure predictions made by the current state-of-the-art AlphaFold2 program using NMR-guided Rosetta modeling.
Collapse
Affiliation(s)
- Julia Koehler Leman
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY 10010, USA
| | - Georg Künze
- Institute for Drug Discovery, Medical Faculty, University of Leipzig, Brüderstr. 34, D-04103 Leipzig, Germany
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Härtelstr. 16-18, D-04107 Leipzig, Germany
| |
Collapse
|
16
|
Haver HN, Wedemeyer M, Butcher E, Peterson FC, Volkman BF, Scaglione KM. Mechanistic Insight into the Suppression of Polyglutamine Aggregation by SRCP1. ACS Chem Biol 2023; 18:549-560. [PMID: 36791332 PMCID: PMC10023506 DOI: 10.1021/acschembio.2c00893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Protein aggregation is a hallmark of the polyglutamine diseases. One potential treatment for these diseases is suppression of polyglutamine aggregation. Previous work identified the cellular slime mold Dictyostelium discoideum as being naturally resistant to polyglutamine aggregation. Further work identified serine-rich chaperone protein 1 (SRCP1) as a protein that is both necessary in Dictyostelium and sufficient in human cells to suppress polyglutamine aggregation. Therefore, understanding how SRCP1 suppresses aggregation may be useful for developing therapeutics for the polyglutamine diseases. Here we utilized a de novo protein modeling approach to generate predictions of SRCP1's structure. Using our best-fit model, we generated mutants that were predicted to alter the stability of SRCP1 and tested these mutants' stability in cells. Using these data, we identified top models of SRCP1's structure that are consistent with the C-terminal region of SRCP1 forming a β-hairpin with a highly dynamic N-terminal region. We next generated a series of peptides that mimic the predicted β-hairpin and validated that they inhibit aggregation of a polyglutamine-expanded mutant huntingtin exon 1 fragment in vitro. To further assess mechanistic details of how SRCP1 inhibits polyglutamine aggregation, we utilized biochemical assays to determine that SRCP1 inhibits secondary nucleation in a manner dependent upon the regions flanking the polyglutamine tract. Finally, to determine if SRCP1 more could generally suppress protein aggregation, we confirmed that it was sufficient to inhibit aggregation of polyglutamine-expanded ataxin-3. Together these studies provide details into the structural and mechanistic basis of the inhibition of protein aggregation by SRCP1.
Collapse
Affiliation(s)
- Holly N. Haver
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27710 USA
| | - Michael Wedemeyer
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226 USA
| | - Erin Butcher
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27710 USA
| | - Francis C. Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226 USA
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226 USA
| | - K. Matthew Scaglione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27710 USA
- Department of Neurology, Duke University, Durham, NC, 27710 USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Durham, NC, 27710 USA
| |
Collapse
|
17
|
Paradis JS, Feng X, Murat B, Jefferson RE, Sokrat B, Szpakowska M, Hogue M, Bergkamp ND, Heydenreich FM, Smit MJ, Chevigné A, Bouvier M, Barth P. Computationally designed GPCR quaternary structures bias signaling pathway activation. Nat Commun 2022; 13:6826. [PMID: 36369272 PMCID: PMC9652377 DOI: 10.1038/s41467-022-34382-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Communication across membranes controls critical cellular processes and is achieved by receptors translating extracellular signals into selective cytoplasmic responses. While receptor tertiary structures can be readily characterized, receptor associations into quaternary structures are challenging to study and their implications in signal transduction remain poorly understood. Here, we report a computational approach for predicting receptor self-associations, and designing receptor oligomers with various quaternary structures and signaling properties. Using this approach, we designed chemokine receptor CXCR4 dimers with reprogrammed binding interactions, conformations, and abilities to activate distinct intracellular signaling proteins. In agreement with our predictions, the designed CXCR4s dimerized through distinct conformations and displayed different quaternary structural changes upon activation. Consistent with the active state models, all engineered CXCR4 oligomers activated the G protein Gi, but only specific dimer structures also recruited β-arrestins. Overall, we demonstrate that quaternary structures represent an important unforeseen mechanism of receptor biased signaling and reveal the existence of a bias switch at the dimer interface of several G protein-coupled receptors including CXCR4, mu-Opioid and type-2 Vasopressin receptors that selectively control the activation of G proteins vs β-arrestin-mediated pathways. The approach should prove useful for predicting and designing receptor associations to uncover and reprogram selective cellular signaling functions.
Collapse
Affiliation(s)
- Justine S Paradis
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Xiang Feng
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Brigitte Murat
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Robert E Jefferson
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
| | - Badr Sokrat
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mireille Hogue
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Nick D Bergkamp
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Franziska M Heydenreich
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Martine J Smit
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland.
| |
Collapse
|
18
|
Qing R, Hao S, Smorodina E, Jin D, Zalevsky A, Zhang S. Protein Design: From the Aspect of Water Solubility and Stability. Chem Rev 2022; 122:14085-14179. [PMID: 35921495 PMCID: PMC9523718 DOI: 10.1021/acs.chemrev.1c00757] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Indexed: 12/13/2022]
Abstract
Water solubility and structural stability are key merits for proteins defined by the primary sequence and 3D-conformation. Their manipulation represents important aspects of the protein design field that relies on the accurate placement of amino acids and molecular interactions, guided by underlying physiochemical principles. Emulated designer proteins with well-defined properties both fuel the knowledge-base for more precise computational design models and are used in various biomedical and nanotechnological applications. The continuous developments in protein science, increasing computing power, new algorithms, and characterization techniques provide sophisticated toolkits for solubility design beyond guess work. In this review, we summarize recent advances in the protein design field with respect to water solubility and structural stability. After introducing fundamental design rules, we discuss the transmembrane protein solubilization and de novo transmembrane protein design. Traditional strategies to enhance protein solubility and structural stability are introduced. The designs of stable protein complexes and high-order assemblies are covered. Computational methodologies behind these endeavors, including structure prediction programs, machine learning algorithms, and specialty software dedicated to the evaluation of protein solubility and aggregation, are discussed. The findings and opportunities for Cryo-EM are presented. This review provides an overview of significant progress and prospects in accurate protein design for solubility and stability.
Collapse
Affiliation(s)
- Rui Qing
- State
Key Laboratory of Microbial Metabolism, School of Life Sciences and
Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Shilei Hao
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Key
Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Eva Smorodina
- Department
of Immunology, University of Oslo and Oslo
University Hospital, Oslo 0424, Norway
| | - David Jin
- Avalon GloboCare
Corp., Freehold, New Jersey 07728, United States
| | - Arthur Zalevsky
- Laboratory
of Bioinformatics Approaches in Combinatorial Chemistry and Biology, Shemyakin−Ovchinnikov Institute of Bioorganic
Chemistry RAS, Moscow 117997, Russia
| | - Shuguang Zhang
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
19
|
Bignold R, Shammout B, Rowley JE, Repici M, Simms J, Johnson JR. Chemokine CXCL12 drives pericyte accumulation and airway remodeling in allergic airway disease. Respir Res 2022; 23:183. [PMID: 35831901 PMCID: PMC9277926 DOI: 10.1186/s12931-022-02108-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Abstract
Background Airway remodeling is a significant contributor to impaired lung function in chronic allergic airway disease. Currently, no therapy exists that is capable of targeting these structural changes and the consequent loss of function. In the context of chronic allergic inflammation, pericytes have been shown to uncouple from the pulmonary microvasculature, migrate to areas of inflammation, and significantly contribute to airway wall remodeling and lung dysfunction. This study aimed to elucidate the mechanism by which pulmonary pericytes accumulate in the airway wall in a model of chronic allergic airway inflammation. Methods Mice were subjected to a protocol of chronic airway inflammation driven by the common environmental aeroallergen house dust mite. Phenotypic changes to lung pericytes were assessed by flow cytometry and immunostaining, and the functional capacity of these cells was evaluated using in vitro migration assays. The molecular mechanisms driving these processes were targeted pharmacologically in vivo and in vitro. Results Pericytes demonstrated increased CXCR4 expression in response to chronic allergic inflammation and migrated more readily to its cognate chemokine, CXCL12. This increase in migratory capacity was accompanied by pericyte accumulation in the airway wall, increased smooth muscle thickness, and symptoms of respiratory distress. Pericyte uncoupling from pulmonary vessels and subsequent migration to the airway wall were abrogated following topical treatment with the CXCL12 neutraligand LIT-927. Conclusion These results provide new insight into the role of the CXCL12/CXCR4 signaling axis in promoting pulmonary pericyte accumulation and airway remodeling and validate a novel target to address tissue remodeling associated with chronic inflammation.
Collapse
Affiliation(s)
- Rebecca Bignold
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Bushra Shammout
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Jessica E Rowley
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Mariaelena Repici
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - John Simms
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Jill R Johnson
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
20
|
McDonald EF, Woods H, Smith ST, Kim M, Schoeder CT, Plate L, Meiler J. Structural Comparative Modeling of Multi-Domain F508del CFTR. Biomolecules 2022; 12:biom12030471. [PMID: 35327663 PMCID: PMC8946492 DOI: 10.3390/biom12030471] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/07/2022] Open
Abstract
Cystic fibrosis (CF) is a rare genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), an epithelial anion channel expressed in several vital organs. Absence of functional CFTR results in imbalanced osmotic equilibrium and subsequent mucus build up in the lungs-which increases the risk of infection and eventually causes death. CFTR is an ATP-binding cassette (ABC) transporter family protein composed of two transmembrane domains (TMDs), two nucleotide binding domains (NBDs), and an unstructured regulatory domain. The most prevalent patient mutation is the deletion of F508 (F508del), making F508del CFTR the primary target for current FDA approved CF therapies. However, no experimental multi-domain F508del CFTR structure has been determined and few studies have modeled F508del using multi-domain WT CFTR structures. Here, we used cryo-EM density data and Rosetta comparative modeling (RosettaCM) to compare a F508del model with published experimental data on CFTR NBD1 thermodynamics. We then apply this modeling method to generate multi-domain WT and F508del CFTR structural models. These models demonstrate the destabilizing effects of F508del on NBD1 and the NBD1/TMD interface in both the inactive and active conformation of CFTR. Furthermore, we modeled F508del/R1070W and F508del bound to the CFTR corrector VX-809. Our models reveal the stabilizing effects of VX-809 on multi-domain models of F508del CFTR and pave the way for rational design of additional drugs that target F508del CFTR for treatment of CF.
Collapse
Affiliation(s)
- Eli Fritz McDonald
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (E.F.M.); (C.T.S.); (L.P.)
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA; (H.W.); (S.T.S.)
| | - Hope Woods
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA; (H.W.); (S.T.S.)
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Shannon T. Smith
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA; (H.W.); (S.T.S.)
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Minsoo Kim
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Clara T. Schoeder
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (E.F.M.); (C.T.S.); (L.P.)
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA; (H.W.); (S.T.S.)
- Leipzig Medical School, Leipzig University, 04109 Leipzig, Germany
| | - Lars Plate
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (E.F.M.); (C.T.S.); (L.P.)
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (E.F.M.); (C.T.S.); (L.P.)
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA; (H.W.); (S.T.S.)
- Leipzig Medical School, Leipzig University, 04109 Leipzig, Germany
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
- Institute for Drug Discovery, Leipzig University, 04109 Leipzig, Germany
- Correspondence: ; Tel.: +1-(615)-936-2211
| |
Collapse
|
21
|
Chen Z, Lin S, Xie T, Lin JM, Canessa CM. A flexible GAS belt responds to pore mutations changing the ion selectivity of proton-gated channels. J Gen Physiol 2022; 154:212811. [PMID: 34766968 PMCID: PMC8594623 DOI: 10.1085/jgp.202112978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/18/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022] Open
Abstract
Proton-gated ion channels conduct mainly Na+ to induce postsynaptic membrane depolarization. Finding the determinants of ion selectivity requires knowledge of the pore structure in the open conformation, but such information is not yet available. Here, the open conformation of the hASIC1a channel was computationally modeled, and functional effects of pore mutations were analyzed in light of the predicted structures. The open pore structure shows two constrictions of similar diameter formed by the backbone of the GAS belt and, right beneath it, by the side chains of H28 from the reentrant loop. Models of nonselective mutant channels, but not those that maintain ion selectivity, predict enlargement of the GAS belt, suggesting that this motif is quite flexible and that the loss of stabilizing interactions in the central pore leads to changes in size/shape of the belt. Our results are consistent with the "close-fit" mechanism governing selectivity of hASIC1a, wherein the backbone of the GAS substitutes at least part of the hydration shell of a permeant ion to enable crossing the pore constriction.
Collapse
Affiliation(s)
- Zhuyuan Chen
- Department of Basic Sciences, Tsinghua University School of Medicine, Beijing, China
| | - Sheng Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Tianze Xie
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Cecilia M Canessa
- Department of Basic Sciences, Tsinghua University School of Medicine, Beijing, China.,Cellular and Molecular Physiology, Yale University, New Haven, CT
| |
Collapse
|
22
|
Mechanism of negative membrane curvature generation by I-BAR domains. Structure 2021; 29:1440-1452.e4. [PMID: 34520736 DOI: 10.1016/j.str.2021.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/16/2021] [Accepted: 07/22/2021] [Indexed: 11/23/2022]
Abstract
The membrane sculpting ability of BAR domains has been attributed to the intrinsic curvature of their banana-shaped dimeric structure. However, there is often a mismatch between this intrinsic curvature and the diameter of the membrane tubules generated. I-BAR domains are especially mysterious since they are almost flat but generate high negative membrane curvature. Here, we use atomistic implicit-solvent computer modeling to show that the membrane bending of the IRSp53 I-BAR domain is dictated by its higher oligomeric structure, whose curvature is completely unrelated to the intrinsic curvature of the dimer. Two other I-BARs give similar results, whereas a flat F-BAR sheet develops a concave membrane-binding interface, consistent with its observed positive membrane curvature generation. Laterally interacting helical spirals of I-BAR dimers on tube interiors are stable and have an enhanced binding energy that is sufficient for membrane bending to experimentally observed tubule diameters at a reasonable surface density.
Collapse
|
23
|
Endocannabinoid activation of the TRPV1 ion channel is distinct from activation by capsaicin. J Biol Chem 2021; 297:101022. [PMID: 34332978 PMCID: PMC8387766 DOI: 10.1016/j.jbc.2021.101022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 11/21/2022] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) ion channel serves as the detector for noxious temperature above 42 °C, pungent chemicals like capsaicin, and acidic extracellular pH. This channel has also been shown to function as an ionotropic cannabinoid receptor. Despite the solving of high-resolution three-dimensional structures of TRPV1, how endocannabinoids such as anandamide and N-arachidonoyl dopamine bind to and activate this channel remains largely unknown. Here we employed a combination of patch-clamp recording, site-directed mutagenesis, and molecular docking techniques to investigate how the endocannabinoids structurally bind to and open the TRPV1 ion channel. We found that these endocannabinoid ligands bind to the vanilloid-binding pocket of TRPV1 in the “tail-up, head-down” configuration, similar to capsaicin; however, there is a unique interaction with TRPV1 Y512 residue critical for endocannabinoid activation of TRPV1 channels. These data suggest that a differential structural mechanism is involved in TRPV1 activation by endocannabinoids compared with the classic agonist capsaicin.
Collapse
|
24
|
Principles and Methods in Computational Membrane Protein Design. J Mol Biol 2021; 433:167154. [PMID: 34271008 DOI: 10.1016/j.jmb.2021.167154] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/13/2023]
Abstract
After decades of progress in computational protein design, the design of proteins folding and functioning in lipid membranes appears today as the next frontier. Some notable successes in the de novo design of simplified model membrane protein systems have helped articulate fundamental principles of protein folding, architecture and interaction in the hydrophobic lipid environment. These principles are reviewed here, together with the computational methods and approaches that were used to identify them. We provide an overview of the methodological innovations in the generation of new protein structures and functions and in the development of membrane-specific energy functions. We highlight the opportunities offered by new machine learning approaches applied to protein design, and by new experimental characterization techniques applied to membrane proteins. Although membrane protein design is in its infancy, it appears more reachable than previously thought.
Collapse
|
25
|
DeMarco KR, Yang PC, Singh V, Furutani K, Dawson JRD, Jeng MT, Fettinger JC, Bekker S, Ngo VA, Noskov SY, Yarov-Yarovoy V, Sack JT, Wulff H, Clancy CE, Vorobyov I. Molecular determinants of pro-arrhythmia proclivity of d- and l-sotalol via a multi-scale modeling pipeline. J Mol Cell Cardiol 2021; 158:163-177. [PMID: 34062207 PMCID: PMC8906354 DOI: 10.1016/j.yjmcc.2021.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/03/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022]
Abstract
Drug isomers may differ in their proarrhythmia risk. An interesting example is the drug sotalol, an antiarrhythmic drug comprising d- and l- enantiomers that both block the hERG cardiac potassium channel and confer differing degrees of proarrhythmic risk. We developed a multi-scale in silico pipeline focusing on hERG channel – drug interactions and used it to probe and predict the mechanisms of pro-arrhythmia risks of the two enantiomers of sotalol. Molecular dynamics (MD) simulations predicted comparable hERG channel binding affinities for d- and l-sotalol, which were validated with electrophysiology experiments. MD derived thermodynamic and kinetic parameters were used to build multi-scale functional computational models of cardiac electrophysiology at the cell and tissue scales. Functional models were used to predict inactivated state binding affinities to recapitulate electrocardiogram (ECG) QT interval prolongation observed in clinical data. Our study demonstrates how modeling and simulation can be applied to predict drug effects from the atom to the rhythm for dl-sotalol and also increased proarrhythmia proclivity of d- vs. l-sotalol when accounting for stereospecific beta-adrenergic receptor blocking.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Kazuharu Furutani
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - John R D Dawson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Biophysics Graduate Group, University of California Davis, Davis, CA 95616, USA
| | - Mao-Tsuen Jeng
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - James C Fettinger
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Slava Bekker
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Science and Engineering, American River College, Sacramento, CA 95841, USA
| | - Van A Ngo
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Sergei Y Noskov
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
26
|
Chen Z, Kuenze G, Meiler J, Canessa CM. An arginine residue in the outer segment of hASIC1a TM1 affects both proton affinity and channel desensitization. J Gen Physiol 2021; 153:211986. [PMID: 33851970 PMCID: PMC8050794 DOI: 10.1085/jgp.202012802] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/14/2020] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Acid-sensing ion channels (ASICs) respond to changes in pH in the central and peripheral nervous systems and participate in synaptic plasticity and pain perception. Understanding the proton-mediated gating mechanism remains elusive despite the of their structures in various conformational states. We report here that R64, an arginine located in the outer segment of the first transmembrane domain of all three isoforms of mammalian ASICs, markedly impacts the apparent proton affinity of activation and the degree of desensitization from the open and preopen states. Rosetta calculations of free energy changes predict that substitutions of R64 in hASIC1a by aromatic residues destabilize the closed conformation while stabilizing the open conformation. Accordingly, F64 enhances the efficacy of proton-mediated gating of hASIC1a, which increases the apparent pH50 and facilitates channel opening when only one or two subunits are activated. F64 also lengthens the duration of opening events, thus keeping channels open for extended periods of time and diminishing low pH-induced desensitization. Our results indicate that activation of a proton sensor(s) with pH50 equal to or greater than pH 7.2–7.1 opens F64hASIC1a, whereas it induces steady-state desensitization in wildtype channels due to the high energy of activation imposed by R64, which prevents opening of the pore. Together, these findings suggest that activation of a high-affinity proton-sensor(s) and a common gating mechanism may mediate the processes of activation and steady-state desensitization of hASIC1a.
Collapse
Affiliation(s)
- Zhuyuan Chen
- Department of Basic Sciences, Tsinghua University School of Medicine, Beijing, China
| | - Georg Kuenze
- Department of Chemistry, Vanderbilt University, Nashville, TN.,Center for Structural Biology, Vanderbilt University, Nashville, TN.,Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN.,Center for Structural Biology, Vanderbilt University, Nashville, TN.,Department of Pharmacology, Vanderbilt University, Nashville, TN.,Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| | - Cecilia M Canessa
- Department of Basic Sciences, Tsinghua University School of Medicine, Beijing, China.,Cellular and Molecular Physiology, Yale University, New Haven, CT
| |
Collapse
|
27
|
Gulsevin A, Meiler J. Prediction of amphipathic helix-membrane interactions with Rosetta. PLoS Comput Biol 2021; 17:e1008818. [PMID: 33730029 PMCID: PMC8007005 DOI: 10.1371/journal.pcbi.1008818] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 03/29/2021] [Accepted: 02/18/2021] [Indexed: 01/17/2023] Open
Abstract
Amphipathic helices have hydrophobic and hydrophilic/charged residues situated on opposite faces of the helix. They can anchor peripheral membrane proteins to the membrane, be attached to integral membrane proteins, or exist as independent peptides. Despite the widespread presence of membrane-interacting amphipathic helices, there is no computational tool within Rosetta to model their interactions with membranes. In order to address this need, we developed the AmphiScan protocol with PyRosetta, which runs a grid search to find the most favorable position of an amphipathic helix with respect to the membrane. The performance of the algorithm was tested in benchmarks with the RosettaMembrane, ref2015_memb, and franklin2019 score functions on six engineered and 44 naturally-occurring amphipathic helices using membrane coordinates from the OPM and PDBTM databases, OREMPRO server, and MD simulations for comparison. The AmphiScan protocol predicted the coordinates of amphipathic helices within less than 3Å of the reference structures and identified membrane-embedded residues with a Matthews Correlation Constant (MCC) of up to 0.57. Overall, AmphiScan stands as fast, accurate, and highly-customizable protocol that can be pipelined with other Rosetta and Python applications. Amphipathic helices are important targets as antibacterial peptides and as domains of membrane proteins that play a role in sensing the membrane environment. Understanding how amphipathic helices interact with membrane enables us to design better peptides and understand how membrane proteins use them to interact with their environment. However, there is a limited number of tools available for the modeling of amphipathic helices in membranes. Implicit membrane models can be used for this purpose as simplistic representations of the membrane environment. In this work, we developed the AmphiScan protocol that can be used to predict membrane coordinates of amphipathic helices starting with a helix structure in an implicit membrane environment. We benchmarked the performance of AmphiScan on engineered LK peptides, naturally-occurring amphipathic helices, and hydrophobic and hydrophilic peptides. Our approach provides a reliable and customizable tool to model amphipathic helix–membrane interactions, and pose a platform for the screening of amphipathic helix properties in silico.
Collapse
Affiliation(s)
- Alican Gulsevin
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jens Meiler
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University Medical School, 04103 Leipzig, Germany
- * E-mail:
| |
Collapse
|
28
|
Winfield I, Barkan K, Routledge S, Robertson NJ, Harris M, Jazayeri A, Simms J, Reynolds CA, Poyner DR, Ladds G. The Role of ICL1 and H8 in Class B1 GPCRs; Implications for Receptor Activation. Front Endocrinol (Lausanne) 2021; 12:792912. [PMID: 35095763 PMCID: PMC8796428 DOI: 10.3389/fendo.2021.792912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
The first intracellular loop (ICL1) of G protein-coupled receptors (GPCRs) has received little attention, although there is evidence that, with the 8th helix (H8), it is involved in early conformational changes following receptor activation as well as contacting the G protein β subunit. In class B1 GPCRs, the distal part of ICL1 contains a conserved R12.48KLRCxR2.46b motif that extends into the base of the second transmembrane helix; this is weakly conserved as a [R/H]12.48KL[R/H] motif in class A GPCRs. In the current study, the role of ICL1 and H8 in signaling through cAMP, iCa2+ and ERK1/2 has been examined in two class B1 GPCRs, using mutagenesis and molecular dynamics. Mutations throughout ICL1 can either enhance or disrupt cAMP production by CGRP at the CGRP receptor. Alanine mutagenesis identified subtle differences with regard elevation of iCa2+, with the distal end of the loop being particularly sensitive. ERK1/2 activation displayed little sensitivity to ICL1 mutation. A broadly similar pattern was observed with the glucagon receptor, although there were differences in significance of individual residues. Extending the study revealed that at the CRF1 receptor, an insertion in ICL1 switched signaling bias between iCa2+ and cAMP. Molecular dynamics suggested that changes in ICL1 altered the conformation of ICL2 and the H8/TM7 junction (ICL4). For H8, alanine mutagenesis showed the importance of E3908.49b for all three signal transduction pathways, for the CGRP receptor, but mutations of other residues largely just altered ERK1/2 activation. Thus, ICL1 may modulate GPCR bias via interactions with ICL2, ICL4 and the Gβ subunit.
Collapse
MESH Headings
- Amino Acid Motifs/physiology
- Calcitonin Receptor-Like Protein/metabolism
- Calcitonin Receptor-Like Protein/physiology
- Calcitonin Receptor-Like Protein/ultrastructure
- Calcium Signaling
- Cyclic AMP/metabolism
- HEK293 Cells
- Humans
- MAP Kinase Signaling System
- Molecular Dynamics Simulation
- Protein Domains
- Protein Structure, Tertiary
- Receptor Activity-Modifying Protein 1/metabolism
- Receptor Activity-Modifying Protein 1/physiology
- Receptor Activity-Modifying Protein 1/ultrastructure
- Receptors, Calcitonin Gene-Related Peptide/metabolism
- Receptors, Calcitonin Gene-Related Peptide/physiology
- Receptors, Calcitonin Gene-Related Peptide/ultrastructure
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Receptors, Corticotropin-Releasing Hormone/physiology
- Receptors, Corticotropin-Releasing Hormone/ultrastructure
- Receptors, G-Protein-Coupled
- Receptors, Glucagon/metabolism
- Receptors, Glucagon/physiology
- Receptors, Glucagon/ultrastructure
Collapse
Affiliation(s)
- Ian Winfield
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Kerry Barkan
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Routledge
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | | | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | | | - John Simms
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | | | - David R. Poyner
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
- *Correspondence: Graham Ladds, ; David R. Poyner,
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Graham Ladds, ; David R. Poyner,
| |
Collapse
|
29
|
Abstract
Protein engineering can yield new molecular tools for nanotechnology and therapeutic applications through modulating physiochemical and biological properties. Engineering membrane proteins is especially attractive because they perform key cellular processes including transport, nutrient uptake, removal of toxins, respiration, motility, and signaling. In this chapter, we describe two protocols for membrane protein engineering with the Rosetta software: (1) ΔΔG calculations for single point mutations and (2) sequence optimization in different membrane lipid compositions. These modular protocols are easily adaptable for more complex problems and serve as a foundation for efficient membrane protein engineering calculations.
Collapse
Affiliation(s)
- Rebecca F Alford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
30
|
Zhang Q, Zhu J, Ju F, Kong L, Sun S, Zheng WM, Bu D. ISSEC: inferring contacts among protein secondary structure elements using deep object detection. BMC Bioinformatics 2020; 21:503. [PMID: 33153432 PMCID: PMC7643357 DOI: 10.1186/s12859-020-03793-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/30/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The formation of contacts among protein secondary structure elements (SSEs) is an important step in protein folding as it determines topology of protein tertiary structure; hence, inferring inter-SSE contacts is crucial to protein structure prediction. One of the existing strategies infers inter-SSE contacts directly from the predicted possibilities of inter-residue contacts without any preprocessing, and thus suffers from the excessive noises existing in the predicted inter-residue contacts. Another strategy defines SSEs based on protein secondary structure prediction first, and then judges whether each candidate SSE pair could form contact or not. However, it is difficult to accurately determine boundary of SSEs due to the errors in secondary structure prediction. The incorrectly-deduced SSEs definitely hinder subsequent prediction of the contacts among them. RESULTS We here report an accurate approach to infer the inter-SSE contacts (thus called as ISSEC) using the deep object detection technique. The design of ISSEC is based on the observation that, in the inter-residue contact map, the contacting SSEs usually form rectangle regions with characteristic patterns. Therefore, ISSEC infers inter-SSE contacts through detecting such rectangle regions. Unlike the existing approach directly using the predicted probabilities of inter-residue contact, ISSEC applies the deep convolution technique to extract high-level features from the inter-residue contacts. More importantly, ISSEC does not rely on the pre-defined SSEs. Instead, ISSEC enumerates multiple candidate rectangle regions in the predicted inter-residue contact map, and for each region, ISSEC calculates a confidence score to measure whether it has characteristic patterns or not. ISSEC employs greedy strategy to select non-overlapping regions with high confidence score, and finally infers inter-SSE contacts according to these regions. CONCLUSIONS Comprehensive experimental results suggested that ISSEC outperformed the state-of-the-art approaches in predicting inter-SSE contacts. We further demonstrated the successful applications of ISSEC to improve prediction of both inter-residue contacts and tertiary structure as well.
Collapse
Affiliation(s)
- Qi Zhang
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jianwei Zhu
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China
| | - Fusong Ju
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China
| | - Lupeng Kong
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shiwei Sun
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wei-Mou Zheng
- Institute of Theoretical Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Dongbo Bu
- Key Lab of Intelligent Information Processing, Big Data Academy, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Computer Science, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Kuenze G, Vanoye CG, Desai RR, Adusumilli S, Brewer KR, Woods H, McDonald EF, Sanders CR, George AL, Meiler J. Allosteric mechanism for KCNE1 modulation of KCNQ1 potassium channel activation. eLife 2020; 9:57680. [PMID: 33095155 PMCID: PMC7584456 DOI: 10.7554/elife.57680] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/28/2020] [Indexed: 01/04/2023] Open
Abstract
The function of the voltage-gated KCNQ1 potassium channel is regulated by co-assembly with KCNE auxiliary subunits. KCNQ1-KCNE1 channels generate the slow delayed rectifier current, IKs, which contributes to the repolarization phase of the cardiac action potential. A three amino acid motif (F57-T58-L59, FTL) in KCNE1 is essential for slow activation of KCNQ1-KCNE1 channels. However, how this motif interacts with KCNQ1 to control its function is unknown. Combining computational modeling with electrophysiological studies, we developed structural models of the KCNQ1-KCNE1 complex that suggest how KCNE1 controls KCNQ1 activation. The FTL motif binds at a cleft between the voltage-sensing and pore domains and appears to affect the channel gate by an allosteric mechanism. Comparison with the KCNQ1-KCNE3 channel structure suggests a common transmembrane-binding mode for different KCNEs and illuminates how specific differences in the interaction of their triplet motifs determine the profound differences in KCNQ1 functional modulation by KCNE1 versus KCNE3.
Collapse
Affiliation(s)
- Georg Kuenze
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States.,Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| | - Carlos G Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Reshma R Desai
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Sneha Adusumilli
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Kathryn R Brewer
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Biochemistry, Vanderbilt University, Nashville, United States
| | - Hope Woods
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Eli F McDonald
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Charles R Sanders
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Biochemistry, Vanderbilt University, Nashville, United States
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States.,Institute for Drug Discovery, Leipzig University, Leipzig, Germany.,Department of Pharmacology, Vanderbilt University, Nashville, United States
| |
Collapse
|
32
|
Xiao Y, Zeng B, Berner N, Frishman D, Langosch D, George Teese M. Experimental determination and data-driven prediction of homotypic transmembrane domain interfaces. Comput Struct Biotechnol J 2020; 18:3230-3242. [PMID: 33209210 PMCID: PMC7649602 DOI: 10.1016/j.csbj.2020.09.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022] Open
Abstract
Homotypic TMD interfaces identified by different techniques share strong similarities. The GxxxG motif is the feature most strongly associated with interfaces. Other features include conservation, polarity, coevolution, and depth in the membrane The role of each of each feature strongly depends on the individual protein. Machine-learning helps predict interfaces from evolutionary sequence data
Interactions between their transmembrane domains (TMDs) frequently support the assembly of single-pass membrane proteins to non-covalent complexes. Yet, the TMD-TMD interactome remains largely uncharted. With a view to predicting homotypic TMD-TMD interfaces from primary structure, we performed a systematic analysis of their physical and evolutionary properties. To this end, we generated a dataset of 50 self-interacting TMDs. This dataset contains interfaces of nine TMDs from bitopic human proteins (Ire1, Armcx6, Tie1, ATP1B1, PTPRO, PTPRU, PTPRG, DDR1, and Siglec7) that were experimentally identified here and combined with literature data. We show that interfacial residues of these homotypic TMD-TMD interfaces tend to be more conserved, coevolved and polar than non-interfacial residues. Further, we suggest for the first time that interface positions are deficient in β-branched residues, and likely to be located deep in the hydrophobic core of the membrane. Overrepresentation of the GxxxG motif at interfaces is strong, but that of (small)xxx(small) motifs is weak. The multiplicity of these features and the individual character of TMD-TMD interfaces, as uncovered here, prompted us to train a machine learning algorithm. The resulting prediction method, THOIPA (www.thoipa.org), excels in the prediction of key interface residues from evolutionary sequence data.
Collapse
Affiliation(s)
- Yao Xiao
- Center for Integrated Protein Science Munich (CIPSM) at the Lehrstuhl für Chemie der Biopolymere, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Bo Zeng
- Department of Bioinformatics, Wissenschaftszentrum, Weihenstephan, Maximus-von-Imhof-Forum 3, Freising 85354, Germany
| | - Nicola Berner
- Center for Integrated Protein Science Munich (CIPSM) at the Lehrstuhl für Chemie der Biopolymere, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Dmitrij Frishman
- Department of Bioinformatics, Wissenschaftszentrum, Weihenstephan, Maximus-von-Imhof-Forum 3, Freising 85354, Germany.,Department of Bioinformatics, Peter the Great Saint Petersburg Polytechnic University, St. Petersburg 195251, Russian Federation
| | - Dieter Langosch
- Center for Integrated Protein Science Munich (CIPSM) at the Lehrstuhl für Chemie der Biopolymere, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Mark George Teese
- Center for Integrated Protein Science Munich (CIPSM) at the Lehrstuhl für Chemie der Biopolymere, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany.,TNG Technology Consulting GmbH, Beta-Straße 13a, 85774 Unterföhring, Germany
| |
Collapse
|
33
|
Determinants of Endoplasmic Reticulum-to-Lipid Droplet Protein Targeting. Dev Cell 2020; 54:471-487.e7. [PMID: 32730754 DOI: 10.1016/j.devcel.2020.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/11/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023]
Abstract
Lipid droplet (LD) formation from the endoplasmic reticulum (ER) is accompanied by the targeting and accumulation of specific hydrophobic, membrane-embedded proteins on LDs. The determinants of this process are unknown. Here, we study the hydrophobic membrane motifs of two Drosophila melanogaster proteins, GPAT4 and ALG14, that utilize this pathway, and we identify crucial sequence features that mediate LD accumulation. Molecular dynamics simulations and studies in cells reveal that LD targeting of these motifs requires deeply inserted tryptophans that have lower free energy in the LD oil phase and positively charged residues near predicted hairpin hinges that become less constrained in the LD environment. Analyzing hydrophobic motifs from similar LD-targeting proteins, it appears that the distribution of tryptophan and positively charged residues distinguishes them from non-LD-targeting membrane motifs. Our studies identify specific sequence features and principles of hydrophobic membrane motifs that mediate their accumulation on LDs.
Collapse
|
34
|
Leman JK, Weitzner BD, Lewis SM, Adolf-Bryfogle J, Alam N, Alford RF, Aprahamian M, Baker D, Barlow KA, Barth P, Basanta B, Bender BJ, Blacklock K, Bonet J, Boyken SE, Bradley P, Bystroff C, Conway P, Cooper S, Correia BE, Coventry B, Das R, De Jong RM, DiMaio F, Dsilva L, Dunbrack R, Ford AS, Frenz B, Fu DY, Geniesse C, Goldschmidt L, Gowthaman R, Gray JJ, Gront D, Guffy S, Horowitz S, Huang PS, Huber T, Jacobs TM, Jeliazkov JR, Johnson DK, Kappel K, Karanicolas J, Khakzad H, Khar KR, Khare SD, Khatib F, Khramushin A, King IC, Kleffner R, Koepnick B, Kortemme T, Kuenze G, Kuhlman B, Kuroda D, Labonte JW, Lai JK, Lapidoth G, Leaver-Fay A, Lindert S, Linsky T, London N, Lubin JH, Lyskov S, Maguire J, Malmström L, Marcos E, Marcu O, Marze NA, Meiler J, Moretti R, Mulligan VK, Nerli S, Norn C, Ó'Conchúir S, Ollikainen N, Ovchinnikov S, Pacella MS, Pan X, Park H, Pavlovicz RE, Pethe M, Pierce BG, Pilla KB, Raveh B, Renfrew PD, Burman SSR, Rubenstein A, Sauer MF, Scheck A, Schief W, Schueler-Furman O, Sedan Y, Sevy AM, Sgourakis NG, Shi L, Siegel JB, Silva DA, Smith S, Song Y, et alLeman JK, Weitzner BD, Lewis SM, Adolf-Bryfogle J, Alam N, Alford RF, Aprahamian M, Baker D, Barlow KA, Barth P, Basanta B, Bender BJ, Blacklock K, Bonet J, Boyken SE, Bradley P, Bystroff C, Conway P, Cooper S, Correia BE, Coventry B, Das R, De Jong RM, DiMaio F, Dsilva L, Dunbrack R, Ford AS, Frenz B, Fu DY, Geniesse C, Goldschmidt L, Gowthaman R, Gray JJ, Gront D, Guffy S, Horowitz S, Huang PS, Huber T, Jacobs TM, Jeliazkov JR, Johnson DK, Kappel K, Karanicolas J, Khakzad H, Khar KR, Khare SD, Khatib F, Khramushin A, King IC, Kleffner R, Koepnick B, Kortemme T, Kuenze G, Kuhlman B, Kuroda D, Labonte JW, Lai JK, Lapidoth G, Leaver-Fay A, Lindert S, Linsky T, London N, Lubin JH, Lyskov S, Maguire J, Malmström L, Marcos E, Marcu O, Marze NA, Meiler J, Moretti R, Mulligan VK, Nerli S, Norn C, Ó'Conchúir S, Ollikainen N, Ovchinnikov S, Pacella MS, Pan X, Park H, Pavlovicz RE, Pethe M, Pierce BG, Pilla KB, Raveh B, Renfrew PD, Burman SSR, Rubenstein A, Sauer MF, Scheck A, Schief W, Schueler-Furman O, Sedan Y, Sevy AM, Sgourakis NG, Shi L, Siegel JB, Silva DA, Smith S, Song Y, Stein A, Szegedy M, Teets FD, Thyme SB, Wang RYR, Watkins A, Zimmerman L, Bonneau R. Macromolecular modeling and design in Rosetta: recent methods and frameworks. Nat Methods 2020; 17:665-680. [PMID: 32483333 PMCID: PMC7603796 DOI: 10.1038/s41592-020-0848-2] [Show More Authors] [Citation(s) in RCA: 494] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022]
Abstract
The Rosetta software for macromolecular modeling, docking and design is extensively used in laboratories worldwide. During two decades of development by a community of laboratories at more than 60 institutions, Rosetta has been continuously refactored and extended. Its advantages are its performance and interoperability between broad modeling capabilities. Here we review tools developed in the last 5 years, including over 80 methods. We discuss improvements to the score function, user interfaces and usability. Rosetta is available at http://www.rosettacommons.org.
Collapse
Affiliation(s)
- Julia Koehler Leman
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA.
- Department of Biology, New York University, New York, New York, USA.
| | - Brian D Weitzner
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Lyell Immunopharma Inc., Seattle, WA, USA
| | - Steven M Lewis
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry, Duke University, Durham, NC, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Jared Adolf-Bryfogle
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rebecca F Alford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Melanie Aprahamian
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kyle A Barlow
- Graduate Program in Bioinformatics, University of California San Francisco, San Francisco, CA, USA
| | - Patrick Barth
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Baylor College of Medicine, Department of Pharmacology, Houston, TX, USA
| | - Benjamin Basanta
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Biological Physics Structure and Design PhD Program, University of Washington, Seattle, WA, USA
| | - Brian J Bender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Kristin Blacklock
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jaume Bonet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Scott E Boyken
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Lyell Immunopharma Inc., Seattle, WA, USA
| | - Phil Bradley
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chris Bystroff
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Patrick Conway
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Seth Cooper
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rhiju Das
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lorna Dsilva
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Roland Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Alexander S Ford
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Brandon Frenz
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Darwin Y Fu
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Caleb Geniesse
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ragul Gowthaman
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Dominik Gront
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Sharon Guffy
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott Horowitz
- Department of Chemistry & Biochemistry, University of Denver, Denver, CO, USA
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Po-Ssu Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Thomas Huber
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Tim M Jacobs
- Program in Bioinformatics and Computational Biology, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - David K Johnson
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| | - Kalli Kappel
- Biophysics Program, Stanford University, Stanford, CA, USA
| | - John Karanicolas
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hamed Khakzad
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute for Computational Science, University of Zurich, Zurich, Switzerland
- S3IT, University of Zurich, Zurich, Switzerland
| | - Karen R Khar
- Cyrus Biotechnology, Seattle, WA, USA
- Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| | - Sagar D Khare
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, NJ, USA
- Center for Integrative Proteomics Research, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Computational Biology and Molecular Biophysics Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Firas Khatib
- Department of Computer and Information Science, University of Massachusetts Dartmouth, Dartmouth, MA, USA
| | - Alisa Khramushin
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Indigo C King
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Robert Kleffner
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Brian Koepnick
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Georg Kuenze
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daisuke Kuroda
- Medical Device Development and Regulation Research Center, School of Engineering, University of Tokyo, Tokyo, Japan
- Department of Bioengineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Jason W Labonte
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, USA
| | - Jason K Lai
- Baylor College of Medicine, Department of Pharmacology, Houston, TX, USA
| | - Gideon Lapidoth
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Andrew Leaver-Fay
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA
| | - Thomas Linsky
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Nir London
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joseph H Lubin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sergey Lyskov
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jack Maguire
- Program in Bioinformatics and Computational Biology, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lars Malmström
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute for Computational Science, University of Zurich, Zurich, Switzerland
- S3IT, University of Zurich, Zurich, Switzerland
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Enrique Marcos
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Research in Biomedicine Barcelona, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Orly Marcu
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nicholas A Marze
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Departments of Chemistry, Pharmacology and Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
- Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Rocco Moretti
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Vikram Khipple Mulligan
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Santrupti Nerli
- Department of Computer Science, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Christoffer Norn
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shane Ó'Conchúir
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Noah Ollikainen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sergey Ovchinnikov
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Michael S Pacella
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Xingjie Pan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Hahnbeom Park
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Ryan E Pavlovicz
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Manasi Pethe
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, NJ, USA
- Center for Integrative Proteomics Research, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brian G Pierce
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Kala Bharath Pilla
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Barak Raveh
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - P Douglas Renfrew
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Shourya S Roy Burman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Aliza Rubenstein
- Institute of Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Computational Biology and Molecular Biophysics Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marion F Sauer
- Chemical and Physical Biology Program, Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN, USA
| | - Andreas Scheck
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - William Schief
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Sedan
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander M Sevy
- Chemical and Physical Biology Program, Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN, USA
| | - Nikolaos G Sgourakis
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Lei Shi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justin B Siegel
- Department of Chemistry, University of California, Davis, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
- Genome Center, University of California, Davis, Davis, CA, USA
| | | | - Shannon Smith
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Yifan Song
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Cyrus Biotechnology, Seattle, WA, USA
| | - Amelie Stein
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Maria Szegedy
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Frank D Teets
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Summer B Thyme
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Ray Yu-Ruei Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Andrew Watkins
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Lior Zimmerman
- Department of Microbiology and Molecular Genetics, IMRIC, Ein Kerem Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA.
- Department of Biology, New York University, New York, New York, USA.
- Department of Computer Science, New York University, New York, NY, USA.
- Center for Data Science, New York University, New York, NY, USA.
| |
Collapse
|
35
|
Alford RF, Smolin N, Young HS, Gray JJ, Robia SL. Protein docking and steered molecular dynamics suggest alternative phospholamban-binding sites on the SERCA calcium transporter. J Biol Chem 2020; 295:11262-11274. [PMID: 32554805 DOI: 10.1074/jbc.ra120.012948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/16/2020] [Indexed: 01/27/2023] Open
Abstract
The transport activity of the sarco(endo)plasmic reticulum calcium ATPase (SERCA) in cardiac myocytes is modulated by an inhibitory interaction with a transmembrane peptide, phospholamban (PLB). Previous biochemical studies have revealed that PLB interacts with a specific inhibitory site on SERCA, and low-resolution structural evidence suggests that PLB interacts with distinct alternative sites on SERCA. High-resolution details of the structural determinants of SERCA regulation have been elusive because of the dynamic nature of the regulatory complex. In this study, we used computational approaches to develop a structural model of SERCA-PLB interactions to gain a mechanistic understanding of PLB-mediated SERCA transport regulation. We combined steered molecular dynamics and membrane protein-protein docking experiments to achieve both a global search and all-atom force calculations to determine the relative affinities of PLB for candidate sites on SERCA. We modeled the binding of PLB to several SERCA conformations, representing different enzymatic states sampled during the calcium transport catalytic cycle. The results of the steered molecular dynamics and docking experiments indicated that the canonical PLB-binding site (comprising transmembrane helices M2, M4, and M9) is the preferred site. This preference was even more stringent for a superinhibitory PLB variant. Interestingly, PLB-binding specificity became more ambivalent for other SERCA conformers. These results provide evidence for polymorphic PLB interactions with novel sites on M3 and with the outside of the SERCA helix M9. Our findings are compatible with previous physical measurements that suggest that PLB interacts with multiple binding sites, conferring dynamic responsiveness to changing physiological conditions.
Collapse
Affiliation(s)
- Rebecca F Alford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nikolai Smolin
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
36
|
Protein Structure Prediction and Design in a Biologically Realistic Implicit Membrane. Biophys J 2020; 118:2042-2055. [PMID: 32224301 DOI: 10.1016/j.bpj.2020.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Accepted: 03/09/2020] [Indexed: 11/19/2022] Open
Abstract
Protein design is a powerful tool for elucidating mechanisms of function and engineering new therapeutics and nanotechnologies. Although soluble protein design has advanced, membrane protein design remains challenging because of difficulties in modeling the lipid bilayer. In this work, we developed an implicit approach that captures the anisotropic structure, shape of water-filled pores, and nanoscale dimensions of membranes with different lipid compositions. The model improves performance in computational benchmarks against experimental targets, including prediction of protein orientations in the bilayer, ΔΔG calculations, native structure discrimination, and native sequence recovery. When applied to de novo protein design, this approach designs sequences with an amino acid distribution near the native amino acid distribution in membrane proteins, overcoming a critical flaw in previous membrane models that were prone to generating leucine-rich designs. Furthermore, the proteins designed in the new membrane model exhibit native-like features including interfacial aromatic side chains, hydrophobic lengths compatible with bilayer thickness, and polar pores. Our method advances high-resolution membrane protein structure prediction and design toward tackling key biological questions and engineering challenges.
Collapse
|
37
|
Yang PC, DeMarco KR, Aghasafari P, Jeng MT, Dawson JRD, Bekker S, Noskov SY, Yarov-Yarovoy V, Vorobyov I, Clancy CE. A Computational Pipeline to Predict Cardiotoxicity: From the Atom to the Rhythm. Circ Res 2020; 126:947-964. [PMID: 32091972 DOI: 10.1161/circresaha.119.316404] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Drug-induced proarrhythmia is so tightly associated with prolongation of the QT interval that QT prolongation is an accepted surrogate marker for arrhythmia. But QT interval is too sensitive a marker and not selective, resulting in many useful drugs eliminated in drug discovery. OBJECTIVE To predict the impact of a drug from the drug chemistry on the cardiac rhythm. METHODS AND RESULTS In a new linkage, we connected atomistic scale information to protein, cell, and tissue scales by predicting drug-binding affinities and rates from simulation of ion channel and drug structure interactions and then used these values to model drug effects on the hERG channel. Model components were integrated into predictive models at the cell and tissue scales to expose fundamental arrhythmia vulnerability mechanisms and complex interactions underlying emergent behaviors. Human clinical data were used for model framework validation and showed excellent agreement, demonstrating feasibility of a new approach for cardiotoxicity prediction. CONCLUSIONS We present a multiscale model framework to predict electrotoxicity in the heart from the atom to the rhythm. Novel mechanistic insights emerged at all scales of the system, from the specific nature of proarrhythmic drug interaction with the hERG channel, to the fundamental cellular and tissue-level arrhythmia mechanisms. Applications of machine learning indicate necessary and sufficient parameters that predict arrhythmia vulnerability. We expect that the model framework may be expanded to make an impact in drug discovery, drug safety screening for a variety of compounds and targets, and in a variety of regulatory processes.
Collapse
Affiliation(s)
- Pei-Chi Yang
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Kevin R DeMarco
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Parya Aghasafari
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Mao-Tsuen Jeng
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - John R D Dawson
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Biophysics Graduate Group (J.R.D.D.), University of California Davis
| | - Slava Bekker
- Department of Science and Engineering, American River College, Sacramento, CA (S.B.)
| | - Sergei Y Noskov
- Faculty of Science, Centre for Molecular Simulations and Department of Biological Sciences, University of Calgary, Alberta, Canada (S.Y.N.)
| | - Vladimir Yarov-Yarovoy
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Igor Vorobyov
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Department of Pharmacology (I.V., C.E.C.), University of California Davis
| | - Colleen E Clancy
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Department of Pharmacology (I.V., C.E.C.), University of California Davis
| |
Collapse
|
38
|
Chen KYM, Keri D, Barth P. Computational design of G Protein-Coupled Receptor allosteric signal transductions. Nat Chem Biol 2019; 16:77-86. [PMID: 31792443 DOI: 10.1038/s41589-019-0407-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
Membrane receptors sense and transduce extracellular stimuli into intracellular signaling responses but the molecular underpinnings remain poorly understood. We report a computational approach for designing protein allosteric signaling functions. By combining molecular dynamics simulations and design calculations, the method engineers amino-acid 'microswitches' at allosteric sites that modulate receptor stability or long-range coupling, to reprogram specific signaling properties. We designed 36 dopamine D2 receptor variants, whose constitutive and ligand-induced signaling agreed well with our predictions, repurposed the D2 receptor into a serotonin biosensor and predicted the signaling effects of more than 100 known G-protein-coupled receptor (GPCR) mutations. Our results reveal the existence of distinct classes of allosteric microswitches and pathways that define an unforeseen molecular mechanism of regulation and evolution of GPCR signaling. Our approach enables the rational design of allosteric receptors with enhanced stability and function to facilitate structural characterization, and reprogram cellular signaling in synthetic biology and cell engineering applications.
Collapse
Affiliation(s)
- Kuang-Yui Michael Chen
- Swiss Federal Institute of Technology (EPFL), Institute of Bioengineering, Lausanne, Switzerland.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, CA, USA
| | - Daniel Keri
- Swiss Federal Institute of Technology (EPFL), Institute of Bioengineering, Lausanne, Switzerland
| | - Patrick Barth
- Swiss Federal Institute of Technology (EPFL), Institute of Bioengineering, Lausanne, Switzerland. .,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA. .,Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA. .,Structural and Computational Biology and Molecular Biophysics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
39
|
Sisco NJ, Helsell CVM, Van Horn WD. Competitive Interactions between PIRT, the Cold Sensing Ion Channel TRPM8, and PIP 2 Suggest a Mechanism for Regulation. Sci Rep 2019; 9:14128. [PMID: 31575973 PMCID: PMC6773951 DOI: 10.1038/s41598-019-49912-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/02/2019] [Indexed: 01/18/2023] Open
Abstract
TRPM8 is a member of the transient receptor potential ion channel family where it functions as a cold and pain sensor in humans and other higher organisms. Previous studies show that TRPM8 requires the signaling phosphoinositide lipid PIP2 to function. TRPM8 function is further regulated by other diverse mechanisms, including the small modulatory membrane protein PIRT (phosphoinositide regulator of TRP). Like TRPM8, PIRT also binds PIP2 and behavioral studies have shown that PIRT is required for normal TRPM8-mediated cold-sensing. To better understand the molecular mechanism of PIRT regulation of TRPM8, solution nuclear magnetic resonance (NMR) spectroscopy was used to assign the backbone resonances of full-length human PIRT and investigate the direct binding of PIRT to PIP2 and the human TRPM8 S1-S4 transmembrane domain. Microscale thermophoresis (MST) binding studies validate the NMR results and identify a competitive PIRT interaction between PIP2 and the TRPM8 S1-S4 domain. Computational PIP2 docking to a human TRPM8 comparative model was performed to help localize where PIRT may bind TRPM8. Taken together, our data suggest a mechanism where TRPM8, PIRT, and PIP2 form a regulatory complex and PIRT modulation of TRPM8 arises, at least in part, by regulating local concentrations of PIP2 accessible to TRPM8.
Collapse
Affiliation(s)
- Nicholas J Sisco
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
- The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
- The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85281, USA
- The Magnetic Resonance Research Center, Arizona State University, Tempe, AZ, 85287, USA
| | - Cole V M Helsell
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
- The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
- The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85281, USA
- The Magnetic Resonance Research Center, Arizona State University, Tempe, AZ, 85287, USA
| | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA.
- The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA.
- The Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85281, USA.
- The Magnetic Resonance Research Center, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
40
|
Kuenze G, Duran AM, Woods H, Brewer KR, McDonald EF, Vanoye CG, George AL, Sanders CR, Meiler J. Upgraded molecular models of the human KCNQ1 potassium channel. PLoS One 2019; 14:e0220415. [PMID: 31518351 PMCID: PMC6743773 DOI: 10.1371/journal.pone.0220415] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/15/2019] [Indexed: 11/29/2022] Open
Abstract
The voltage-gated potassium channel KCNQ1 (KV7.1) assembles with the KCNE1 accessory protein to generate the slow delayed rectifier current, IKS, which is critical for membrane repolarization as part of the cardiac action potential. Loss-of-function (LOF) mutations in KCNQ1 are the most common cause of congenital long QT syndrome (LQTS), type 1 LQTS, an inherited genetic predisposition to cardiac arrhythmia and sudden cardiac death. A detailed structural understanding of KCNQ1 is needed to elucidate the molecular basis for KCNQ1 LOF in disease and to enable structure-guided design of new anti-arrhythmic drugs. In this work, advanced structural models of human KCNQ1 in the resting/closed and activated/open states were developed by Rosetta homology modeling guided by newly available experimentally-based templates: X. leavis KCNQ1 and various resting voltage sensor structures. Using molecular dynamics (MD) simulations, the capacity of the models to describe experimentally established channel properties including state-dependent voltage sensor gating charge interactions and pore conformations, PIP2 binding sites, and voltage sensor–pore domain interactions were validated. Rosetta energy calculations were applied to assess the utility of each model in interpreting mutation-evoked KCNQ1 dysfunction by predicting the change in protein thermodynamic stability for 50 experimentally characterized KCNQ1 variants with mutations located in the voltage-sensing domain. Energetic destabilization was successfully predicted for folding-defective KCNQ1 LOF mutants whereas wild type-like mutants exhibited no significant energetic frustrations, which supports growing evidence that mutation-induced protein destabilization is an especially common cause of KCNQ1 dysfunction. The new KCNQ1 Rosetta models provide helpful tools in the study of the structural basis for KCNQ1 function and can be used to generate hypotheses to explain KCNQ1 dysfunction.
Collapse
Affiliation(s)
- Georg Kuenze
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Amanda M. Duran
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Hope Woods
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Kathryn R. Brewer
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Eli Fritz McDonald
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Alfred L. George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Charles R. Sanders
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
41
|
Yin Y, Dong Y, Vu S, Yang F, Yarov‐Yarovoy V, Tian Y, Zheng J. Structural mechanisms underlying activation of TRPV1 channels by pungent compounds in gingers. Br J Pharmacol 2019; 176:3364-3377. [PMID: 31207668 PMCID: PMC6692589 DOI: 10.1111/bph.14766] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Like chili peppers, gingers produce pungent stimuli by a group of vanilloid compounds that activate the nociceptive transient receptor potential vanilloid 1 (TRPV1) ion channel. How these compounds interact with TRPV1 remains unclear. EXPERIMENTAL APPROACH We used computational structural modelling, functional tests (electrophysiology and calcium imaging), and mutagenesis to investigate the structural mechanisms underlying ligand-channel interactions. KEY RESULTS The potency of three principal pungent compounds from ginger -shogaol, gingerol, and zingerone-depends on the same two residues in the TRPV1 channel that form a hydrogen bond with the chili pepper pungent compound, capsaicin. Computational modelling revealed binding poses of these ginger compounds similar to those of capsaicin, including a "head-down tail-up" orientation, two specific hydrogen bonds, and important contributions of van der Waals interactions by the aliphatic tail. Our study also identified a novel horizontal binding pose of zingerone that allows it to directly interact with the channel pore when bound inside the ligand-binding pocket. These observations offer a molecular level explanation for how unique structures in the ginger compounds affect their channel activation potency. CONCLUSIONS AND IMPLICATIONS Mechanistic insights into the interactions of ginger compounds and the TRPV1 cation channel should help guide drug discovery efforts to modulate nociception.
Collapse
Affiliation(s)
- Yue Yin
- Department of PharmacologyQingdao University School of PharmacyQingdaoShandongChina
| | - Yawen Dong
- Department of PharmacologyQingdao University School of PharmacyQingdaoShandongChina
| | - Simon Vu
- Department of Physiology and Membrane BiologyUC Davis School of MedicineDavisCAUSA
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | | | - Yuhua Tian
- Department of PharmacologyQingdao University School of PharmacyQingdaoShandongChina
| | - Jie Zheng
- Department of Physiology and Membrane BiologyUC Davis School of MedicineDavisCAUSA
| |
Collapse
|
42
|
Weinstein JY, Elazar A, Fleishman SJ. A lipophilicity-based energy function for membrane-protein modelling and design. PLoS Comput Biol 2019; 15:e1007318. [PMID: 31461441 PMCID: PMC6736313 DOI: 10.1371/journal.pcbi.1007318] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/10/2019] [Accepted: 08/01/2019] [Indexed: 01/14/2023] Open
Abstract
Membrane-protein design is an exciting and increasingly successful research area which has led to landmarks including the design of stable and accurate membrane-integral proteins based on coiled-coil motifs. Design of topologically more complex proteins, such as most receptors, channels, and transporters, however, demands an energy function that balances contributions from intra-protein contacts and protein-membrane interactions. Recent advances in water-soluble all-atom energy functions have increased the accuracy in structure-prediction benchmarks. The plasma membrane, however, imposes different physical constraints on protein solvation. To understand these constraints, we recently developed a high-throughput experimental screen, called dsTβL, and inferred apparent insertion energies for each amino acid at dozens of positions across the bacterial plasma membrane. Here, we express these profiles as lipophilicity energy terms in Rosetta and demonstrate that the new energy function outperforms previous ones in modelling and design benchmarks. Rosetta ab initio simulations starting from an extended chain recapitulate two-thirds of the experimentally determined structures of membrane-spanning homo-oligomers with <2.5Å root-mean-square deviation within the top-predicted five models (available online: http://tmhop.weizmann.ac.il). Furthermore, in two sequence-design benchmarks, the energy function improves discrimination of stabilizing point mutations and recapitulates natural membrane-protein sequences of known structure, thereby recommending this new energy function for membrane-protein modelling and design.
Collapse
Affiliation(s)
| | - Assaf Elazar
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Sarel Jacob Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
43
|
Xia Y, Ledwitch K, Kuenze G, Duran A, Li J, Sanders CR, Manning C, Meiler J. A unified structural model of the mammalian translocator protein (TSPO). JOURNAL OF BIOMOLECULAR NMR 2019; 73:347-364. [PMID: 31243635 PMCID: PMC8006375 DOI: 10.1007/s10858-019-00257-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/10/2019] [Indexed: 05/10/2023]
Abstract
The translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), is a membrane protein located on the outer mitochondrial membrane. Experimentally-derived structures of mouse TSPO (mTSPO) and its homologs from bacterial species have been determined by NMR spectroscopy and X-ray crystallography, respectively. These structures and ligand interactions within the TSPO binding pocket display distinct differences. Here, we leverage experimental and computational studies to derive a unified structural model of mTSPO in the presence and absence of the TSPO ligand, PK11195, and study the effects of DPC detergent micelles on the TSPO structure and ligand binding. From this work, we conclude that that the lipid-mimetic system used to solubilize mTSPO for NMR studies thermodynamically destabilizes the protein, introduces structural perturbations, and alters the characteristics of ligand binding. Furthermore, we used Rosetta to construct a unified mTSPO model that reconciles deviating features of the mammalian and bacterial TSPO. These deviating features are likely a consequence of the detergent system used for structure determination of mTSPO by NMR. The unified mTSPO model agrees with available experimental NMR data, appears to be physically realistic (i.e. thermodynamically not frustrated as judged by the Rosetta energy function), and simultaneously shares the structural features observed in sequence-conserved regions of the bacterial proteins. Finally, we identified the binding site for an imaging ligand VUIIS8310 that is currently positioned for clinical translation using NMR spectroscopy and propose a computational model of the VUIIS8310-mTSPO complex.
Collapse
Affiliation(s)
- Yan Xia
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Kaitlyn Ledwitch
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Georg Kuenze
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Amanda Duran
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jun Li
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Charles R Sanders
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37240, USA
| | - Charles Manning
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA.
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, MRBIII 5144B, Nashville, TN, 37232, USA.
| |
Collapse
|
44
|
Katayama K, Gulati S, Ortega JT, Alexander NS, Sun W, Shenouda MM, Palczewski K, Jastrzebska B. Specificity of the chromophore-binding site in human cone opsins. J Biol Chem 2019; 294:6082-6093. [PMID: 30770468 DOI: 10.1074/jbc.ra119.007587] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Indexed: 01/21/2023] Open
Abstract
The variable composition of the chromophore-binding pocket in visual receptors is essential for vision. The visual phototransduction starts with the cis-trans isomerization of the retinal chromophore upon absorption of photons. Despite sharing the common 11-cis-retinal chromophore, rod and cone photoreceptors possess distinct photochemical properties. Thus, a detailed molecular characterization of the chromophore-binding pocket of these receptors is critical to understanding the differences in the photochemistry of vision between rods and cones. Unlike for rhodopsin (Rh), the crystal structures of cone opsins remain to be determined. To obtain insights into the specific chromophore-protein interactions that govern spectral tuning in human visual pigments, here we harnessed the unique binding properties of 11-cis-6-membered-ring-retinal (11-cis-6mr-retinal) with human blue, green, and red cone opsins. To unravel the specificity of the chromophore-binding pocket of cone opsins, we applied 11-cis-6mr-retinal analog-binding analyses to human blue, green, and red cone opsins. Our results revealed that among the three cone opsins, only blue cone opsin can accommodate the 11-cis-6mr-retinal in its chromophore-binding pocket, resulting in the formation of a synthetic blue pigment (B6mr) that absorbs visible light. A combination of primary sequence alignment, molecular modeling, and mutagenesis experiments revealed the specific amino acid residue 6.48 (Tyr-262 in blue cone opsins and Trp-281 in green and red cone opsins) as a selectivity filter in human cone opsins. Altogether, the results of our study uncover the molecular basis underlying the binding selectivity of 11-cis-6mr-retinal to the cone opsins.
Collapse
Affiliation(s)
- Kota Katayama
- From the Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Department of Life Science and Applied Chemistry, Showa-ku, Nagoya 466-8555, Japan; OptoBio Technology Research Center, Nagoya Institute of Technology, Showa-ku, Nagoya 466-8555, Japan
| | - Sahil Gulati
- Gavin Herbert Eye Institute and the Department of Ophthalmology, University of California, Irvine, California 92697
| | - Joseph T Ortega
- From the Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nathan S Alexander
- From the Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wenyu Sun
- Polgenix Inc., Cleveland, Ohio 44106
| | - Marina M Shenouda
- From the Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute and the Department of Ophthalmology, University of California, Irvine, California 92697; Polgenix Inc., Cleveland, Ohio 44106.
| | - Beata Jastrzebska
- From the Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
45
|
Bulla M, Gyimesi G, Kim JH, Bhardwaj R, Hediger MA, Frieden M, Demaurex N. ORAI1 channel gating and selectivity is differentially altered by natural mutations in the first or third transmembrane domain. J Physiol 2018; 597:561-582. [PMID: 30382595 PMCID: PMC6332830 DOI: 10.1113/jp277079] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Gain-of-function mutations in the highly selective Ca2+ channel ORAI1 cause tubular aggregate myopathy (TAM) characterized by muscular pain, weakness and cramping. TAM-associated mutations in ORAI1 first and third transmembrane domain facilitate channel opening by STIM1, causing constitutive Ca2+ influx and increasing the currents evoked by Ca2+ store depletion. Mutation V107M additionally decreases the channel selectivity for Ca2+ ions and its inhibition by acidic pH, while mutation T184M does not alter the channel sensitivity to pH or to reactive oxygen species. The ORAI blocker GSK-7975A prevents the constitutive activity of TAM-associated channels and might be used in therapy for patients suffering from TAM. ABSTRACT Skeletal muscle differentiation relies on store-operated Ca2+ entry (SOCE) mediated by STIM proteins linking the depletion of endoplasmic/sarcoplasmic reticulum Ca2+ stores to the activation of membrane Ca2+ -permeable ORAI channels. Gain-of-function mutations in STIM1 or ORAI1 isoforms cause tubular aggregate myopathy (TAM), a skeletal muscle disorder with muscular pain, weakness and cramping. Here, we characterize two overactive ORAI1 mutants from patients with TAM: V107M and T184M, located in the first and third transmembrane domain of the channel. When ectopically expressed in HEK-293T cells or human primary myoblasts, the mutated channels increased basal and store-operated Ca2+ entry. The constitutive activity of V107M, L138F, T184M and P245L mutants was prevented by low concentrations of GSK-7975A while the G98S mutant was resistant to inhibition. Electrophysiological recordings confirmed ORAI1-V107M constitutive activity and revealed larger STIM1-gated V107M- and T184M-mediated currents with conserved fast and slow Ca2+ -dependent inactivation. Mutation V107M altered the channel selectivity for Ca2+ ions and conferred resistance to acidic inhibition. Ca2+ imaging and molecular dynamics simulations showed a preserved sensitivity of T184M to the negative regulation by reactive oxygen species. Both mutants were able to mediate SOCE in Stim1-/- /Stim2-/- mouse embryonic fibroblasts expressing the binding-deficient STIM1-F394H mutant, indicating a higher sensitivity for STIM1-mediated gating, with ORAI1-T184M gain-of-function being strictly dependent on STIM1. These findings provide new insights into the permeation and regulatory properties of ORAI1 mutants that might translate into therapies against diseases with gain-of-function mutations in ORAI1.
Collapse
Affiliation(s)
- M Bulla
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - G Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - J H Kim
- Departments of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - R Bhardwaj
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - M A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - M Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - N Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
Wang Z, Jumper JM, Wang S, Freed KF, Sosnick TR. A Membrane Burial Potential with H-Bonds and Applications to Curved Membranes and Fast Simulations. Biophys J 2018; 115:1872-1884. [PMID: 30413241 DOI: 10.1016/j.bpj.2018.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/21/2018] [Accepted: 10/10/2018] [Indexed: 10/28/2022] Open
Abstract
We use the statistics of a large and curated training set of transmembrane helical proteins to develop a knowledge-based potential that accounts for the dependence on both the depth of burial of the protein in the membrane and the degree of side-chain exposure. Additionally, the statistical potential includes depth-dependent energies for unsatisfied backbone hydrogen bond donors and acceptors, which are found to be relatively small, ∼2 RT. Our potential accurately places known proteins within the bilayer. The potential is applied to the mechanosensing MscL channel in membranes of varying thickness and curvature, as well as to the prediction of protein structure. The potential is incorporated into our new Upside molecular dynamics algorithm. Notably, we account for the exchange of protein-lipid interactions for protein-protein interactions as helices contact each other, thereby avoiding overestimating the energetics of helix association within the membrane. Simulations of most multimeric complexes find that isolated monomers and the oligomers retain the same orientation in the membrane, suggesting that the assembly of prepositioned monomers presents a viable mechanism of oligomerization.
Collapse
Affiliation(s)
- Zongan Wang
- Department of Chemistry, The University of Chicago, Chicago, Illinois; James Franck Institute, The University of Chicago, Chicago, Illinois
| | - John M Jumper
- Department of Chemistry, The University of Chicago, Chicago, Illinois; James Franck Institute, The University of Chicago, Chicago, Illinois; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - Sheng Wang
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia; Toyota Technological Institute at Chicago, Chicago, Illinois
| | - Karl F Freed
- Department of Chemistry, The University of Chicago, Chicago, Illinois; James Franck Institute, The University of Chicago, Chicago, Illinois.
| | - Tobin R Sosnick
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois; Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
47
|
Mackie DI, Al Mutairi F, Davis RB, Kechele DO, Nielsen NR, Snyder JC, Caron MG, Kliman HJ, Berg JS, Simms J, Poyner DR, Caron KM. h CALCRL mutation causes autosomal recessive nonimmune hydrops fetalis with lymphatic dysplasia. J Exp Med 2018; 215:2339-2353. [PMID: 30115739 PMCID: PMC6122977 DOI: 10.1084/jem.20180528] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/15/2018] [Accepted: 07/26/2018] [Indexed: 01/19/2023] Open
Abstract
We report the first case of nonimmune hydrops fetalis (NIHF) associated with a recessive, in-frame deletion of V205 in the G protein-coupled receptor, Calcitonin Receptor-Like Receptor (hCALCRL). Homozygosity results in fetal demise from hydrops fetalis, while heterozygosity in females is associated with spontaneous miscarriage and subfertility. Using molecular dynamic modeling and in vitro biochemical assays, we show that the hCLR(V205del) mutant results in misfolding of the first extracellular loop, reducing association with its requisite receptor chaperone, receptor activity modifying protein (RAMP), translocation to the plasma membrane and signaling. Using three independent genetic mouse models we establish that the adrenomedullin-CLR-RAMP2 axis is both necessary and sufficient for driving lymphatic vascular proliferation. Genetic ablation of either lymphatic endothelial Calcrl or nonendothelial Ramp2 leads to severe NIHF with embryonic demise and placental pathologies, similar to that observed in humans. Our results highlight a novel candidate gene for human congenital NIHF and provide structure-function insights of this signaling axis for human physiology.
Collapse
Affiliation(s)
- Duncan I Mackie
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Fuad Al Mutairi
- Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Centre (KAIMRC), Riyadh, Saudi Arabia
| | - Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Daniel O Kechele
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Joshua C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, NC
- Department of Surgery, Duke University Medical Center, Durham, NC
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC
| | - Harvey J Kliman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - John Simms
- School of Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, England, UK
| | - David R Poyner
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, England, UK
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
48
|
Simms J, Uddin R, Sakmar TP, Gingell JJ, Garelja ML, Hay DL, Brimble MA, Harris PW, Reynolds CA, Poyner DR. Photoaffinity Cross-Linking and Unnatural Amino Acid Mutagenesis Reveal Insights into Calcitonin Gene-Related Peptide Binding to the Calcitonin Receptor-like Receptor/Receptor Activity-Modifying Protein 1 (CLR/RAMP1) Complex. Biochemistry 2018; 57:4915-4922. [PMID: 30004692 DOI: 10.1021/acs.biochem.8b00502] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Calcitonin gene-related peptide (CGRP) binds to the complex of the calcitonin receptor-like receptor (CLR) with receptor activity-modifying protein 1 (RAMP1). How CGRP interacts with the transmembrane domain (including the extracellular loops) of this family B receptor remains unclear. In this study, a photoaffinity cross-linker, p-azido l-phenylalanine (azF), was incorporated into CLR, chiefly in the second extracellular loop (ECL2) using genetic code expansion and unnatural amino acid mutagenesis. The method was optimized to ensure efficient photolysis of azF residues near the transmembrane bundle of the receptor. A CGRP analogue modified with fluorescein at position 15 was used for detection of ultraviolet-induced cross-linking. The methodology was verified by confirming the known contacts of CGRP to the extracellular domain of CLR. Within ECL2, the chief contacts were I284 on the loop itself and L291, at the top of the fifth transmembrane helix (TM5). Minor contacts were noted along the lip of ECL2 between S286 and L290 and also with M223 in TM3 and F349 in TM6. Full length molecular models of the bound receptor complex suggest that CGRP sits at the top of the TM bundle, with Thr6 of the peptide making contacts with L291 and H295. I284 is likely to contact Leu12 and Ala13 of CGRP, and Leu16 of CGRP is at the ECL/extracellular domain boundary of CLR. The reduced potency, Emax, and affinity of [Leu16Ala]-human α CGRP are consistent with this model. Contacts between Thr6 of CGRP and H295 may be particularly important for receptor activation.
Collapse
Affiliation(s)
- John Simms
- Aston University , Birmingham B4 7ET , U.K
- Coventry University , Priory Street , Coventry CV1 5FB , U.K
| | | | - Thomas P Sakmar
- The Rockefeller University , 1230 York Avenue , New York , New York 10065 , United States
| | - Joseph J Gingell
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Michael L Garelja
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Debbie L Hay
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Margaret A Brimble
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Paul W Harris
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | | | | |
Collapse
|
49
|
Kharade SV, Kurata H, Bender AM, Blobaum AL, Figueroa EE, Duran A, Kramer M, Days E, Vinson P, Flores D, Satlin LM, Meiler J, Weaver CD, Lindsley CW, Hopkins CR, Denton JS. Discovery, Characterization, and Effects on Renal Fluid and Electrolyte Excretion of the Kir4.1 Potassium Channel Pore Blocker, VU0134992. Mol Pharmacol 2018; 94:926-937. [PMID: 29895592 PMCID: PMC6041953 DOI: 10.1124/mol.118.112359] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/30/2018] [Indexed: 12/28/2022] Open
Abstract
The inward rectifier potassium (Kir) channel Kir4.1 (KCNJ10) carries out important physiologic roles in epithelial cells of the kidney, astrocytes in the central nervous system, and stria vascularis of the inner ear. Loss-of-function mutations in KCNJ10 lead to EAST/SeSAME syndrome, which is characterized by epilepsy, ataxia, renal salt wasting, and sensorineural deafness. Although genetic approaches have been indispensable for establishing the importance of Kir4.1 in the normal function of these tissues, the availability of pharmacological tools for acutely manipulating the activity of Kir4.1 in genetically normal animals has been lacking. We therefore carried out a high-throughput screen of 76,575 compounds from the Vanderbilt Institute of Chemical Biology library for small-molecule modulators of Kir4.1. The most potent inhibitor identified was 2-(2-bromo-4-isopropylphenoxy)-N-(2,2,6,6-tetramethylpiperidin-4-yl)acetamide (VU0134992). In whole-cell patch-clamp electrophysiology experiments, VU0134992 inhibits Kir4.1 with an IC50 value of 0.97 µM and is 9-fold selective for homomeric Kir4.1 over Kir4.1/5.1 concatemeric channels (IC50 = 9 µM) at -120 mV. In thallium (Tl+) flux assays, VU0134992 is greater than 30-fold selective for Kir4.1 over Kir1.1, Kir2.1, and Kir2.2; is weakly active toward Kir2.3, Kir6.2/SUR1, and Kir7.1; and is equally active toward Kir3.1/3.2, Kir3.1/3.4, and Kir4.2. This potency and selectivity profile is superior to Kir4.1 inhibitors amitriptyline, nortriptyline, and fluoxetine. Medicinal chemistry identified components of VU0134992 that are critical for inhibiting Kir4.1. Patch-clamp electrophysiology, molecular modeling, and site-directed mutagenesis identified pore-lining glutamate 158 and isoleucine 159 as critical residues for block of the channel. VU0134992 displayed a large free unbound fraction (fu) in rat plasma (fu = 0.213). Consistent with the known role of Kir4.1 in renal function, oral dosing of VU0134992 led to a dose-dependent diuresis, natriuresis, and kaliuresis in rats. Thus, VU0134992 represents the first in vivo active tool compound for probing the therapeutic potential of Kir4.1 as a novel diuretic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Sujay V Kharade
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Haruto Kurata
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Aaron M Bender
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Anna L Blobaum
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Eric E Figueroa
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Amanda Duran
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Meghan Kramer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Emily Days
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Paige Vinson
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Daniel Flores
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Lisa M Satlin
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Jens Meiler
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - C David Weaver
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Craig W Lindsley
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Corey R Hopkins
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| |
Collapse
|
50
|
Afonin AV, Sterkhova IV, Vashchenko AV, Sigalov MV. Estimating the energy of intramolecular bifurcated (three-centered) hydrogen bond by X-ray, IR and 1 H NMR spectroscopy, and QTAIM calculations. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2018.02.106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|