1
|
Prokaeva T, Jayaraman S, Klimtchuk E, Burke N, Spencer B, Nedelkov D, Chen H, Dasari S, McPhail ED, Pereira L, Payne MC, Wong S, Burks EJ, Sanchorawala V, Gursky O. An unusual phenotype of hereditary AApoAI amyloidosis caused by a novel Asp20Tyr substitution is linked to pH-dependent aggregation of apolipoprotein A-I. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167820. [PMID: 40164396 PMCID: PMC11998993 DOI: 10.1016/j.bbadis.2025.167820] [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: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Apolipoprotein A-I (apoA-I) plays beneficial roles as the major structural and functional protein on plasma high-density lipoproteins (HDL). However, APOA1 gene mutations can cause protein misfolding and pathologic amyloid deposition in various organs in human hereditary AApoAI amyloidosis, a potentially lethal systemic disease. We report esophageal and duodenal AApoAI amyloidosis in a 56-year-old patient with Barrett's esophagus, a condition involving chronic acid reflux. Amyloid deposits contained full-length apoA-I featuring a novel D20Y mutation identified by gene sequencing and protein mass spectrometry. Genetic analysis of asymptomatic family members revealed autosomal dominant inheritance. Fibril formation by the full-length variant apoA-I rather than its fragments and the location of the mutation in a conserved amyloid-prone N-terminal segment were highly unusual for hereditary AApoA-I amyloidosis. Structural and stability studies of the recombinant D20Y and wild-type apoA-I showed small but significant mutation-induced structural perturbations in the native lipid-free protein at pH 7.4. Major destabilization and aggregation of the variant protein were observed at pH 4.0. We propose that acidic conditions in Barrett's esophagus promoted protein misfolding and amyloid formation by the D20Y variant. These findings expand our understanding of the clinical features and molecular basis of AApoAI amyloidosis and suggest clinical strategies.
Collapse
Affiliation(s)
- Tatiana Prokaeva
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA.
| | - Shobini Jayaraman
- Department of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Elena Klimtchuk
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Natasha Burke
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Brian Spencer
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | | | - Hui Chen
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Ellen D McPhail
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lucas Pereira
- Department of Hematology & Medical Oncology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Michael C Payne
- Division of Gastroenterology, Department of Internal Medicine, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Sherry Wong
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Eric J Burks
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Vaishali Sanchorawala
- Amyloidosis Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Olga Gursky
- Department of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA.
| |
Collapse
|
2
|
Bonnin-Marquez A, Jankowski J, Maas SL, Hermann J, Kahles F, Lellig M, Fliser D, Schunk S, Stamellou E, Berger M, Speer T, Kalim S, Leong Wong DW, van der Vorst EPC, Jankowski V. Guanidinylation compromises the anti-inflammatory and anti-oxidative properties of apolipoprotein A-I in chronic kidney disease progression. Kidney Int 2025; 107:916-929. [PMID: 40010491 DOI: 10.1016/j.kint.2025.02.010] [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: 08/08/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Chronic kidney disease (CKD) substantially heightens the likelihood of cardiovascular events, in part due to the impaired functionality of high-density lipoprotein (HDL) and its connection with atherosclerosis. Here, 82 patients with CKD stages 2-5 had their plasma isolated and analyzed using mass spectrometry to detect post-translational modifications of apolipoprotein A-I (apoA-I), the main protein component of HDL. Guanidinylation, a non-enzymatic post-translational modification, led to increased levels of apoA-I with CKD progression. The increase in guanidinylated apoA-I became significant from CKD stage 3 onwards. The modification patterns of apoA-I in patients with CKD were mimicked in vitro by exposure to O-methylisourea bisulfate. The thus modified apoA-I was used for functional assays which revealed that guanidinylation compromised the anti-inflammatory and anti-oxidative properties of apoA-I, of potential relevance for clinical findings. Specifically, guanidinylated apoA-I activated inflammatory kinases in macrophages, suggesting a mechanistic link between apoA-I modifications and inflammatory responses. These findings are in favor of alterations in the functional properties of apoA-I in patients with CKD due to guanidinylation. The identification of high guanidinylated apoA-I peptide levels in plasma highlights a novel aspect of protein modification in CKD pathophysiology. The results of our study may provide a better understanding of the molecular mechanisms underlying CKD-related cardiovascular complications and highlight the importance and the need to minimize post-translational modifications in patients with CKD.
Collapse
Affiliation(s)
- Andrea Bonnin-Marquez
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, the Netherlands
| | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
| | - Juliane Hermann
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Florian Kahles
- Department of Cardiology, Angiology and Intensive Care Medicine, University Hospital Aachen, Aachen, Germany
| | - Michaela Lellig
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Stefan Schunk
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Eleni Stamellou
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Martin Berger
- Department of Cardiology, Angiology and Intensive Care Medicine, University Hospital Aachen, Aachen, Germany
| | - Thimoteus Speer
- Department of Internal Medicine 4, Nephrology, Goethe University, Frankfurt/Main, Germany; Else Kroener Fresenius Center for Nephrological Research, Goethe University, Frankfurt/Main, Germany
| | - Sahir Kalim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | | | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
| |
Collapse
|
3
|
Sarker H, Panigrahi R, Lopez-Campistrous A, McMullen T, Reyes K, Anderson E, Krishnan V, Hernandez-Anzaldo S, Zheng XL, Glover JNM, Hardy E, Fernandez-Patron C. Apolipoprotein-A1 transports and regulates MMP2 in the blood. Nat Commun 2025; 16:3752. [PMID: 40263360 PMCID: PMC12015353 DOI: 10.1038/s41467-025-59062-0] [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: 01/25/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
Synthesized in the liver and intestines, apolipoprotein A1 (APOA1) transports cholesterol in high density lipoproteins from atherosclerotic lesions to the liver, protecting against atherosclerotic plaque rupture. Here, we show that proMMP2 (zymogen of matrix metalloproteinase-2) circulates associated with APOA1 in humans and APOA1-expressing mice. This is noteworthy because MMP2 is the most abundant MMP in blood, and MMPs promote atherosclerotic plaque rupture. Artificial intelligence (AlphaFold)-based modeling suggested that APOA1 and MMP2 interact; direct interactions were confirmed using five orthogonal interaction assays, showing that APOA1 binds to MMP2 catalytic and hemopexin-like domains. APOA1 inhibited MMP2 autolysis and allosterically increased MMP2 activity-an effect specifically reproduced by plasma from humans and APOA1-expressing mice but not albumin nor plasma from APOA1 knockout mice. These function-altering interactions with APOA1 may increase MMP2 bioavailability and lay the foundation for future research on how apolipoproteins and MMPs influence atherosclerotic plaque rupture, independently of cholesterol transport.
Collapse
Affiliation(s)
- Hassan Sarker
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rashmi Panigrahi
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ana Lopez-Campistrous
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Todd McMullen
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ken Reyes
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Elena Anderson
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vidhya Krishnan
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Samuel Hernandez-Anzaldo
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Inorganic Chemistry, Institute of Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Xi-Long Zheng
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - J N Mark Glover
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Eugenio Hardy
- Center of Molecular Immunology, P.O. Box 16040, Havana, Cuba
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- Cardiovascular Research Centre, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Kim S, Park JG, Choi SH, Kim JW, Jin MS. Cryo-EM structures reveal the H +/citrate symport mechanism of Drosophila INDY. Life Sci Alliance 2025; 8:e202402992. [PMID: 39884835 PMCID: PMC11782487 DOI: 10.26508/lsa.202402992] [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: 08/12/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Drosophila I'm Not Dead Yet (INDY) functions as a transporter for citrate, a key metabolite in the citric acid cycle, across the plasma membrane. Partial deficiency of INDY extends lifespan, akin to the effects of caloric restriction. In this work, we use cryo-electron microscopy to determine structures of INDY in the presence and absence of citrate and in complex with the well-known inhibitor 4,4'-diisothiocyano-2,2'-disulfonic acid stilbene (DIDS) at resolutions ranging from 2.7 to 3.6 Å. Together with functional data obtained in vitro, the INDY structures reveal the H+/citrate co-transport mechanism, in which aromatic residue F119 serves as a one-gate element. They also provide insight into how protein-lipid interactions at the dimerization interface affect the stability and function of the transporter, and how DIDS disrupts the transport cycle.
Collapse
Affiliation(s)
- Subin Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jun Gyou Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Seung Hun Choi
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Ji Won Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Mi Sun Jin
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
5
|
Koh YH, Kim SJ, Roh SH. Unraveling membrane protein localization and interactions in nanodiscs. FEBS Lett 2025; 599:512-529. [PMID: 39607859 DOI: 10.1002/1873-3468.15059] [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: 07/24/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024]
Abstract
Nanodiscs, consisting of a lipid bilayer surrounded by membrane scaffold proteins (MSPs), are extensively used to study membrane proteins (MPs) because they provide a stable lipid environment. However, the precise mechanism governing MP behavior within the nanodisc remains elusive. Here, we examined the cryo-EM structures of various MPs reconstituted in nanodiscs from EMPIAR. By analyzing the heterogeneity and interactions in the nanodiscs, we discovered that MPs display a distinct spatial preference toward the edges of the nanodisc shells. Furthermore, MPs can establish direct, amphipathic interactions with the MSPs, causing a reduction in local protein dynamics. These interactions may rearrange MSP-MSP interactions into MP-MSP interactions. Collectively, we provide structural insights into how nanodiscs contribute to MP structural behavior and dynamics. Impact statement Nanodiscs are used to study membrane proteins (MPs), but the mechanisms governing the behavior of MPs within nanodiscs remain elusive. Here, we provide structural insights into how nanodiscs contribute to the behavior of MPs, which will aid the interpretation of cryo-EM studies performed using nanodiscs.
Collapse
Affiliation(s)
- Young Hoon Koh
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, South Korea
| | - So-Jung Kim
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, South Korea
| | - Soung-Hun Roh
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, South Korea
| |
Collapse
|
6
|
Uddin M, Coombe D. A Coarse-Grained Simulation Approach for Protein Molecular Conformation Dynamics. J Phys Chem A 2025; 129:607-622. [PMID: 39754120 DOI: 10.1021/acs.jpca.4c06977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Coarse-grained molecular dynamics simulation is widely accepted for assessment of a large complex biological system, but it may also lead to a misleading conclusion. The challenge is to simulate protein structural dynamics (such as folding-unfolding behavior) due to the lack of a necessary backbone flexibility. This study developed a standard coarse-grained model directly from the protein atomic structure and amino acid coarse-grained FF (such as MARTINI FF v2.2). The atomic structure is used as a parent template to set up the coarse model, which naturally gives a better representation of the initial conditions. We have formulated a computational algorithm to set up protein coarse-grained coordinates and force field topology (such as bonds, angles, and dihedrals). The model was validated by a systematic all atom and coarse-grained simulation of a system containing protein human serum albumin and the drug paclitaxel in a water bath. The bonded force constants were optimized locally by neighboring residue-free energy data and globally by history matching against all atom simulation. The coarse-grained model was then applied for several other proteins and justified its general reliability for modeling protein conformations dynamics. We arrived at such a conclusion with great satisfaction because it describes the initial conditions accurately, applies only standard bonded force constants, and provides a significant backbone flexibility.
Collapse
Affiliation(s)
- Mafiz Uddin
- Alberta Computational Biochemistry Lab, 208, 8909-100 Street, Edmonton, Alberta T6E 6T4, Canada
| | - Dennis Coombe
- Computer Modelling Group, 3710 33 St NW, Calgary, Alberta T2L 2M1, Canada
| |
Collapse
|
7
|
Wang B, Tieleman DP. The structure, self-assembly and dynamics of lipid nanodiscs revealed by computational approaches. Biophys Chem 2024; 309:107231. [PMID: 38569455 DOI: 10.1016/j.bpc.2024.107231] [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: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Nanodisc technology is increasingly being used in structural, biochemical and biophysical studies of membrane proteins. The computational approaches have revealed many important features of nanodisc assembly, structures and dynamics. Therefore, we reviewed the application of computational approaches, especially molecular modeling and molecular dyncamics (MD) simulations, to characterize nanodiscs, including the structural models, assembly and disassembly, protocols for modeling, structural properties and dynamics, and protein-lipid interactions in nanodiscs. More amazing computational studies about nanodiscs are looked forward to in the future.
Collapse
Affiliation(s)
- Beibei Wang
- Centre for Advanced Materials Research, Beijing Normal University, Zhuhai 519087, China.
| | - D Peter Tieleman
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary T2N 1N4, Canada.
| |
Collapse
|
8
|
Malajczuk CJ, Mancera RL. An atomistic characterization of high-density lipoproteins and the conserved "LN" region of apoA-I. Biophys J 2024; 123:1116-1128. [PMID: 38555508 PMCID: PMC11079945 DOI: 10.1016/j.bpj.2024.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
The physicochemical characteristics of the various subpopulations of high-density lipoproteins (HDLs) and, in particular, their surface properties determine their ability to scavenge lipids and interact with specific receptors and peptides. Five representative spheroidal HDL subpopulation models were mapped from a previously reported equilibrated coarse-grained (CG) description to an atomistic representation for subsequent molecular dynamics simulation. For each HDL model a range of finer-level analyses was undertaken, including the component-wise characterization of HDL surfaces, the average size and composition of hydrophobic surface patches, dynamic protein secondary structure monitoring, and the proclivity for solvent exposure of the proposed β-amyloid (Aβ) binding region of apolipoprotein A-I (apoA-I), "LN." This study reveals that previously characterized ellipsoidal HDL3a and HDL2a models revert to a more spherical geometry in an atomistic representation due to the enhanced conformational flexibility afforded to the apoA-I protein secondary structure, allowing for enhanced surface lipid packing and lower overall surface hydrophobicity. Indeed, the proportional surface hydrophobicity and apoA-I exposure reduced with increasing HDL size, consistent with previous characterizations. Furthermore, solvent exposure of the "LN" region of apoA-I was exclusively limited to the smallest HDL3c model within the timescale of the simulations, and typically corresponded to a distinct loss in secondary structure across the "LN" region to form part of a significant contiguous hydrophobic patch on the HDL surface. Taken together, these findings provide preliminary evidence for a subpopulation-specific interaction between HDL3c particles and circulating hydrophobic species such as Aβ via the exposed "LN" region of apoA-I.
Collapse
Affiliation(s)
- Chris J Malajczuk
- Curtin Medical School, Curtin Health Innovation Research Institute and Curtin Institute for Data Science, Curtin University, Perth, WA, Australia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute and Curtin Institute for Data Science, Curtin University, Perth, WA, Australia.
| |
Collapse
|
9
|
Nady A, Reichheld SE, Sharpe S. Structural studies of a serum amyloid A octamer that is primed to scaffold lipid nanodiscs. Protein Sci 2024; 33:e4983. [PMID: 38659173 PMCID: PMC11043621 DOI: 10.1002/pro.4983] [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: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
Serum amyloid A (SAA) is a highly conserved acute-phase protein that plays roles in activating multiple pro-inflammatory pathways during the acute inflammatory response and is commonly used as a biomarker of inflammation. It has been linked to beneficial roles in tissue repair through improved clearance of lipids and cholesterol from sites of damage. In patients with chronic inflammatory diseases, elevated levels of SAA may contribute to increased severity of the underlying condition. The majority of circulating SAA is bound to lipoproteins, primarily high-density lipoprotein (HDL). Interaction with HDL not only stabilizes SAA but also alters its functional properties, likely through altered accessibility of protein-protein interaction sites on SAA. While high-resolution structures for lipid-free, or apo-, forms of SAA have been reported, their relationship with the HDL-bound form of the protein, and with other possible mechanisms of SAA binding to lipids, has not been established. Here, we have used multiple biophysical techniques, including SAXS, TEM, SEC-MALS, native gel electrophoresis, glutaraldehyde crosslinking, and trypsin digestion to characterize the lipid-free and lipid-bound forms of SAA. The SAXS and TEM data show the presence of soluble octamers of SAA with structural similarity to the ring-like structures reported for lipid-free ApoA-I. These SAA octamers represent a previously uncharacterized structure for lipid-free SAA and are capable of scaffolding lipid nanodiscs with similar morphology to those formed by ApoA-I. The SAA-lipid nanodiscs contain four SAA molecules and have similar exterior dimensions as the lipid-free SAA octamer, suggesting that relatively few conformational rearrangements may be required to allow SAA interactions with lipid-containing particles such as HDL. This study suggests a new model for SAA-lipid interactions and provides new insight into how SAA might stabilize protein-lipid nanodiscs or even replace ApoA-I as a scaffold for HDL particles during inflammation.
Collapse
Affiliation(s)
- Asal Nady
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| | - Sean E. Reichheld
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
| | - Simon Sharpe
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| |
Collapse
|
10
|
Smith JW, Carnevale LN, Das A, Chen Q. Electron videography of a lipid-protein tango. SCIENCE ADVANCES 2024; 10:eadk0217. [PMID: 38630809 PMCID: PMC11023515 DOI: 10.1126/sciadv.adk0217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
Biological phenomena, from enzymatic catalysis to synaptic transmission, originate in the structural transformations of biomolecules and biomolecular assemblies in liquid water. However, directly imaging these nanoscopic dynamics without probes or labels has been a fundamental methodological challenge. Here, we developed an approach for "electron videography"-combining liquid phase electron microscopy with molecular modeling-with which we filmed the nanoscale structural fluctuations of individual, suspended, and unlabeled membrane protein nanodiscs in liquid. Systematic comparisons with biochemical data and simulation indicate the graphene encapsulation involved can afford sufficiently reduced effects of the illuminating electron beam for these observations to yield quantitative fingerprints of nanoscale lipid-protein interactions. Our results suggest that lipid-protein interactions delineate dynamically modified membrane domains across unexpectedly long ranges. Moreover, they contribute to the molecular mechanics of the nanodisc as a whole in a manner specific to the protein within. Overall, this work illustrates an experimental approach to film, quantify, and understand biomolecular dynamics at the nanometer scale.
Collapse
Affiliation(s)
- John W. Smith
- Department of Materials Science and Engineering, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Lauren N. Carnevale
- Department of Biochemistry, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Aditi Das
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Qian Chen
- Department of Materials Science and Engineering, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
- Materials Research Laboratory, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| |
Collapse
|
11
|
Strickland MR, Rau MJ, Summers B, Basore K, Wulf J, Jiang H, Chen Y, Ulrich JD, Randolph GJ, Zhang R, Fitzpatrick JAJ, Cashikar AG, Holtzman DM. Apolipoprotein E secreted by astrocytes forms antiparallel dimers in discoidal lipoproteins. Neuron 2024; 112:1100-1109.e5. [PMID: 38266643 PMCID: PMC10994765 DOI: 10.1016/j.neuron.2023.12.018] [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: 09/11/2023] [Revised: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
The Apolipoprotein E gene (APOE) is of great interest due to its role as a risk factor for late-onset Alzheimer's disease. ApoE is secreted by astrocytes in the central nervous system in high-density lipoprotein (HDL)-like lipoproteins. Structural models of lipidated ApoE of high resolution could aid in a mechanistic understanding of how ApoE functions in health and disease. Using monoclonal Fab and F(ab')2 fragments, we characterize the structure of lipidated ApoE on astrocyte-secreted lipoproteins. Our results provide support for the "double-belt" model of ApoE in nascent discoidal HDL-like lipoproteins, where two ApoE proteins wrap around the nanodisc in an antiparallel conformation. We further show that lipidated, recombinant ApoE accurately models astrocyte-secreted ApoE lipoproteins. Cryogenic electron microscopy of recombinant lipidated ApoE further supports ApoE adopting antiparallel dimers in nascent discoidal lipoproteins.
Collapse
Affiliation(s)
| | - Michael J Rau
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Brock Summers
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Katherine Basore
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - John Wulf
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Hong Jiang
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Yun Chen
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Anil G Cashikar
- Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Psychiatry, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Taylor Family institute for Innovative Psychiatric Research, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA.
| |
Collapse
|
12
|
Li Y, Xu W, Wang X, Lai R, Qiu X, Zeng Z, Wang Z, Wang J. Molecular Dynamics and In Vitro Studies Elucidating the Tunable Features of Reconfigurable Nanodiscs for Guiding the Optimal Design of Curcumin Formulation. Bioengineering (Basel) 2024; 11:245. [PMID: 38534519 DOI: 10.3390/bioengineering11030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
In this study, we advance our exploration of Apolipoprotein A-I (apoA-I) peptide analogs (APAs) for their application in nanodisc (ND) assembly, focusing on the dynamic conformational characteristics and the potential for drug delivery. We explore APA-ND interactions with an emphasis on curcumin encapsulation, utilizing molecular dynamic simulations and in vitro assessments to evaluate the efficacy of various APA-ND formulations as drug carriers. The methodological approach involved the generation of three unique apoA-I α-11/3 helical mimics, resulting in fifteen distinct APAs. Their structural integrity was rigorously assessed using ColabFold-AF2, with particular attention to pLDDT and pTM scores. Extensive molecular dynamics simulations, covering 1.7 μs across 17 ND systems, were conducted to investigate the influence of APA sequence variations on ND stability and interactions. This study reveals that the composition of APAs, notably the presence of Proline, Serine, and Tryptophan, significantly impacts ND stability and morphology. Oligomeric APAs, in particular, demonstrated superior stability and distinct interaction patterns compared to their monomeric counterparts. Additionally, hydrodynamic diameter measurements over eight weeks indicated sequence-dependent stability, highlighting the potential of specific APA configurations for sustained colloidal stability. In vitro study successfully encapsulated curcumin in [AA]3/DMPC ND formulations, revealing concentration-dependent stability and interaction dynamics. The findings underscore the remarkable capability of APA-NDs to maintain structural integrity and efficient drug encapsulation, positioning them as a promising platform for drug delivery. The study concludes by emphasizing the tunability and versatility of APA-NDs in drug formulation, potentially revolutionizing nanomedicine by enabling customized APA sequences and ND properties for targeted drug delivery.
Collapse
Affiliation(s)
- Yongxiao Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wanting Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ruizhi Lai
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Xiaohui Qiu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zekai Zeng
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
13
|
Banach M. Structural Outlier Detection and Zernike-Canterakis Moments for Molecular Surface Meshes-Fast Implementation in Python. Molecules 2023; 29:52. [PMID: 38202635 PMCID: PMC10779519 DOI: 10.3390/molecules29010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Object retrieval systems measure the degree of similarity of the shape of 3D models. They search for the elements of the 3D model databases that resemble the query model. In structural bioinformatics, the query model is a protein tertiary/quaternary structure and the objective is to find similarly shaped molecules in the Protein Data Bank. With the ever-growing size of the PDB, a direct atomic coordinate comparison with all its members is impractical. To overcome this problem, the shape of the molecules can be encoded by fixed-length feature vectors. The distance of a protein to the entire PDB can be measured in this low-dimensional domain in linear time. The state-of-the-art approaches utilize Zernike-Canterakis moments for the shape encoding and supply the retrieval process with geometric data of the input structures. The BioZernike descriptors are a standard utility of the PDB since 2020. However, when trying to calculate the ZC moments locally, the issue of the deficiency of libraries readily available for use in custom programs (i.e., without relying on external binaries) is encountered, in particular programs written in Python. Here, a fast and well-documented Python implementation of the Pozo-Koehl algorithm is presented. In contrast to the more popular algorithm by Novotni and Klein, which is based on the voxelized volume, the PK algorithm produces ZC moments directly from the triangular surface meshes of 3D models. In particular, it can accept the molecular surfaces of proteins as its input. In the presented PK-Zernike library, owing to Numba's just-in-time compilation, a mesh with 50,000 facets is processed by a single thread in a second at the moment order 20. Since this is the first time the PK algorithm is used in structural bioinformatics, it is employed in a novel, simple, but efficient protein structure retrieval pipeline. The elimination of the outlying chain fragments via a fast PCA-based subroutine improves the discrimination ability, allowing for this pipeline to achieve an 0.961 area under the ROC curve in the BioZernike validation suite (0.997 for the assemblies). The correlation between the results of the proposed approach and of the 3D Surfer program attains values up to 0.99.
Collapse
Affiliation(s)
- Mateusz Banach
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Kraków, Poland
| |
Collapse
|
14
|
Aly AEE, Caron NS, Black HF, Schmidt ME, Anderson C, Ko S, Baddeley HJE, Anderson L, Casal LL, Rahavi RSM, Martin DDO, Hayden MR. Delivery of mutant huntingtin-lowering antisense oligonucleotides to the brain by intranasally administered apolipoprotein A-I nanodisks. J Control Release 2023; 360:913-927. [PMID: 37468110 DOI: 10.1016/j.jconrel.2023.07.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
Lowering mutant huntingtin (mHTT) in the central nervous system (CNS) using antisense oligonucleotides (ASOs) is a promising approach currently being evaluated in clinical trials for Huntington disease (HD). However, the therapeutic potential of ASOs in HD patients is limited by their inability to cross the blood-brain barrier (BBB). In non-human primates, intrathecal infusion of ASOs results in limited brain distribution, with higher ASO concentrations in superficial regions and lower concentrations in deeper regions, such as the basal ganglia. To address the need for improved delivery of ASOs to the brain, we are evaluating the therapeutic potential of apolipoprotein A-I nanodisks (apoA-I NDs) as novel delivery vehicles for mHTT-lowering ASOs to the CNS after intranasal administration. Here, we have demonstrated the ability of apoA-I nanodisks to bypass the BBB after intranasal delivery in the BACHD model of HD. Following intranasal administration of apoA-I NDs, apoA-I protein levels were elevated along the rostral-caudal brain axis, with highest levels in the most rostral brain regions including the olfactory bulb and frontal cortex. Double-label immunohistochemistry indicates that both the apoA-I and ASO deposit in neurons. Most importantly, a single intranasal dose of apoA-I ASO-NDs significantly reduces mHTT levels in the brain regions most affected in HD, namely the cortex and striatum. This approach represents a novel non-invasive means for improving delivery and brain distribution of oligonucleotide therapies and enhancing likelihood of efficacy. Improved ASO delivery to the brain has widespread application for treatment of many other CNS disorders.
Collapse
Affiliation(s)
- Amirah E-E Aly
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Nicholas S Caron
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Hailey Findlay Black
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Mandi E Schmidt
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Christine Anderson
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC V5Z 4H4, Canada
| | - Seunghyun Ko
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC V5Z 4H4, Canada
| | - Helen J E Baddeley
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC V5Z 4H4, Canada
| | - Lisa Anderson
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC V5Z 4H4, Canada
| | - Lorenzo L Casal
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC V5Z 4H4, Canada
| | - Reza S M Rahavi
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children's a Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Dale D O Martin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; Department of Biology, University of Waterloo, Ontario, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
15
|
Nakagawa F, Kikkawa M, Chen S, Miyashita Y, Hamaguchi-Suzuki N, Shibuya M, Yamashita S, Nagase L, Yasuda S, Shiroishi M, Senda T, Ito K, Murata T, Ogasawara S. Anti-nanodisc antibodies specifically capture nanodiscs and facilitate molecular interaction kinetics studies for membrane protein. Sci Rep 2023; 13:11627. [PMID: 37468499 DOI: 10.1038/s41598-023-38547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Nanodisc technology has dramatically advanced the analysis of molecular interactions for membrane proteins. A nanodisc is designed as a vehicle for membrane proteins that provide a native-like phospholipid environment and better thermostability in a detergent-free buffer. This enables the determination of the thermodynamic and kinetic parameters of small molecule binding by surface plasmon resonance. In this study, we generated a nanodisc specific anti-MSP (membrane scaffold protein) monoclonal antibody biND5 for molecular interaction analysis of nanodiscs. The antibody, biND5 bound to various types of nanodiscs with sub-nanomolar to nanomolar affinity. Epitope mapping analysis revealed specific recognition of 8 amino acid residues in the exposed helix-4 structure of MSP. Further, we performed kinetics binding analysis between adenosine A2a receptor reconstituted nanodiscs and small molecule antagonist ZM241385 using biND5 immobilized sensor chips. These results show that biND5 facilitates the molecular interaction kinetics analysis of membrane proteins substituted in nanodiscs.
Collapse
Affiliation(s)
- Fuhito Nakagawa
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Marin Kikkawa
- Department of Food and Nutritional Sciences, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan
| | - Sisi Chen
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
- Membrane Protein Research Center, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Yasuomi Miyashita
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Norie Hamaguchi-Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Minami Shibuya
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Soichi Yamashita
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Lisa Nagase
- Structure Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki, 305-0801, Japan
| | - Satoshi Yasuda
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
- Membrane Protein Research Center, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
- Department of Quantum Life Science, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan
| | - Mitsunori Shiroishi
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-Ku, Tokyo, 125-8585, Japan
| | - Toshiya Senda
- Structure Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki, 305-0801, Japan
- Department of Materials Structure Science, School of High Energy Accelerator Science, The Graduate University of Advanced Studies (Soken-Dai), 1-1 Oho, Tsukuba, Ibaraki, 305-0801, Japan
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Keisuke Ito
- Department of Food and Nutritional Sciences, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
- Membrane Protein Research Center, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
- Structure Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki, 305-0801, Japan.
- Department of Quantum Life Science, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
| | - Satoshi Ogasawara
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
- Membrane Protein Research Center, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
- Department of Quantum Life Science, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
- Institute for Advanced Academic Research, Chiba University, 1-33 Yayoi-Cho, Inage, Chiba, 263-8522, Japan.
| |
Collapse
|
16
|
Xu D, Chen X, Li Y, Chen Z, Xu W, Wang X, Lv Y, Wang Z, Wu M, Liu G, Wang J. Reconfigurable Peptide Analogs of Apolipoprotein A-I Reveal Tunable Features of Nanodisc Assembly. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1262-1276. [PMID: 36626237 DOI: 10.1021/acs.langmuir.2c03082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nanodisc (ND)-forming membrane scaffold proteins or peptides developed from apolipoprotein A-I (apoA-I) have led to considerable promise in structural biology and therapeutic applications. However, the rationale and regularity characteristics in peptide sequence design remain inconclusive. Here, we proposed a consensus-based normalization approach through the reversed engineering of apoA-IΔ1-45 to design reconfigurable apoA-I peptide analogs (APAs) for tunable ND assembly. We present extensive morphological validations and computational simulation analyses on divergent APA-NDs that are generated by our method. Fifteen divergent APAs were generated accordingly to study the assembly machinery of NDs. We show that APA designs exhibit multifactorial influence in terms of varying APA tandem repeats, sequence composition, and lipid-to-APA ratio to form tunable diameters of NDs. There is a strong positive correlation between DMPC-to-APA ratios and ND diameters. Longer APA with more tandem repeats tends to yield higher particle size homogeneity. Our results also suggest proline is a dispensable residue for the APA-ND formation. Interestingly, proline-rich substitution not only provides an inward-bending effect in forming smaller NDs but also induces the cumulative chain flexibility that enables larger ND formation at higher lipid ratios. Additionally, proline-tryptophan residues in APAs play a dominant role in forming larger NDs. Molecular simulation shows that enriched basic and acidic residues in APAs evoke abundant hydrogen-bond and salt bridge networks to reinforce the structural stability of APA-NDs. Together, our findings provide a rational basis for understanding APA design. The proposed model could be extended to other apolipoproteins for desired ND engineering.
Collapse
Affiliation(s)
- Daiyun Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Yongxiao Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Zhidong Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Wanting Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Yonghui Lv
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen518033, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen361102, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| |
Collapse
|
17
|
Lin HJ, James I, Hyer CD, Haderlie CT, Zackrison MJ, Bateman TM, Berg M, Park JS, Daley SA, Zuniga Pina NR, Tseng YJJ, Moody JD, Price JC. Quantifying In Situ Structural Stabilities of Human Blood Plasma Proteins Using a Novel Iodination Protein Stability Assay. J Proteome Res 2022; 21:2920-2935. [PMID: 36356215 PMCID: PMC9724711 DOI: 10.1021/acs.jproteome.2c00323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Indexed: 11/12/2022]
Abstract
Many of the diseases that plague society today are driven by a loss of protein quality. One method to quantify protein quality is to measure the protein folding stability (PFS). Here, we present a novel mass spectrometry (MS)-based approach for PFS measurement, iodination protein stability assay (IPSA). IPSA quantifies the PFS by tracking the surface-accessibility differences of tyrosine, histidine, methionine, and cysteine under denaturing conditions. Relative to current methods, IPSA increases protein coverage and granularity to track the PFS changes of a protein along its sequence. To our knowledge, this study is the first time the PFS of human serum proteins has been measured in the context of the blood serum (in situ). We show that IPSA can quantify the PFS differences between different transferrin iron-binding states in near in vivo conditions. We also show that the direction of the denaturation curve reflects the in vivo surface accessibility of the amino acid residue and reproducibly reports a residue-specific PFS. Along with IPSA, we introduce an analysis tool Chalf that provides a simple workflow to calculate the residue-specific PFS. The introduction of IPSA increases the potential to use protein structural stability as a structural quality metric in understanding the etiology and progression of human disease. Data is openly available at Chorusproject.org (project ID 1771).
Collapse
Affiliation(s)
- Hsien-Jung
L. Lin
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Isabella James
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Chad D. Hyer
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Connor T. Haderlie
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Michael J. Zackrison
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Tyler M. Bateman
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Monica Berg
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Ji-Sun Park
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - S. Anisha Daley
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Nathan R. Zuniga Pina
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - Yi-Jie J. Tseng
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - James D. Moody
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| | - John C. Price
- Department of Chemistry and
Biochemistry, Brigham Young University, Provo, Utah84602, United States
| |
Collapse
|
18
|
Rijal H, Goggin L, Muriph R, Evans J, Hamad-Schifferli K. The Influence of Preforming Protein Coronas on the Performance of Dengue NS1 Immunoassays. Pharmaceutics 2022; 14:2439. [PMID: 36432630 PMCID: PMC9694804 DOI: 10.3390/pharmaceutics14112439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
The effect of preformed protein coronas on immunoassays for Dengue nonstructural protein 1 (NS1) immunoassays was investigated. The composition of the protein corona that forms around nanoparticle-antibody conjugates in human serum was characterized, and selected proteins from the corona were used for preformed coronas (human serum albumin and apolipoprotein A1). Coronas were formed and characterized by dynamic light scattering (DLS), and the nanoparticle-conjugate was probed by optical absorption spectroscopy. Immunoassays were run, and performance was quantified by analyzing the strip intensity as a function of NS1 concentration. The preformed coronas influenced the limit of detection (LOD) of the assay and the affinity for the NS1 target (KD). The resulting KD and LODs for the NP-Ab-ApoA1 immunoprobes were 0.83 nM and 1.24 nM, respectively. For the NP-Ab -HSA coronas, the test line intensity was lower by 33% at a given NS1 concentration than for the NP-Ab immunoprobes, and KD was 0.14 nM, a slightly higher affinity. Due to the relatively large error of the negative control, a meaningful LOD for the NP-Ab with HSA coronas could not be determined.
Collapse
Affiliation(s)
- Hom Rijal
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Laura Goggin
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Rachel Muriph
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Jason Evans
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Kimberly Hamad-Schifferli
- Department of Engineering, University of Massachusetts Boston, Boston, MA 02125, USA
- School for the Environment, University of Massachusetts Boston, Boston, MA 02125, USA
| |
Collapse
|
19
|
Bhale AS, Venkataraman K. Leveraging knowledge of HDLs major protein ApoA1: Structure, function, mutations, and potential therapeutics. Biomed Pharmacother 2022; 154:113634. [PMID: 36063649 DOI: 10.1016/j.biopha.2022.113634] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is a member of the Apolipoprotein family of proteins. It's a vital protein that helps in the production of high-density lipoprotein (HDL) particles, which are crucial for reverse cholesterol transport (RCT). It also has anti-inflammatory, anti-atherogenic, anti-apoptotic, and anti-thrombotic properties. These functions interact to give HDL particles their cardioprotective characteristics. ApoA1 has recently been investigated for its potential role in atherosclerosis, diabetes, neurological diseases, cancer, and certain infectious diseases. Since ApoA1's discovery, numerous mutations have been reported that affect its structural integrity and alter its function. Hence these insights have led to the development of clinically relevant peptides and synthetic reconstituted HDL (rHDL) that mimics the function of ApoA1. As a result, this review has aimed to provide an organized explanation of our understanding of the ApoA1 protein structure and its role in various essential pathways. Furthermore, we have comprehensively reviewed the important ApoA1 mutations (24 mutations) that are reported to be involved in various diseases. Finally, we've focused on the therapeutic potentials of some of the beneficial mutations, small peptides, and synthetic rHDL that are currently being researched or developed, since these will aid in the development of novel therapeutics in the future.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
20
|
Bedi S, Morris J, Shah A, Hart RC, Jerome WG, Aller SG, Tang C, Vaisar T, Bornfeldt KE, Segrest JP, Heinecke JW, Davidson WS. Conformational flexibility of apolipoprotein A-I amino- and carboxy-termini is necessary for lipid binding but not cholesterol efflux. J Lipid Res 2022; 63:100168. [PMID: 35051413 PMCID: PMC8953623 DOI: 10.1016/j.jlr.2022.100168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/03/2022] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
Because of its critical role in HDL formation, significant efforts have been devoted to studying apolipoprotein A-I (APOA1) structural transitions in response to lipid binding. To assess the requirements for the conformational freedom of its termini during HDL particle formation, we generated three dimeric APOA1 molecules with their termini covalently joined in different combinations. The dimeric (d)-APOA1C-N mutant coupled the C-terminus of one APOA1 molecule to the N-terminus of a second with a short alanine linker, whereas the d-APOA1C-C and d-APOA1N-N mutants coupled the C-termini and the N-termini of two APOA1 molecules, respectively, using introduced cysteine residues to form disulfide linkages. We then tested the ability of these constructs to generate reconstituted HDL by detergent-assisted and spontaneous phospholipid microsolubilization methods. Using cholate dialysis, we demonstrate WT and all APOA1 mutants generated reconstituted HDL particles of similar sizes, morphologies, compositions, and abilities to activate lecithin:cholesterol acyltransferase. Unlike WT, however, the mutants were incapable of spontaneously solubilizing short chain phospholipids into discoidal particles. We found lipid-free d-APOA1C-N and d-APOA1N-N retained most of WT APOA1's ability to promote cholesterol efflux via the ATP binding cassette transporter A1, whereas d-APOA1C-C exhibited impaired cholesterol efflux. Our data support the double belt model for a lipid-bound APOA1 structure in nascent HDL particles and refute other postulated arrangements like the "double super helix." Furthermore, we conclude the conformational freedom of both the N- and C-termini of APOA1 is important in spontaneous microsolubilization of bulk phospholipid but is not critical for ABCA1-mediated cholesterol efflux.
Collapse
Affiliation(s)
- Shimpi Bedi
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jamie Morris
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Amy Shah
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rachel C Hart
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - W Gray Jerome
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Stephen G Aller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chongren Tang
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Tomas Vaisar
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jere P Segrest
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
21
|
Fusion pores with low conductance are cation selective. Cell Rep 2021; 36:109580. [PMID: 34433034 PMCID: PMC8500334 DOI: 10.1016/j.celrep.2021.109580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/01/2021] [Accepted: 07/30/2021] [Indexed: 11/23/2022] Open
Abstract
Many neurotransmitters are organic ions that carry a net charge, and their release from secretory vesicles is therefore an electrodiffusion process. The selectivity of early exocytotic fusion pores is investigated by combining electrodiffusion theory, measurements of amperometric foot signals from chromaffin cells with anion substitution, and molecular dynamics simulation. The results reveal that very narrow fusion pores are cation selective, but more dilated fusion pores become anion permeable. The transition occurs around a fusion pore conductance of ~300 pS. The cation selectivity of a narrow fusion pore accelerates the release of positively charged transmitters such as dopamine, noradrenaline, adrenaline, serotonin, and acetylcholine, while glutamate release may require a more dilated fusion pore. For transmission, a fusion pore forms when vesicle and target membranes are brought together by SNARE proteins. Delacruz et al. demonstrate that selectivity of the pore accelerates release of positively charged transmitters such as dopamine, noradrenaline, adrenaline, serotonin, and acetylcholine, while glutamate release may require a more dilated fusion pore.
Collapse
|
22
|
Melchior JT, Street SE, Vaisar T, Hart R, Jerome J, Kuklenyik Z, Clouet-Foraison N, Thornock C, Bedi S, Shah AS, Segrest JP, Heinecke JW, Davidson WS. Apolipoprotein A-I modulates HDL particle size in the absence of apolipoprotein A-II. J Lipid Res 2021; 62:100099. [PMID: 34324889 PMCID: PMC8385444 DOI: 10.1016/j.jlr.2021.100099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/01/2021] [Accepted: 07/15/2021] [Indexed: 11/30/2022] Open
Abstract
Human high-density lipoproteins (HDL) are a complex mixture of structurally-related nanoparticles that perform distinct physiological functions. We previously showed human HDL containing apolipoprotein A-I (APOA1) but not apolipoprotein A-II (APOA2), designated LpA-I, is composed primarily of two discretely sized populations. Here, we isolated these particles directly from human plasma by antibody affinity chromatography, separated them by high-resolution size exclusion chromatography and performed a deep molecular characterization of each species. The large and small LpA-I populations were spherical with mean diameters of 109 Å and 91 Å, respectively. Unexpectedly, isotope dilution MS/MS with [15N]-APOA1 in concert with quantitation of particle concentration by calibrated ion mobility analysis demonstrated that the large particles contained fewer APOA1 molecules than the small particles; the stoichiometries were 3.0 and 3.7 molecules of APOA1 per particle, respectively. MS/MS experiments showed that the protein cargo of large LpA-I particles was more diverse. Human HDL and isolated particles containing both APOA1 and APOA2 exhibit a much wider range and variation of particle sizes than LpA-I, indicating that APOA2 is likely the major contributor to HDL size heterogeneity. We propose a ratchet model based on the trefoil structure of APOA1 whereby the helical cage maintaining particle structure has two 'settings' - large and small - that accounts for these findings. This understanding of the determinants of HDL particle size and protein cargo distribution serves as a basis for determining the roles of HDL subpopulations in metabolism and disease states.
Collapse
Affiliation(s)
- John T Melchior
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio 45237; Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, 99354
| | - Scott E Street
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio 45237
| | - Tomas Vaisar
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109
| | - Rachel Hart
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Jay Jerome
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Zsuzsanna Kuklenyik
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341
| | - Noemie Clouet-Foraison
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109
| | - Carissa Thornock
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109
| | - Shimpi Bedi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, 99354
| | - Amy S Shah
- Division of Endocrinology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio 45229
| | - Jere P Segrest
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Jay W Heinecke
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109
| | - W Sean Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio 45237.
| |
Collapse
|
23
|
Schrijver DP, Dreu A, Hofstraat SRJ, Kluza E, Zwolsman R, Deckers J, Anbergen T, Bruin K, Trines MM, Nugraha EG, Ummels F, Röring RJ, Beldman TJ, Teunissen AJP, Fayad ZA, Meel R, Mulder WJM. Nanoengineering Apolipoprotein A1‐Based Immunotherapeutics. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- David P. Schrijver
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Anne Dreu
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Stijn R. J. Hofstraat
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Ewelina Kluza
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Robby Zwolsman
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Jeroen Deckers
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Tom Anbergen
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Koen Bruin
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Mirre M. Trines
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Eveline G. Nugraha
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Floor Ummels
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Rutger J. Röring
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| | - Abraham J. P. Teunissen
- Biomedical Engineering and Imaging Institute Icahn School of Medicine at Mount Sinai New York NY 10029‐6574 USA
| | - Zahi A. Fayad
- Biomedical Engineering and Imaging Institute Icahn School of Medicine at Mount Sinai New York NY 10029‐6574 USA
| | - Roy Meel
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Willem J. M. Mulder
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| |
Collapse
|
24
|
Kjølbye LR, De Maria L, Wassenaar TA, Abdizadeh H, Marrink SJ, Ferkinghoff-Borg J, Schiøtt B. General Protocol for Constructing Molecular Models of Nanodiscs. J Chem Inf Model 2021; 61:2869-2883. [PMID: 34048229 DOI: 10.1021/acs.jcim.1c00157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nanodisc technology is increasingly being applied for structural and biophysical studies of membrane proteins. In this work, we present a general protocol for constructing molecular models of nanodiscs for molecular dynamics simulations. The protocol is written in python and based on geometric equations, making it fast and easy to modify, enabling automation and customization of nanodiscs in silico. The novelty being the ability to construct any membrane scaffold protein (MSP) variant fast and easy given only an input sequence. We validated and tested the protocol by simulating seven different nanodiscs of various sizes and with different membrane scaffold proteins, both circularized and noncircularized. The structural and biophysical properties were analyzed and shown to be in good agreement with previously reported experimental data and simulation studies.
Collapse
Affiliation(s)
- Lisbeth R Kjølbye
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus C, Denmark.,Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | | | - Tsjerk A Wassenaar
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Haleh Abdizadeh
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | | | - Birgit Schiøtt
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds vej 14, 8000 Aarhus C, Denmark
| |
Collapse
|
25
|
Interaction of lipid-free apolipoprotein A-I with cholesterol revealed by molecular modeling. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140614. [PMID: 33548491 DOI: 10.1016/j.bbapap.2021.140614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/23/2021] [Accepted: 01/28/2021] [Indexed: 11/22/2022]
Abstract
We report the modeling of the interaction of differently self-associated lipid-free apoA-I with cholesterol monomer and tail-to-tail (TT) or face-to-face (FF) cholesterol dimer. Cholesterol dimerization is exploited to reconcile the existing experimental data on cholesterol binding to apoA-I with extremely low critical micelle concentration of cholesterol. Two crystal structures of 1-43 N-truncated apolipoprotein Δ(1-43)A-I tetramer (PDB ID: 1AV1, structure B), 185-243 C-truncated apolipoprotein Δ(185-243)A-I dimer (PDB ID: 3R2P, structure M) were analyzed. Cholesterol monomers bind to multiple binding sites in apoA-I monomer, dimer and tetramer with low, moderate and high energy (-10 to -28 kJ/mol with Schrödinger package), still insufficient to overcome the thermodynamic restriction by cholesterol micellization (-52.8 kJ/mol). The binding sites partially coincide with the putative cholesterol-binding motifs. However, apoA-I monomer and dimer existing in structure B, that contain nonoverlapping and non-interacting pairs of binding sites with high affinity for TT and FF cholesterol dimers, can bind in common 14 cholesterol molecules that correspond to existing values. ApoA-I monomer and dimer in structure M can bind in common 6 cholesterol molecules. The values of respective total energy of cholesterol binding up to 64.5 and 67.0 kJ/mol for both B and M structures exceed the free energy of cholesterol micellization. We hypothesize that cholesterol dimers may simultaneously interact with extracellular monomer and dimer of lipid-free apoA-I, that accumulate at acid pH in atheroma. The thermodynamically allowed apolipoprotein-cholesterol interaction outside the macrophage may represent a new mechanism of cholesterol transport by apoA-I from atheroma, in addition to ABCA1-mediated cholesterol efflux.
Collapse
|
26
|
Gkolfinopoulou C, Soukou F, Dafnis I, Kellici TF, Sanoudou D, Mavromoustakos T, Stratikos E, Chroni A. Structure-function analysis of naturally occurring apolipoprotein A-I L144R, A164S and L178P mutants provides insight on their role on HDL levels and cardiovascular risk. Cell Mol Life Sci 2021; 78:1523-1544. [PMID: 32666307 PMCID: PMC11072781 DOI: 10.1007/s00018-020-03583-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 01/04/2023]
Abstract
Naturally occurring point mutations in apolipoprotein A-I (apoA-I), the major protein component of high-density lipoprotein (HDL), may affect plasma HDL-cholesterol levels and cardiovascular risk. Here, we evaluated the effect of human apoA-I mutations L144R (associated with low HDL-cholesterol), L178P (associated with low HDL-cholesterol and increased cardiovascular risk) and A164S (associated with increased cardiovascular risk and mortality without low HDL-cholesterol) on the structural integrity and functions of lipid-free and lipoprotein-associated apoA-I in an effort to explain the phenotypes of subjects carrying these mutations. All three mutants, in lipid-free form, presented structural and thermodynamic aberrations, with apoA-I[L178P] presenting the greatest thermodynamic destabilization. Additionally, apoA-I[L178P] displayed reduced ABCA1-mediated cholesterol efflux capacity. When in reconstituted HDL (rHDL), apoA-I[L144R] and apoA-I[L178P] were more thermodynamically destabilized compared to wild-type apoA-I, both displayed reduced SR-BI-mediated cholesterol efflux capacity and apoA-I[L144R] showed severe LCAT activation defect. ApoA-I[A164S] was thermodynamically unaffected when in rHDL, but exhibited a series of functional defects. Specifically, it had reduced ABCG1-mediated cholesterol and 7-ketocholesterol efflux capacity, failed to reduce ROS formation in endothelial cells and had reduced capacity to induce endothelial cell migration. Mechanistically, the latter was due to decreased capacity of rHDL-apoA-I[A164S] to activate Akt kinase possibly by interacting with endothelial LOX-1 receptor. The impaired capacity of rHDL-apoA-I[A164S] to preserve endothelial function may be related to the increased cardiovascular risk for this mutation. Overall, our structure-function analysis of L144R, A164S and L178P apoA-I mutants provides insights on how HDL-cholesterol levels and/or atheroprotective properties of apoA-I/HDL are impaired in carriers of these mutations.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Apolipoprotein A-I/genetics
- Apolipoprotein A-I/metabolism
- Apolipoprotein A-I/ultrastructure
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/pathology
- Cell Movement/genetics
- Cholesterol, HDL/genetics
- Cholesterol, HDL/metabolism
- Cholesterol, HDL/ultrastructure
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Heart Disease Risk Factors
- Humans
- Ketocholesterols/genetics
- Ketocholesterols/metabolism
- Lipoproteins, HDL/genetics
- Lipoproteins, HDL/metabolism
- Lipoproteins, HDL/ultrastructure
- Mutant Proteins/genetics
- Mutant Proteins/metabolism
- Mutant Proteins/ultrastructure
- Mutation/genetics
- Scavenger Receptors, Class E/genetics
- Scavenger Receptors, Class E/metabolism
- Structure-Activity Relationship
- Thermodynamics
Collapse
Affiliation(s)
- Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, 15341, Athens, Greece
| | - Faye Soukou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, 15341, Athens, Greece
| | - Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, 15341, Athens, Greece
| | - Tahsin F Kellici
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, Athens, Greece
| | - Despina Sanoudou
- 4th Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, 'Attikon' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, Athens, Greece
| | - Efstratios Stratikos
- Protein Chemistry Laboratory, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, 15341, Athens, Greece.
| |
Collapse
|
27
|
Abstract
The properties of natural lipid bilayers are vital to the regulation of many membrane proteins. Scaffolded nanodiscs provide an in vitro lipid bilayer platform to host membrane proteins in an environment that approximates native lipid bilayers. However, the properties of scaffold-enclosed bilayers may depart significantly from those of bulk cellular membranes. Therefore, to improve the usefulness of nanodiscs it is essential to understand the properties of lipids restricted by scaffolds. We used computational molecular dynamics and modeling approaches to understand the effects of nanodisc size, scaffold type (DNA or protein), and hydrophobic modification of DNA scaffolds on bilayer stability and degree to which the properties of enclosed bilayers approximate bulk bilayers. With respect to achieving bulk bilayer behavior, we found that charge neutralization of DNA scaffolds was more important than the total hydrophobic content of their modifications: bilayer properties were better for scaffolds having a large number of short alkyl chains than those having fewer long alkyl chains. Further, complete charge neutralization of DNA scaffolds enabled better lipid binding, and more stable bilayers, as shown by steered molecular dynamics simulations that measured the force required to dislodge scaffolds from lipid bilayer patches. Considered together, our simulations provide a guide to the design of DNA-scaffolded nanodiscs suitable for studying membrane proteins.
Collapse
Affiliation(s)
- Vishal Maingi
- Department of Bioengineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Paul W K Rothemund
- Departments of Bioengineering, Computing + Mathematical Sciences, and Computation & Neural Systems, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
28
|
Laurenzi T, Parravicini C, Palazzolo L, Guerrini U, Gianazza E, Calabresi L, Eberini I. rHDL modeling and the anchoring mechanism of LCAT activation. J Lipid Res 2020; 62:100006. [PMID: 33518511 PMCID: PMC7859856 DOI: 10.1194/jlr.ra120000843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 11/29/2022] Open
Abstract
Lecithin:cholesterol-acyl transferase (LCAT) plays a major role in cholesterol metabolism as it is the only extracellular enzyme able to esterify cholesterol. LCAT activity is required for lipoprotein remodeling and, most specifically, for the growth and maturation of HDLs. In fact, genetic alterations affecting LCAT functionality may cause a severe reduction in plasma levels of HDL-cholesterol with important clinical consequences. Although several hypotheses were formulated, the exact molecular recognition mechanism between LCAT and HDLs is still unknown. We employed a combination of structural bioinformatics procedures to deepen the insights into the HDL-LCAT interplay that promotes LCAT activation and cholesterol esterification. We have generated a data-driven model of reconstituted HDL (rHDL) and studied the dynamics of an assembled rHDL::LCAT supramolecular complex, pinpointing the conformational changes originating from the interaction between LCAT and apolipoprotein A-I (apoA-I) that are necessary for LCAT activation. Specifically, we propose a mechanism in which the anchoring of LCAT lid to apoA-I helices allows the formation of a hydrophobic hood that expands the LCAT active site and shields it from the solvent, allowing the enzyme to process large hydrophobic substrates.
Collapse
Affiliation(s)
- Tommaso Laurenzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Chiara Parravicini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Gianazza
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Laura Calabresi
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari & DSRC, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
29
|
Malajczuk CJ, Gandhi NS, Mancera RL. Structure and intermolecular interactions in spheroidal high-density lipoprotein subpopulations. JOURNAL OF STRUCTURAL BIOLOGY-X 2020; 5:100042. [PMID: 33437963 PMCID: PMC7788233 DOI: 10.1016/j.yjsbx.2020.100042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/13/2022]
Abstract
High-density lipoprotein subpopulations have unique surface profiles and dynamics. Relative hydrophobic surface area decreases with increasing lipoprotein size. Core lipid exposure at the lipoprotein surface decreases with increasing size. Cholesterol molecules localise near apolipoprotein A-I central helices. Lipid and protein interactions stabilise multifoil models of apolipoprotein A-I.
Human serum high-density lipoproteins (HDLs) are a population of small, dense protein-lipid aggregates that are crucial for intravascular lipid trafficking and are protective against cardiovascular disease. The spheroidal HDL subfraction can be separated by size and density into five major subpopulations with distinct molecular compositions and unique biological functionalities: HDL3c, HDL3b, HDL3a, HDL2a and HDL2b. Representative molecular models of these five subpopulations were developed and characterised for the first time in the presence of multiple copies of its primary protein component apolipoprotein A-I (apoA-I) using coarse-grained molecular dynamics simulations. Each HDL model exhibited size, morphological and compositional profiles consistent with experimental observables. With increasing particle size the separation of core and surface molecules became progressively more defined, resulting in enhanced core lipid mixing, reduced core lipid exposure at the surface, and the formation of an interstitial region between core and surface molecules in HDL2b. Cholesterol molecules tended to localise around the central helix-5 of apoA-I, whilst triglyceride molecules predominantly interacted with aromatic, hydrophobic residues located within the terminal helix-10 across all subpopulation models. The three intermediate HDL models exhibited similar surface profiles despite having distinct molecular compositions. ApoA-I in trefoil, quatrefoil and pentafoil arrangements across the surface of HDL particles exhibited significant warping and twisting, but largely retained intermolecular contacts between adjacent apoA-I chains. Representative HDL subpopulations differed in particle size, morphology, intermolecular interaction profiles and lipid and protein dynamics. These findings reveal how different HDL subpopulations might exhibit distinct functional associations depending on particle size, form and composition.
Collapse
Affiliation(s)
- Chris J Malajczuk
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Neha S Gandhi
- School of Mathematical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Ricardo L Mancera
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| |
Collapse
|
30
|
Roh SH, Shekhar M, Pintilie G, Chipot C, Wilkens S, Singharoy A, Chiu W. Cryo-EM and MD infer water-mediated proton transport and autoinhibition mechanisms of V o complex. SCIENCE ADVANCES 2020; 6:6/41/eabb9605. [PMID: 33028525 PMCID: PMC7541076 DOI: 10.1126/sciadv.abb9605] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/17/2020] [Indexed: 05/15/2023]
Abstract
Rotary vacuolar adenosine triphosphatases (V-ATPases) drive transmembrane proton transport through a Vo proton channel subcomplex. Despite recent high-resolution structures of several rotary ATPases, the dynamic mechanism of proton pumping remains elusive. Here, we determined a 2.7-Å cryo-electron microscopy (cryo-EM) structure of yeast Vo proton channel in nanodisc that reveals the location of ordered water molecules along the proton path, details of specific protein-lipid interactions, and the architecture of the membrane scaffold protein. Moreover, we uncover a state of Vo that shows the c-ring rotated by ~14°. Molecular dynamics simulations demonstrate that the two rotary states are in thermal equilibrium and depict how the protonation state of essential glutamic acid residues couples water-mediated proton transfer with c-ring rotation. Our cryo-EM models and simulations also rationalize a mechanism for inhibition of passive proton transport as observed for free Vo that is generated as a result of V-ATPase regulation by reversible disassembly in vivo.
Collapse
Affiliation(s)
- Soung-Hun Roh
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea.
| | - Mrinal Shekhar
- Biodesign Institute, School of Molecular Sciences, Arizona State University, Tempe, AZ 85801, USA
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Grigore Pintilie
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Christophe Chipot
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Laboratoire International Associé CNRS-UIUC, UMR 7019, Université de Lorraine, 54506 Vandœuvre-lès-Nancy, France
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | - Abhishek Singharoy
- Biodesign Institute, School of Molecular Sciences, Arizona State University, Tempe, AZ 85801, USA.
| | - Wah Chiu
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, CA 94305, USA.
- Division of Cryo-EM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA 94025, USA
| |
Collapse
|
31
|
Dergunov AD, Litvinov DY, Malkov AA, Baserova VB, Nosova EV, Dergunova LV. Denaturation of human plasma high-density lipoproteins by urea studied by apolipoprotein A-I dissociation. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158814. [PMID: 32961276 DOI: 10.1016/j.bbalip.2020.158814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/30/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
We studied the mechanism of HDL denaturation with concomitant apoA-I dissociation with HDL preparations from 48 patients with a wide range of plasma HDL-C and evaluated the contribution of lipid-free apoA-I into cholesterol efflux from macrophage, in particular, mediated by cholesterol transporter ABCA1. We prepared HDL by precipitation of apoB-containing lipoproteins by polyethylene glycol and used the chaotropic agent urea to denature HDL preparations. Apo-I dissociation from urea-treated HDL was assessed by the increase of preβ-band fraction with agarose gel electrophoresis followed by electro transfer and immunodetection and by the increase of ABCA1-mediated efflux of fluorescent analogue BODIPY-Cholesterol from RAW 264.7 macrophages. The HDL denaturation is governed by a single transition to fully dissociated apoA-I and the transition cooperativity decreases with increasing HDL-C. The apoA-I release depends on phospholipid concentration of HDL preparation and HDL compositional and structural heterogeneity and is well described by apolipoprotein partition between aqueous and lipid phases. Dissociated apoA-I determines the increase of ABCA1-mediated efflux of BODIPY-Cholesterol from RAW 264.7 macrophages to patient HDL. The increase in apoA-I dissociation is associated with the increase of ABCA1 gene transcript in peripheral blood mononuclear cells from patients. The low level of plasma HDL particles may be compensated by their increased potency for apoA-I release, thus suggesting apoA-I dissociation as a new HDL functional property.
Collapse
Affiliation(s)
- Alexander D Dergunov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Research Centre for Preventive Medicine, Moscow, Russia.
| | - Dmitry Y Litvinov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Research Centre for Preventive Medicine, Moscow, Russia
| | - Artem A Malkov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Research Centre for Preventive Medicine, Moscow, Russia
| | - Veronika B Baserova
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Research Centre for Preventive Medicine, Moscow, Russia
| | - Elena V Nosova
- Laboratory of Functional Genomics, Institute of Molecular Genetics of the Russian Academy of Sciences, Moscow, Russia
| | - Liudmila V Dergunova
- Laboratory of Functional Genomics, Institute of Molecular Genetics of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
32
|
Rangubpit W, Paritanon P, Pandey RB, Sompornpisut P. Thermally induced structural organization of nanodiscs by coarse-grained molecular dynamics simulations. Biophys Chem 2020; 267:106464. [PMID: 32927312 DOI: 10.1016/j.bpc.2020.106464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Abstract
Membrane scaffold proteins (MSP) nanodiscs have been extensively used in structural study of membrane proteins. In cryo-EM, an incorporation of target proteins into nanodiscs is conducted under a rapid change from cryogenic to ambient temperatures. We present a coarse-grained molecular dynamics (CGMD) study for investigating an effect of temperature on the structural organization of DPPC-nanodisc and POPC-nanodisc. A non-monotonic response of physical quantities (i.e. the lipid order parameter, nanodisc flatness, structural change, solvation property, radius of gyration) with increase in temperature (T = 200-350 K) is found to be associated with the gel-ripple-liquid crystalline phase change within nanodiscs. The reorganization of lipids upon temperature variation induced conformational changes of MSP to minimize hydrophobic exposure of the lipid membrane to an aqueous environment. Structural response to temperature is different to a certain extent between the saturated DPPC and unsaturated POPC.
Collapse
Affiliation(s)
- Warin Rangubpit
- Center of Excellence in Computational Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, 10330, Thailand
| | - Pasawan Paritanon
- Center of Excellence in Computational Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, 10330, Thailand
| | - Ras B Pandey
- School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Pornthep Sompornpisut
- Center of Excellence in Computational Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, 10330, Thailand.
| |
Collapse
|
33
|
Yelamanchili D, Liu J, Gotto AM, Hurley AE, Lagor WR, Gillard BK, Davidson WS, Pownall HJ, Rosales C. Highly conserved amino acid residues in apolipoprotein A1 discordantly induce high density lipoprotein assembly in vitro and in vivo. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158794. [PMID: 32810603 DOI: 10.1016/j.bbalip.2020.158794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Apolipoprotein A1 (APOA1) is essential to reverse cholesterol transport, a physiologically important process that protects against atherosclerotic cardiovascular disease. APOA1 is a 28 kDa protein comprising multiple lipid-binding amphiphatic helices initialized by proline residues, which are conserved across multiple species. We tested the hypothesis that the evolutionarily conserved residues are essential to high density lipoprotein (HDL) function. APPROACH We used biophysical and physiological assays of the function of APOA1P➔A variants, i.e., rHDL formation via dimyristoylphosphatidylcholine (DMPC) microsolubilization, activation of lecithin: cholesterol acyltransferase, cholesterol efflux from human monocyte-derived macrophages (THP-1) to each variant, and comparison of the size and composition of HDL from APOA1-/- mice receiving adeno-associated virus delivery of each human variant. RESULTS Differences in microsolubilization were profound and showed that conserved prolines, especially those in the C-terminus of APOA1, are essential to efficient rHDL formation. In contrast, P➔A substitutions produced small changes (-25 to +25%) in rates of cholesterol efflux and no differences in the rates of LCAT activation. The HDL particles formed following ectopic expression of each variant in APOA1-/- mice were smaller and more heterogeneous than those from control animals. CONCLUSION Studies of DMPC microsolubilization show that proline residues are essential to the optimal interaction of APOA1 with membranes, the initial step in cholesterol efflux and HDL production. In contrast, P➔A substitutions modestly reduce the cholesterol efflux capacity of APOA1, have no effect on LCAT activation, but according to the profound reduction in the size of HDL formed in vivo, P➔A substitutions alter HDL biogenesis, thereby implicating other cellular and in vivo processes as determinants of HDL metabolism and function.
Collapse
Affiliation(s)
- Dedipya Yelamanchili
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA.
| | - Jing Liu
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Antonio M Gotto
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Weill Cornell Medicine, 1305 York Avenue, New York, NY 10065, USA.
| | - Ayrea E Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Willam R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Baiba K Gillard
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Weill Cornell Medicine, 1305 York Avenue, New York, NY 10065, USA.
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA.
| | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Weill Cornell Medicine, 1305 York Avenue, New York, NY 10065, USA.
| | - Corina Rosales
- Center for Bioenergetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Weill Cornell Medicine, 1305 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
34
|
Pinals RL, Chio L, Ledesma F, Landry MP. Engineering at the nano-bio interface: harnessing the protein corona towards nanoparticle design and function. Analyst 2020; 145:5090-5112. [PMID: 32608460 PMCID: PMC7439532 DOI: 10.1039/d0an00633e] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Unpredictable and uncontrollable protein adsorption on nanoparticles remains a considerable challenge to achieving effective application of nanotechnologies within biological environments. Nevertheless, engineered nanoparticles offer unprecedented functionality and control in probing and altering biological systems. In this review, we highlight recent advances in harnessing the "protein corona" formed on nanoparticles as a handle to tune functional properties of the protein-nanoparticle complex. Towards this end, we first review nanoparticle properties that influence protein adsorption and design strategies to facilitate selective corona formation, with the corresponding characterization techniques. We next focus on literature detailing corona-mediated functionalities, including stealth to avoid recognition and sequestration while in circulation, targeting of predetermined in vivo locations, and controlled activation once localized to the intended biological compartment. We conclude with a discussion of biocompatibility outcomes for these protein-nanoparticle complexes applied in vivo. While formation of the nanoparticle-corona complex may impede our control over its use for the projected nanobiotechnology application, it concurrently presents an opportunity to create improved protein-nanoparticle architectures by exploiting natural or guiding selective protein adsorption to the nanoparticle surface.
Collapse
Affiliation(s)
- Rebecca L Pinals
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, USA.
| | | | | | | |
Collapse
|
35
|
Florens N, Calzada C, Lemoine S, Boulet MM, Guillot N, Barba C, Roux J, Delolme F, Page A, Poux JM, Laville M, Moulin P, Soulère L, Guebre-Egziabher F, Juillard L, Soulage CO. CKD Increases Carbonylation of HDL and Is Associated with Impaired Antiaggregant Properties. J Am Soc Nephrol 2020; 31:1462-1477. [PMID: 32518085 DOI: 10.1681/asn.2019111205] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 03/22/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND CKD is associated with increased oxidative stress that correlates with occurrence of cardiovascular events. Modifications induced by increased oxidative stress particularly affect circulating lipoproteins such as HDL that exhibit antiatheromatous and antithrombotic properties in vitro. METHODS To explore the specific role of oxidative modifications of HDL in CKD and their effect on the platelet-targeting antiaggregant properties of HDL, we used a CKD (5/6 nephrectomy) rabbit model. For ex vivo assessment of the antiaggregant properties of HDL, we collected blood samples from 15 healthy volunteers, 25 patients on hemodialysis, and 20 on peritoneal dialysis. We analyzed malondialdehyde, 4-hydroxynonenal (HNE), and 4-hydroxy-2-hexenal protein adduct levels. Platelet aggregation and activation were assessed by aggregometry, thromboxane B2 assay, or FACS. We modified HDL from controls by incubating it overnight at 37°C with 100 µM of HNE. RESULTS HDL from CKD rabbits and patients on hemodialysis had HNE adducts. The percentage of platelet aggregation or activation induced by collagen was significantly higher when platelets were incubated with HDL from CKD rabbit and hemodialysis groups than with HDL from the control group. In both rabbits and humans, platelet aggregation and activation were significantly higher in the presence of HNE-modified HDL than with HDL from their respective controls. Incubation of platelets with a blocking antibody directed against CD36 or with a pharmacologic inhibitor of SRC kinases restored the antiaggregative phenotype in the presence of HDL from CKD rabbits, patients on hemodialysis and peritoneal dialysis, and HNE-modified HDL. CONCLUSIONS HDL from CKD rabbits and patients on hemodialysis exhibited an impaired ability to inhibit platelet aggregation, suggesting that altered HDL properties may contribute to the increased cardiovascular risk in this population.
Collapse
Affiliation(s)
- Nans Florens
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France .,Department of Nephrology, University Hospital of Lyon, E. Herriot Hospital, Lyon, France
| | - Catherine Calzada
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| | - Sandrine Lemoine
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France.,Department of Nephrology, University Hospital of Lyon, E. Herriot Hospital, Lyon, France
| | - Marie Michèle Boulet
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| | - Nicolas Guillot
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| | - Christophe Barba
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| | - Julie Roux
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| | - Fréderic Delolme
- Protein Science Facility, SFR BioSciences, Centre National de la Recherche Scientifique (CNRS) UMS3444, INSERM US8, Claude Bernard University Lyon 1, École Normale Supérieure de Lyon (ENS de Lyon), Lyon, France
| | - Adeline Page
- Protein Science Facility, SFR BioSciences, Centre National de la Recherche Scientifique (CNRS) UMS3444, INSERM US8, Claude Bernard University Lyon 1, École Normale Supérieure de Lyon (ENS de Lyon), Lyon, France
| | - Jean Michel Poux
- Association Pour l'Utilisation du Rein Artificiel dans la Région Lyonnaise (AURAL), Lyon, France
| | - Maurice Laville
- Association Pour l'Utilisation du Rein Artificiel dans la Région Lyonnaise (AURAL), Lyon, France
| | - Philippe Moulin
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France.,Department of Endocrinology, University Hospital of Lyon, L. Pradel Hospital, Bron, France
| | - Laurent Soulère
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, University of Lyon, INSA-Lyon, UMR 5246 CNRS, Villeurbanne, France
| | - Fitsum Guebre-Egziabher
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France.,Department of Nephrology, University Hospital of Lyon, E. Herriot Hospital, Lyon, France
| | - Laurent Juillard
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France.,Department of Nephrology, University Hospital of Lyon, E. Herriot Hospital, Lyon, France
| | - Christophe O Soulage
- CarMeN Laboratory, University of Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) U1060, Institut National des Sciences Appliquées de Lyon (INSA-Lyon), Claude Bernard University Lyon 1, Institut National de la Recherche Agronomique (INRA) U1397, Villeurbanne, France
| |
Collapse
|
36
|
Andreeva AM, Toropygin IY, Garina DV, Lamash NE, Vasiliev AS. The Role of High-Density Lipoproteins
in Maintaining Osmotic Homeostasis in the Goldfish Carassius auratus L. (Cyprinidae). J EVOL BIOCHEM PHYS+ 2020. [DOI: 10.1134/s0022093020020027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Ciccone L, Shi C, di Lorenzo D, Van Baelen AC, Tonali N. The Positive Side of the Alzheimer's Disease Amyloid Cross-Interactions: The Case of the Aβ 1-42 Peptide with Tau, TTR, CysC, and ApoA1. Molecules 2020; 25:E2439. [PMID: 32456156 PMCID: PMC7288020 DOI: 10.3390/molecules25102439] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) represents a progressive amyloidogenic disorder whose advancement is widely recognized to be connected to amyloid-β peptides and Tau aggregation. However, several other processes likely contribute to the development of AD and some of them might be related to protein-protein interactions. Amyloid aggregates usually contain not only single type of amyloid protein, but also other type of proteins and this phenomenon can be rationally explained by the process of protein cross-seeding and co-assembly. Amyloid cross-interaction is ubiquitous in amyloid fibril formation and so a better knowledge of the amyloid interactome could help to further understand the mechanisms of amyloid related diseases. In this review, we discuss about the cross-interactions of amyloid-β peptides, and in particular Aβ1-42, with other amyloids, which have been presented either as integrated part of Aβ neurotoxicity process (such as Tau) or conversely with a preventive role in AD pathogenesis by directly binding to Aβ (such as transthyretin, cystatin C and apolipoprotein A1). Particularly, we will focus on all the possible therapeutic strategies aiming to rescue the Aβ toxicity by taking inspiration from these protein-protein interactions.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Chenghui Shi
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| | - Davide di Lorenzo
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| | - Anne-Cécile Van Baelen
- Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris Saclay, SIMoS, 91191 Gif-sur-Yvette, France;
| | - Nicolo Tonali
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| |
Collapse
|
38
|
Chuang ST, Cruz S, Narayanaswami V. Reconfiguring Nature's Cholesterol Accepting Lipoproteins as Nanoparticle Platforms for Transport and Delivery of Therapeutic and Imaging Agents. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E906. [PMID: 32397159 PMCID: PMC7279153 DOI: 10.3390/nano10050906] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
Apolipoproteins are critical structural and functional components of lipoproteins, which are large supramolecular assemblies composed predominantly of lipids and proteins, and other biomolecules such as nucleic acids. A signature feature of apolipoproteins is the preponderance of amphipathic α-helical motifs that dictate their ability to make extensive non-covalent inter- or intra-molecular helix-helix interactions in lipid-free states or helix-lipid interactions with hydrophobic biomolecules in lipid-associated states. This review focuses on the latter ability of apolipoproteins, which has been capitalized on to reconstitute synthetic nanoscale binary/ternary lipoprotein complexes composed of apolipoproteins/peptides and lipids that mimic native high-density lipoproteins (HDLs) with the goal to transport drugs. It traces the historical development of our understanding of these nanostructures and how the cholesterol accepting property of HDL has been reconfigured to develop them as drug-loading platforms. The review provides the structural perspective of these platforms with different types of apolipoproteins and an overview of their synthesis. It also examines the cargo that have been loaded into the core for therapeutic and imaging purposes. Finally, it lays out the merits and challenges associated with apolipoprotein-based nanostructures with a future perspective calling for a need to develop "zip-code"-based delivery for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
| | | | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry, California State University, Long Beach, 1250 Bellflower Blvd, Long Beach, CA 90840, USA; (S.T.C.); (S.C.)
| |
Collapse
|
39
|
Wzorek J, Bednarek R, Watala C, Boncler M. Binding of adenosine derivatives to carrier proteins may reduce their antiplatelet activity. Biochem Pharmacol 2020; 174:113827. [PMID: 31987853 DOI: 10.1016/j.bcp.2020.113827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/22/2020] [Indexed: 01/03/2023]
Abstract
Adenosine analogues have high affinity and selectivity for adenosine receptors (AR), and exhibit anti-platelet activity. Plasma proteins play an important role in the regulation of platelet function and may influence the action of anti-platelet compounds. Little is known about the interactions of AR agonists with plasma proteins. This study investigates the interplay between AR agonists and plasma proteins and the consequences of those interactions. Surface plasmon resonance was employed together with molecular docking study to determine the binding kinetics of four selected ARagonists (PSB0777, Cl-Ado, MRE0094, UK432097) to several carrier proteins and to clarify the nature of these interactions. The influence of a whole plasma and of some plasma components on the effectiveness of ARagonists in the inhibition of platelet function was assessed by flow cytometry (platelet activation) and ELISA (platelet adhesion). Plasma proteins remarkably diminished the effectiveness of ARagonists in inhibiting platelet activation and adhesion in vitro. ARagonists were found to strongly bind to human serum albumin (HSA) and the protein components of lipoproteins - apolipoproteins; HSA was essential for the binding of water-soluble PSB0777, whereas apolipoproteins were needed for interactions with poorly-water soluble compounds such as UK432097 and MRE0094. In addition, HSA was shown to significantly reduce the effectiveness of PSB0777 in inhibiting ADP-induced platelet activation. In conclusion, HSA and lipoproteins are important carriers for ARagonists, which can affect pharmacodynamics of ARagonists used as platelet inhibitors.
Collapse
Affiliation(s)
- Joanna Wzorek
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Radosław Bednarek
- Department of Cytobiology and Proteomics, Medical University of Lodz, Lodz, Poland
| | - Cezary Watala
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Magdalena Boncler
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
40
|
Abstract
The apolipoproteins are well known for their roles in both health and disease, as components of plasma lipoprotein particles, such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), very-low-density lipoprotein (VLDL), chylomicrons, and metabolic, vascular- and inflammation-related disorders, such as cardiovascular disease, atherosclerosis, metabolic syndrome, and diabetes. Increasingly, their roles in neurovascular and neurodegenerative disorders are also being elucidated. They play major roles in lipid and cholesterol transport between blood and organs and are, therefore, critical to maintenance and homeostasis of the lipidome, with apolipoprotein-lipid interactions, including cholesterol, fatty acids, triglycerides, phospholipids, and isoprostanes. Further, they have important pleiotropic roles related to aging and longevity, which are largely managed through their many structural variants, including multiple isoforms, and a diversity of post-translational modifications. Consequently, tools for the characterization and accurate quantification of apolipoproteins, including their diverse array of variant forms, are required to understand their salutary and disease related roles. In this chapter we outline three distinct quantitative approaches suitable for targeting apolipoproteins: (1) multiplex immunoassays, (2) mass spectrometric immunoassay, and (3) multiple reaction monitoring, mass spectrometric quantification. We also discuss management of pre-analytical and experimental design variables.
Collapse
|
41
|
Gkolfinopoulou C, Bourtsala A, Chroni A. Structural and functional basis for increased HDL-cholesterol levels due to the naturally occurring V19L mutation in human apolipoprotein A-I. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158593. [PMID: 31863971 DOI: 10.1016/j.bbalip.2019.158593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/09/2019] [Accepted: 12/16/2019] [Indexed: 11/26/2022]
Abstract
Several hereditary point mutations in human apolipoprotein A-I (apoA-I) have been associated with low HDL-cholesterol levels and/or increased coronary artery disease (CAD) risk. However, one apoA-I mutation, the V19L, recently identified in Icelanders, has been associated with increased HDL-cholesterol levels and decreased CAD risk. In an effort to gain mechanistic insight linking the presence of this mutation in apoA-I with the increase of HDL-cholesterol levels we evaluated the effect of V19L mutation on the conformational integrity and functional properties of apoA-I in lipid-free and lipidated form. ApoA-I[V19L] was found to be thermodynamically destabilized in lipid-free form and displays an increased capacity to associate with phospholipids compared to WT apoA-I. When associated to reconstituted HDL (rHDL), apoA-I[V19L] was more thermodynamically stabilized than WT apoA-I. ApoA-I[V19L] displayed normal capacity to promote ABCA1-mediated cholesterol efflux and to activate the enzyme LCAT, in lipid-free and rHDL-associated forms, respectively. Additionally, rHDL-associated apoA-I[V19L] showed normal capacity to promote ABCG1-mediated cholesterol efflux, but 45% increased capacity to promote SR-BI-mediated cholesterol efflux, while the SR-BI-mediated HDL-lipid uptake was normal. Overall, our findings show that the apoA-I V19L mutation does not affect the first steps of HDL biogenesis pathway. However, the increased capacity of apoA-I[V19L] to associate with phospholipids, in combination with the enhanced thermodynamic stability of lipoprotein-associated apoA-I[V19L] and increased capacity of apoA-I[V19L]-containing lipoprotein particles to accept additional cholesterol by SR-BI could account for the increased HDL-cholesterol levels observed in human carriers of the mutation.
Collapse
Affiliation(s)
- Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Angeliki Bourtsala
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece.
| |
Collapse
|
42
|
Qi Y, Lee J, Klauda JB, Im W. CHARMM-GUI Nanodisc Builder for modeling and simulation of various nanodisc systems. J Comput Chem 2019; 40:893-899. [PMID: 30677169 DOI: 10.1002/jcc.25773] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/13/2023]
Abstract
Nanodiscs are discoidal protein-lipid complexes that have wide applications in membrane protein studies. Modeling and simulation of nanodiscs are challenging due to the absence of structures of many membrane scaffold proteins (MSPs) that wrap around the membrane bilayer. We have developed CHARMM-GUI Nanodisc Builder (http://www.charmm-gui.org/input/nanodisc) to facilitate the setup of nanodisc simulation systems by modeling the MSPs with defined size and known structural features. A total of 11 different nanodiscs with a diameter from 80 to 180 Å are made available in both the all-atom CHARMM and two coarse-grained (PACE and Martini) force fields. The usage of the Nanodisc Builder is demonstrated with various simulation systems. The structures and dynamics of proteins and lipids in these systems were analyzed, showing similar behaviors to those from previous all-atom and coarse-grained nanodisc simulations. We expect the Nanodisc Builder to be a convenient and reliable tool for modeling and simulation of nanodisc systems. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yifei Qi
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Jumin Lee
- Departments of Biological Sciences and Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering and the Biophysics Program, University of Maryland, College Park, Maryland
| | - Wonpil Im
- Departments of Biological Sciences and Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
43
|
Magro M, Baratella D, Bonaiuto E, de Almeida Roger J, Chemello G, Pasquaroli S, Mancini L, Olivotto I, Zoppellaro G, Ugolotti J, Aparicio C, Fifi AP, Cozza G, Miotto G, Radaelli G, Bertotto D, Zboril R, Vianello F. Stealth Iron Oxide Nanoparticles for Organotropic Drug Targeting. Biomacromolecules 2019; 20:1375-1384. [PMID: 30694655 DOI: 10.1021/acs.biomac.8b01750] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The ability of peculiar iron oxide nanoparticles (IONPs) to evade the immune system was investigated in vivo. The nanomaterial was provided directly into the farming water of zebrafish ( Danio rerio) and the distribution of IONPs and the delivery of oxytetracycline (OTC) was studied evidencing the successful overcoming of the intestinal barrier and the specific and prolonged (28 days) organotropic delivery of OTC to the fish ovary. Noteworthy, no sign of adverse effects was observed. In fish blood, IONPs were able to specifically bind apolipoprotein A1 (Apo A1) and molecular modeling showed the structural analogy between the IONP@Apo A1 nanoconjugate and high-density lipoprotein (HDL). Thus, the preservation of the biological identity of the protein suggests a plausible explanation of the observed overcoming of the intestinal barrier, of the great biocompatibity of the nanomaterial, and of the prolonged drug delivery (benefiting of the lipoprotein transport route). The present study promises novel and unexpected stealth materials in nanomedicine.
Collapse
Affiliation(s)
- Massimiliano Magro
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Davide Baratella
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Emanuela Bonaiuto
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Jessica de Almeida Roger
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Giulia Chemello
- Department of Life and Environmental Sciences , Marche Polytechnic University , via Brecce Bianche , Ancona , 60131 , Italy
| | - Sonia Pasquaroli
- Department of Life and Environmental Sciences , Marche Polytechnic University , via Brecce Bianche , Ancona , 60131 , Italy
| | - Leonardo Mancini
- Department of Life and Environmental Sciences , Marche Polytechnic University , via Brecce Bianche , Ancona , 60131 , Italy
| | - Ike Olivotto
- Department of Life and Environmental Sciences , Marche Polytechnic University , via Brecce Bianche , Ancona , 60131 , Italy
| | - Giorgio Zoppellaro
- Department of Physical Chemistry, Regional Centre of Advanced Technologies and Materials , Palacky University in Olomouc , Šlechtitelů , Olomouc 78371 , Czech Republic
| | - Juri Ugolotti
- Department of Physical Chemistry, Regional Centre of Advanced Technologies and Materials , Palacky University in Olomouc , Šlechtitelů , Olomouc 78371 , Czech Republic
| | - Claudia Aparicio
- Department of Physical Chemistry, Regional Centre of Advanced Technologies and Materials , Palacky University in Olomouc , Šlechtitelů , Olomouc 78371 , Czech Republic
| | - Anna P Fifi
- BioTecnologie BT S.r.l. , Agrifood Technology Park of Umbria , Frazione Pantalla , Pantalla , 06059 , Italy
| | - Giorgio Cozza
- Department of Molecular Medicine , University of Padua , Viale G. Colombo , Padova , 35121 , Italy
| | - Giovanni Miotto
- Department of Molecular Medicine , University of Padua , Viale G. Colombo , Padova , 35121 , Italy
| | - Giuseppe Radaelli
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Daniela Bertotto
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| | - Radek Zboril
- Department of Physical Chemistry, Regional Centre of Advanced Technologies and Materials , Palacky University in Olomouc , Šlechtitelů , Olomouc 78371 , Czech Republic
| | - Fabio Vianello
- Department of Comparative Biomedicine and Food Science , University of Padua , Viale dell'Università , Legnaro , 35020 , Italy
| |
Collapse
|
44
|
He Y, Song HD, Anantharamaiah GM, Palgunachari MN, Bornfeldt KE, Segrest JP, Heinecke JW. Apolipoprotein A1 Forms 5/5 and 5/4 Antiparallel Dimers in Human High-density Lipoprotein. Mol Cell Proteomics 2019; 18:854-864. [PMID: 30659061 DOI: 10.1074/mcp.ra118.000878] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
Apolipoprotein A1 (APOA1), the major protein of high-density lipoprotein (HDL), contains 10 helical repeats that play key roles in protein-protein and protein-lipid interactions. The current structural model for HDL proposes that APOA1 forms an antiparallel dimer in which helix 5 in monomer 1 associates with helix 5 in monomer 2 along a left-left (LL5/5) interface, forming a protein complex with a 2-fold axis of symmetry centered on helix 5. However, computational studies suggest that other orientations are possible. To test this idea, we used a zero-length chemical cross-linking reagent that forms covalent bonds between closely apposed basic and acidic residues. Using proteolytic digestion and tandem mass spectrometry, we identified amino acids in the central region of the antiparallel APOA1 dimer of HDL that were in close contact. As predicted by the current model, we found six intermolecular cross-links that were consistent with the antiparallel LL5/5 registry. However, we also identified three intermolecular cross-links that were consistent with the antiparallel LL5/4 registry. The LL5/5 is the major structural conformation of the two complexes in both reconstituted discoidal HDL particles and in spherical HDL from human plasma. Molecular dynamic simulations suggest that that LL5/5 and LL5/4 APOA1 dimers possess similar free energies of dimerization, with LL5/5 having the lowest free energy. Our observations indicate that phospholipidated APOA1 in HDL forms different antiparallel dimers that could play distinct roles in enzyme regulation, assembly of specific protein complexes, and the functional properties of HDL in humans.
Collapse
Affiliation(s)
- Yi He
- From the Departments of ‡Medicine and
| | - Hyun D Song
- ‖Department of Medicine, Vanderbilt University, Nashville, Tennessee, 37240
| | - G M Anantharamaiah
- ¶Department of Medicine, University of Alabama at Birmingham, Alabama 35233
| | - M N Palgunachari
- ¶Department of Medicine, University of Alabama at Birmingham, Alabama 35233
| | - Karin E Bornfeldt
- From the Departments of ‡Medicine and; §Pathology, University of Washington, Seattle, Washington, 98109
| | - Jere P Segrest
- ‖Department of Medicine, Vanderbilt University, Nashville, Tennessee, 37240
| | | |
Collapse
|
45
|
McPherson A, Larson SB. The structure of human apolipoprotein C-1 in four different crystal forms. J Lipid Res 2018; 60:400-411. [PMID: 30559175 PMCID: PMC6358290 DOI: 10.1194/jlr.m089441] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/28/2018] [Indexed: 02/02/2023] Open
Abstract
Human apolipoprotein C1 (APOC1) is a 57 amino acid long polypeptide that, through its potent inhibition of cholesteryl ester transferase protein, helps regulate the transfer of lipids between lipid particles. We have now determined the structure of APOC1 in four crystal forms by X-ray diffraction. A molecule of APOC1 is a single, slightly bent, α-helix having 13–14 turns and a length of about 80 Å. APOC1 exists as a dimer, but the dimers are not the same in the four crystals. In two monoclinic crystals, two helices closely engage one another in an antiparallel fashion. The interactions between monomers are almost entirely hydrophobic with sparse electrostatic complements. In the third monoclinic crystal, the two monomers spread at one end of the dimer, like a scissor opening, and, by translation along the crystallographic a axis, form a continuous, contiguous sheet through the crystal. In the orthorhombic crystals, two molecules of APOC1 are related by a noncrystallographic 2-fold axis to create an arc of about 120 Å length. This symmetrical dimer utilizes interactions not present in dimers of the monoclinic crystals. Versatility of APOC1 monomer association shown by these crystals is suggestive of physiological function.
Collapse
Affiliation(s)
- Alexander McPherson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900
| | - Steven B Larson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900
| |
Collapse
|
46
|
Ho YT, Azman N‘A, Loh FWY, Ong GKT, Engudar G, Kriz SA, Kah JCY. Protein Corona Formed from Different Blood Plasma Proteins Affects the Colloidal Stability of Nanoparticles Differently. Bioconjug Chem 2018; 29:3923-3934. [DOI: 10.1021/acs.bioconjchem.8b00743] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yan Teck Ho
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), 28 Medical Drive, #05-01, Singapore 117456
| | - Nurul ‘Ain Azman
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583
| | - Fion Wen Yee Loh
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583
| | - Gabriella Kai Teng Ong
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583
| | - Gokce Engudar
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583
| | - Seth Allan Kriz
- Department of Chemical Engineering, Michigan Technological University, Building 203, 1400 Townsend Drive, Houghton, Michigan 49931, United States
| | - James Chen Yong Kah
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Centre for Life Sciences (CeLS), 28 Medical Drive, #05-01, Singapore 117456
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583
| |
Collapse
|
47
|
Zhao Y, Cieplak M. Stability of structurally entangled protein dimers. Proteins 2018; 86:945-955. [DOI: 10.1002/prot.25526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Yani Zhao
- Institute of Physics, Polish Academy of Sciences; Aleja Lotników 32/46, Warsaw 02668 Poland
| | - Marek Cieplak
- Institute of Physics, Polish Academy of Sciences; Aleja Lotników 32/46, Warsaw 02668 Poland
| |
Collapse
|
48
|
Liu D, Ji L, Zhao M, Wang Y, Guo Y, Li L, Zhang D, Xu L, Pan B, Su J, Xiang S, Pennathur S, Li J, Gao J, Liu P, Willard B, Zheng L. Lysine glycation of apolipoprotein A-I impairs its anti-inflammatory function in type 2 diabetes mellitus. J Mol Cell Cardiol 2018; 122:47-57. [PMID: 30092227 DOI: 10.1016/j.yjmcc.2018.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/16/2022]
Abstract
Apolipoprotein A-I (apoA-I), the major protein compontent of high-density lipoprotein (HDL), exerts many anti-atherogenic functions. This study aimed to reveal whether nonenzymatic glycation of specific sites of apoA-I impaired its anti-inflammatory effects in type 2 diabetes mellitus (T2DM). LC-MS/MS was used to analyze the specific sites and the extent of apoA-I glycation either modified by glucose in vitro or isolated from T2DM patients. Cytokine release in THP-1 monocyte-derived macrophages was tested by ELISA. Activation of NF-kappa B pathway was detected by western blot. The binding affinity of apoA-I to THP-1 cells was measured using 125I-labeled apoA-I. We identified seven specific lysine (Lys, K) residues of apoA-I (K12, K23, K40, K96, K106, K107 and K238) that were susceptible to be glycated either in vitro or in vivo. Glycation of apoA-I impaired its abilities to inhibit the release of TNF-α and IL-1β against lipopolysaccharide (LPS) in THP-1 cells. Besides, the glycation levels of these seven K sites in apoA-I were inversely correlated with its anti-inflammatory abilities. Furthermore, glycated apoA-I had a lower affinity to THP-1 cells than native apoA-I had. We generated mutant apoA-I (K107E, M-apoA-I) with a substitution of glutamic acid (Glu, E) for lysine at the 107th site, and found that compared to wild type apoA-I (WT-apoA-I), M-apoA-I decreased its anti-inflammatory effects in THP-1 cells. We also modeled the location of these seven K residues on apoA-I which allowed us to infer the conformational alteration of glycated apoA-I and HDL. In summary, glycation of these seven K residues altered the conformation of apoA-I and consequently impaired the protective effects of apoA-I, which may partly account for the increased risk of cardiovascular disease (CVD) in diabetic subjects.
Collapse
Affiliation(s)
- Donghui Liu
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China; Department of Cardiology, the Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, Fujian 361004, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China
| | - Yang Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yansong Guo
- Department of Cardiovascular Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Ling Li
- Proteomics Laboratory, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongmei Zhang
- Proteomics Laboratory, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Liang Xu
- Department of Cardiology, the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China
| | - Jinzi Su
- Department of Cardiology, the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Song Xiang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 294 Taiyuan Road, Shanghai 200031, China
| | | | - Jingxuan Li
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China
| | - Jianing Gao
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Belinda Willard
- Proteomics Laboratory, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Health Science Center, 100191 Beijing, China.
| |
Collapse
|
49
|
Wilson CJ, Das M, Jayaraman S, Gursky O, Engen JR. Effects of Disease-Causing Mutations on the Conformation of Human Apolipoprotein A-I in Model Lipoproteins. Biochemistry 2018; 57:4583-4596. [PMID: 30004693 DOI: 10.1021/acs.biochem.8b00538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Plasma high-density lipoproteins (HDLs) are protein-lipid nanoparticles that transport lipids and protect against atherosclerosis. Human apolipoprotein A-I (apoA-I) is the principal HDL protein whose mutations can cause either aberrant lipid metabolism or amyloid disease. Hydrogen-deuterium exchange (HDX) mass spectrometry (MS) was used to study the apoA-I conformation in model discoidal lipoproteins similar in size to large plasma HDL. We examined how point mutations associated with hereditary amyloidosis (F71Y and L170P) or atherosclerosis (L159R) influence the local apoA-I conformation in model lipoproteins. Unlike other apoA-I forms, the large particles showed minimal conformational heterogeneity, suggesting a fully extended protein conformation. Mutation-induced structural perturbations in lipid-bound protein were attenuated compared to the free protein and indicated close coupling between the two belt-forming apoA-I molecules. These perturbations propagated to distant lipoprotein sites, either increasing or decreasing their protection. This HDX MS study of large model HDL, compared with previous studies of smaller particles, ascertained that apoA-I's central region helps accommodate the protein conformation to lipoproteins of various sizes. This study also reveals that the effects of mutations on lipoprotein conformational dynamics are much weaker than those in a lipid-free protein. Interestingly, the mutation-induced perturbations propagate to distant sites nearly 10 nm away and alter their protection in ways that cannot be predicted from the lipoprotein structure and stability. We propose that long-range mutational effects are mediated by both protein and lipid and can influence lipoprotein functionality.
Collapse
Affiliation(s)
- Christopher J Wilson
- Department of Chemistry and Chemical Biology , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - Madhurima Das
- Department of Physiology & Biophysics , Boston University School of Medicine , 700 Albany Street , Boston , Massachusetts 02118 , United States
| | - Shobini Jayaraman
- Department of Physiology & Biophysics , Boston University School of Medicine , 700 Albany Street , Boston , Massachusetts 02118 , United States
| | - Olga Gursky
- Department of Physiology & Biophysics , Boston University School of Medicine , 700 Albany Street , Boston , Massachusetts 02118 , United States.,Amyloidosis Research Center , Boston University School of Medicine , Boston , Massachusetts 02118 , United States
| | - John R Engen
- Department of Chemistry and Chemical Biology , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
50
|
Zhao Y, Chwastyk M, Cieplak M. Structural entanglements in protein complexes. J Chem Phys 2018; 146:225102. [PMID: 29166058 DOI: 10.1063/1.4985221] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We consider multi-chain protein native structures and propose a criterion that determines whether two chains in the system are entangled or not. The criterion is based on the behavior observed by pulling at both termini of each chain simultaneously in the two chains. We have identified about 900 entangled systems in the Protein Data Bank and provided a more detailed analysis for several of them. We argue that entanglement enhances the thermodynamic stability of the system but it may have other functions: burying the hydrophobic residues at the interface and increasing the DNA or RNA binding area. We also study the folding and stretching properties of the knotted dimeric proteins MJ0366, YibK, and bacteriophytochrome. These proteins have been studied theoretically in their monomeric versions so far. The dimers are seen to separate on stretching through the tensile mechanism and the characteristic unraveling force depends on the pulling direction.
Collapse
Affiliation(s)
- Yani Zhao
- Institute of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL-02668 Warsaw, Poland
| | - Mateusz Chwastyk
- Institute of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL-02668 Warsaw, Poland
| | - Marek Cieplak
- Institute of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL-02668 Warsaw, Poland
| |
Collapse
|