1
|
Apley KD, Bass LE, King J, Downes G, Wang K, Forchetti MV, Moore DJ, Kendall P, Bonami RH, Berkland CJ. Evaluation of proinsulin(F25D) as a targeting ligand for insulin-binding B cells in autoimmune diabetes. Drug Deliv Transl Res 2025:10.1007/s13346-025-01869-x. [PMID: 40402465 DOI: 10.1007/s13346-025-01869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2025] [Indexed: 05/23/2025]
Abstract
Insulin-binding B cells are implicated in Type 1 Diabetes (T1D) pathology. Antigen-specific immunotherapy (ASIT) holds promise in T1D. However, ASIT-targeted suppression of insulin-binding B cells is hampered by insulin's hormonal activity and the resulting binding and endocytosis of insulin by insulin receptors (INSR). To evaluate ASIT strategies that target insulin-binding B cells in vivo, non-hormonally active insulin variants are needed. In this work, we aimed to improve upon prior non-hormonal insulin variants by making mutations to the insulin precursor, proinsulin, and including a c-terminal sortase (SrtA) tag (LPETGGHG) to enable facile site-selective bioconjugation to scaffolds or payloads. Of the insulin variants investigated that retained low-nM binding to the murine-derived insulin autoantibody mAb 125, proinsulin(F25D)-SrtA had the lowest INSR binding and activity and the greatest fibrillation resistance. Compared to desoctapeptide insulin, a previously proposed non-hormonal insulin variant, proinsulin(F25D)-SrtA demonstrated 50-fold lower INSR binding and 100-fold greater fibrillation lag time. However, insulin(F25D)-SrtA bound to the anti-insulin antibody 12M4 isolated from a presymptomatic T1D individual, whereas proinsulin(F25D)-SrtA and desoctapeptide insulin did not, highlighting the potential for anti-insulin B cells to develop in human T1D that would escape this ASIT moiety. The characteristics of proinsulin(F25D)-SrtA make it a well-suited non-hormonal insulin variant for insulin-binding B cell targeting and warrants additional study with other anti-insulin B cell specificities derived from T1D individuals.
Collapse
Affiliation(s)
- Kyle D Apley
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63130, USA
| | - Lindsay E Bass
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jaylyn King
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Grant Downes
- Bioengineering Program, University of Kansas, Lawrence, KS, 66045, USA
| | - Kristen Wang
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA
| | - Mason V Forchetti
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Daniel J Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Division of Endocrinology & Diabetes, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Peggy Kendall
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - Rachel H Bonami
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Cory J Berkland
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63130, USA.
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA.
| |
Collapse
|
2
|
Li M, Qi L, Huang J, Li H, Cheng W, Shi Z, Jiang X, Zhou Y, Jiang W. The Novel Long-Acting Peptide S6-FA Attenuates Liver Fibrosis In Vitro and In Vivo. ACS OMEGA 2025; 10:9661-9674. [PMID: 40092780 PMCID: PMC11904669 DOI: 10.1021/acsomega.4c10956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025]
Abstract
Liver fibrosis can progress to cirrhosis and hepatocellular carcinoma. Currently, there is no effective drug for liver fibrosis. The peptide 6 (T6) is an endogenous peptide derived from human intrauterine adhesion tissues and has antifibrotic potential. Here, to improve the long-term efficacy and activity of T6, we conducted the rational modified of T6 through studying structure-activity, and synthesized a series of analogues. Among them, S6 and S6-FA exhibited optimal antihepatic fibrosis activity, and S6-FA had a stronger long-acting effect than T6 and S6. The two analogues inhibited the expression of α-SMA and Collagen 1 in TGF-β-induced LX2 cells model and CCl4-induced mouse model of liver fibrosis. Besides, we discovered that S6 and S6-FA remarkably reduced the AST and ALT serum levels. Mechanistic studies have demonstrated that analogues inhibited liver fibrosis through inhibiting Erk, Smad and P65 pathways. This study provided that the novel peptide S6 and S6-FA is potential candidate compounds for treating liver fibrosis.
Collapse
Affiliation(s)
- Mingmin Li
- College
of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Liang Qi
- State
Key Laboratory of Anti-Infective Drug Discovery and Development, School
of Pharmaceutical Sciences, Sun Yat-sen
University, Guangzhou, Guangdong 510006, China
| | - Jin Huang
- Guangzhou
Dorsay Biotechnology Co., Ltd, Guangzhou, Guangdong 510006, China
| | - Haonan Li
- State
Key Laboratory of Anti-Infective Drug Discovery and Development, School
of Pharmaceutical Sciences, Sun Yat-sen
University, Guangzhou, Guangdong 510006, China
| | - Wei Cheng
- State
Key Laboratory of Anti-Infective Drug Discovery and Development, School
of Pharmaceutical Sciences, Sun Yat-sen
University, Guangzhou, Guangdong 510006, China
| | - Zihan Shi
- State
Key Laboratory of Anti-Infective Drug Discovery and Development, School
of Pharmaceutical Sciences, Sun Yat-sen
University, Guangzhou, Guangdong 510006, China
| | - Xianxing Jiang
- State
Key Laboratory of Anti-Infective Drug Discovery and Development, School
of Pharmaceutical Sciences, Sun Yat-sen
University, Guangzhou, Guangdong 510006, China
| | - Yifeng Zhou
- College
of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Wanxiang Jiang
- Sichuan
Greentech Bioscience Co., Ltd, Meishan, Sichuan 620031, China
| |
Collapse
|
3
|
Hassan MN, Hussain M, Khan RH. Strategies for inhibiting amyloid fibrillation: Current status and future prospects. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 211:145-168. [PMID: 39947747 DOI: 10.1016/bs.pmbts.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
One of the hallmarks of multiple neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases, is deposition of insoluble amyloid fibrils, which are toxic proteinaceous structures containing cross β-sheets. Several inhibitory strategies have been devised by researchers to impede or slow down the generation of such toxic species. Small compounds, peptides, and antibodies have been studied as possible inhibitors to interfere with key steps in amyloid production. Furthermore, adjusting environmental variables, such as temperature and pH have been known to impact the amyloid fibrillation process. Additionally, strategies are also available to reduce the possibility of protein misfolding so as to inhibit the subsequent development of fibrils, simply by stabilizing native protein conformations. It is very promising to develop targeted inhibitory therapies and comprehend the complexities of amyloid fibrillation in order to develop effective therapeutics to slow the progression of neurodegenerative disorders linked to misfolding and aggregation of proteins.
Collapse
Affiliation(s)
- Md Nadir Hassan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Murtaza Hussain
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| |
Collapse
|
4
|
Jeong K, Guo SC, Allaw S, Dinner AR. Analysis of the Dynamics of a Complex, Multipathway Reaction: Insulin Dimer Dissociation. J Phys Chem B 2024; 128:12728-12740. [PMID: 39670451 DOI: 10.1021/acs.jpcb.4c06933] [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: 12/14/2024]
Abstract
The protein hormone insulin forms a homodimer that must dissociate to bind to its receptor. Understanding the kinetics and mechanism of dissociation is essential for the rational design of therapeutic analogs. In addition to its physiological importance, this dissociation process serves as a paradigm for coupled (un)folding and (un)binding. Based on previous free energy simulations, insulin dissociation is thought to involve multiple pathways with comparable free energy barriers. Here, we analyze the mechanism of insulin dimer dissociation using a recently developed computational framework for estimating kinetic statistics from short-trajectory data. These statistics indicate that the likelihood of dissociation (the committor) closely tracks the decrease in the number of (native and nonnative) intermonomer contacts and the increase in the number of water contacts at the dimer interface; the transition state with equal likelihood of association and dissociation corresponds to an encounter complex with relatively few native contacts and many nonnative contacts. We identify four pathways out of the dimer state and quantify their contributions to the rate as well as their exchange by computing reactive fluxes. We show that both the pathways and their extents of exchange can be understood in terms of rotations around three axes of the dimer structure. Our results provide insights into the kinetics of insulin analogs and, more generally, how to characterize complex, multipathway processes.
Collapse
Affiliation(s)
- Kwanghoon Jeong
- Department of Chemistry, the University of Chicago, Chicago, Illinois 60637, United States
| | - Spencer C Guo
- Department of Chemistry, the University of Chicago, Chicago, Illinois 60637, United States
| | - Sammy Allaw
- Department of Chemistry, the University of Chicago, Chicago, Illinois 60637, United States
| | - Aaron R Dinner
- Department of Chemistry, the University of Chicago, Chicago, Illinois 60637, United States
- James Franck Institute, the University of Chicago, Chicago, Illinois 60637, United States
- Institute for Biophysical Dynamics, the University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
5
|
Wang S, Rienstra CM, Chen K. Higher Order Structure Differences Among Insulin Crystalline Drugs Revealed by 2D heteronuclear NMR. ChemMedChem 2024; 19:e202400340. [PMID: 39116305 DOI: 10.1002/cmdc.202400340] [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: 05/03/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/10/2024]
Abstract
During therapeutic protein development, two-dimensional (2D) heteronuclear NMR spectra can be a powerful analytical method for measuring protein higher order structure (HOS) in solution since the spectra exhibit much higher resolution than homonuclear 1H spectra. However, 2D NMR capabilities for characterizing protein HOS in crystalline states remain to be assessed, given the low 13C natural abundance and intrinsically broader lines in solid-state NMR (SSNMR). Herein, high-resolution heteronuclear correlation (HETCOR) SSNMR was utilized to directly measure intact crystal drug products of insulin human, insulin analogs of insulin lispro and insulin aspart. The fingerprint regions in 2D 1H-13C HETCOR spectra were identified, which distinguished the insulin crystals in their primary structure, HOS heterogeneity and dynamics, as well as the manufacturing processes. The HOS heterogeneity in insulin analogs is consistent with their therapeutic effect of rapid action; while insulin human crystals showed more structural homogeneity, consistent with their slower pharmacokinetics (PK) peak time than insulin analogs. Therefore, heteronuclear NMR could be broadly applicable to study protein drug dosage forms from liquid to solid, yielding improved molecular level structure data for assessing drug HOS in biosimilar drug development.
Collapse
Affiliation(s)
- Songlin Wang
- National Magnetic Resonance Facility at Madison (NMRFAM), University of Wisconsin-Madison, Madison, WI-53706, United States
| | - Chad M Rienstra
- National Magnetic Resonance Facility at Madison (NMRFAM), University of Wisconsin-Madison, Madison, WI-53706, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI-53706, United States
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI-53706, United States
| | - Kang Chen
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD-20993, United States
| |
Collapse
|
6
|
Jeong K, Guo SC, Allaw S, Dinner AR. Analysis of the dynamics of a complex, multipathway reaction: Insulin dimer dissociation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617297. [PMID: 39416150 PMCID: PMC11482781 DOI: 10.1101/2024.10.08.617297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The protein hormone insulin forms a homodimer that must dissociate to bind to its receptor. Understanding the kinetics and mechanism of dissociation is essential for rational design of therapeutic analogs. In addition to its physiological importance, this dissociation process serves as a paradigm for coupled (un)folding and (un)binding. Based on previous free energy simulations, insulin dissociation is thought to involve multiple pathways with comparable free energy barriers. Here, we analyze the mechanism of insulin dimer dissociation using a recently developed computational framework for estimating kinetic statistics from short-trajectory data. These statistics indicate that the likelihood of dissociation (the committor) closely tracks the decrease in the number of (native and nonnative) intermonomer contacts and the increase in the number of water contacts at the dimer interface; the transition state with equal likelihood of association and dissociation corresponds to an encounter complex with relatively few native contacts and many nonnative contacts. We identify four pathways out of the dimer state and quantify their contributions to the rate, as well as their exchange, by computing reactive fluxes. We show that both the pathways and their extents of exchange can be understood in terms of rotations around three axes of the dimer structure. Our results provide insights into the kinetics of insulin analogues and, more generally, how to characterize complex, multipathway processes.
Collapse
|
7
|
Breunig SL, Chapman AM, LeBon J, Quijano JC, Ranasinghe M, Rawson J, Demeler B, Ku HT, Tirrell DA. 4S-fluorination of ProB29 in insulin lispro slows fibril formation. J Biol Chem 2024; 300:107332. [PMID: 38703998 PMCID: PMC11154709 DOI: 10.1016/j.jbc.2024.107332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/06/2024] Open
Abstract
Recombinant insulin is a life-saving therapeutic for millions of patients affected by diabetes mellitus. Standard mutagenesis has led to insulin variants with improved control of blood glucose; for instance, the fast-acting insulin lispro contains two point mutations that suppress dimer formation and expedite absorption. However, insulins undergo irreversible denaturation, a process accelerated for the insulin monomer. Here we replace ProB29 of insulin lispro with 4R-fluoroproline, 4S-fluoroproline, and 4,4-difluoroproline. All three fluorinated lispro variants reduce blood glucose in diabetic mice, exhibit similar secondary structure as measured by CD, and rapidly dissociate from the zinc- and resorcinol-bound hexamer upon dilution. Notably, however, we find that 4S-fluorination of ProB29 delays the formation of undesired insulin fibrils that can accumulate at the injection site in vivo and can complicate insulin production and storage. These results demonstrate how subtle molecular changes achieved through non-canonical amino acid mutagenesis can improve the stability of protein therapeutics.
Collapse
Affiliation(s)
- Stephanie L Breunig
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Alex M Chapman
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Jeanne LeBon
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope, Duarte, California, USA
| | - Janine C Quijano
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope, Duarte, California, USA
| | - Maduni Ranasinghe
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope, Duarte, California, USA
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada; Department of Chemistry and Biochemistry, University of Montana, Missoula, Montana, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope, Duarte, California, USA; Irell & Manella Graduate School of Biological Science, City of Hope, Duarte, California, USA
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, USA.
| |
Collapse
|
8
|
Chikunova A, Manley MP, Heijjer CN, Drenth CS, Cramer-Blok AJ, Ahmad MUD, Perrakis A, Ubbink M. Conserved proline residues prevent dimerization and aggregation in the β-lactamase BlaC. Protein Sci 2024; 33:e4972. [PMID: 38533527 DOI: 10.1002/pro.4972] [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: 12/08/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
Evolution leads to conservation of amino acid residues in protein families. Conserved proline residues are usually considered to ensure the correct folding and to stabilize the three-dimensional structure. Surprisingly, proline residues that are highly conserved in class A β-lactamases were found to tolerate various substitutions without large losses in enzyme activity. We investigated the roles of three conserved prolines at positions 107, 226, and 258 in the β-lactamase BlaC from Mycobacterium tuberculosis and found that mutations can lead to dimerization of the enzyme and an overall less stable protein that is prone to aggregate over time. For the variant Pro107Thr, the crystal structure shows dimer formation resembling domain swapping. It is concluded that the proline substitutions loosen the structure, enhancing multimerization. Even though the enzyme does not lose its properties without the conserved proline residues, the prolines ensure the long-term structural integrity of the enzyme.
Collapse
Affiliation(s)
- A Chikunova
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - M P Manley
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - C N Heijjer
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - C S Drenth
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - A J Cramer-Blok
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - M Ud Din Ahmad
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Perrakis
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Ubbink
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Department of Infectious Diseases, Imperial College, London, UK
- Zocdoc, New York City, New York, USA
- ZoBio BV, Leiden, The Netherlands
| |
Collapse
|
9
|
Weil-Ktorza O, Dhayalan B, Chen YS, Weiss MA, Metanis N. Se-Glargine: Chemical Synthesis of a Basal Insulin Analogue Stabilized by an Internal Diselenide Bridge. Chembiochem 2024; 25:e202300818. [PMID: 38149322 DOI: 10.1002/cbic.202300818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 12/28/2023]
Abstract
Insulin has long provided a model for studies of protein folding and stability, enabling enhanced treatment of diabetes mellitus via analogue design. We describe the chemical synthesis of a basal insulin analogue stabilized by substitution of an internal cystine (A6-A11) by a diselenide bridge. The studies focused on insulin glargine (formulated as Lantus® and Toujeo®; Sanofi). Prepared at pH 4 in the presence of zinc ions, glargine exhibits a shifted isoelectric point due to a basic B chain extension (ArgB31 -ArgB32 ). Subcutaneous injection leads to pH-dependent precipitation of a long-lived depot. Pairwise substitution of CysA6 and CysA11 by selenocysteine was effected by solid-phase peptide synthesis; the modified A chain also contained substitution of AsnA21 by Gly, circumventing acid-catalyzed deamidation. Although chain combination of native glargine yielded negligible product, in accordance with previous synthetic studies, the pairwise selenocysteine substitution partially rescued this reaction: substantial product was obtained through repeated combination, yielding a stabilized insulin analogue. This strategy thus exploited both (a) the unique redox properties of selenocysteine in protein folding and (b) favorable packing of an internal diselenide bridge in the native state, once achieved. Such rational optimization of protein folding and stability may be generalizable to diverse disulfide-stabilized proteins of therapeutic interest.
Collapse
Affiliation(s)
- Orit Weil-Ktorza
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Norman Metanis
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
- Casali Center for Applied Chemistry, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| |
Collapse
|
10
|
Breunig S, Quijano JC, Donohue C, Henrickson A, Demeler B, Ku HT, Tirrell DA. Incorporation of Aliphatic Proline Residues into Recombinantly Produced Insulin. ACS Chem Biol 2023; 18:2574-2581. [PMID: 37960878 PMCID: PMC10728891 DOI: 10.1021/acschembio.3c00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023]
Abstract
Analogs of proline can be used to expand the chemical space about the residue while maintaining its uniquely restricted conformational space. Here, we demonstrate the incorporation of 4R-methylproline, 4S-methylproline, and 4-methyleneproline into recombinant insulin expressed in Escherichia coli. These modified proline residues, introduced at position B28, change the biophysical properties of insulin: Incorporation of 4-methyleneproline at B28 accelerates fibril formation, while 4-methylation speeds dissociation from the pharmaceutically formulated hexamer. This work expands the scope of proline analogs amenable to incorporation into recombinant proteins and demonstrates how noncanonical amino acid mutagenesis can be used to engineer the therapeutically relevant properties of protein drugs.
Collapse
Affiliation(s)
- Stephanie
L. Breunig
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Janine C. Quijano
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
| | - Cecile Donohue
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
| | - Amy Henrickson
- Department
of Chemistry and Biochemistry, University
of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Borries Demeler
- Department
of Chemistry and Biochemistry, University
of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
- Department
of Chemistry and Biochemistry, University
of Montana, Missoula, Montana 59801, United States
| | - Hsun Teresa Ku
- Department
of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Institute City
of Hope, Duarte, California 91010, United States
- Irell &
Manella Graduate School of Biological Science, City of Hope, Duarte, California 91010, United
States
| | - David A. Tirrell
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
11
|
Bohr F, Bohr SSR, Mishra NK, González-Foutel NS, Pinholt HD, Wu S, Nielsen EM, Zhang M, Kjaergaard M, Jensen KJ, Hatzakis NS. Enhanced hexamerization of insulin via assembly pathway rerouting revealed by single particle studies. Commun Biol 2023; 6:178. [PMID: 36792809 PMCID: PMC9932072 DOI: 10.1038/s42003-022-04386-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/20/2022] [Indexed: 02/17/2023] Open
Abstract
Insulin formulations with diverse oligomerization states are the hallmark of interventions for the treatment of diabetes. Here using single-molecule recordings we firstly reveal that insulin oligomerization can operate via monomeric additions and secondly quantify the existence, abundance and kinetic characterization of diverse insulin assembly and disassembly pathways involving addition of monomeric, dimeric or tetrameric insulin species. We propose and experimentally validate a model where the insulin self-assembly pathway is rerouted, favoring monomeric or oligomeric assembly, by solution concentration, additives and formulations. Combining our practically complete kinetic characterization with rate simulations, we calculate the abundance of each oligomeric species from nM to mM offering mechanistic insights and the relative abundance of all oligomeric forms at concentrations relevant both for secreted and administrated insulin. These reveal a high abundance of all oligomers and a significant fraction of hexamer resulting in practically halved bioavailable monomer concentration. In addition to providing fundamental new insights, the results and toolbox presented here can be universally applied, contributing to the development of optimal insulin formulations and the deciphering of oligomerization mechanisms for additional proteins.
Collapse
Affiliation(s)
- Freja Bohr
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren S-R Bohr
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Narendra Kumar Mishra
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
| | - Nicolás Sebastian González-Foutel
- Department of Molecular Biology and Genetics, The Danish Research Institute for Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, and Center for Proteins in Memory PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Henrik Dahl Pinholt
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Physics Department, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shunliang Wu
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Milan Nielsen
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Min Zhang
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, The Danish Research Institute for Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, and Center for Proteins in Memory PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Knud J Jensen
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark.
| | - Nikos S Hatzakis
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Aggarwal S, Tanwar N, Singh A, Munde M. Formation of Protamine and Zn-Insulin Assembly: Exploring Biophysical Consequences. ACS OMEGA 2022; 7:41044-41057. [PMID: 36406544 PMCID: PMC9670714 DOI: 10.1021/acsomega.2c04419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
The insulin-protamine interaction is at the core of the mode of action in many insulin formulations (Zn + insulin + protamine) and to treat diabetes, in which protamine is added to the stable form of hexameric insulin (Zn-insulin). However, due to the unavailability of quantitative data and a high-resolution structure, the binding mechanism of the insulin-protamine complex remains unknown. In this study, it was observed that Zn-insulin experiences destabilization as observed by the loss of secondary structure in circular dichroism (CD), and reduction in thermal stability in melting study, upon protamine binding. In isothermal titration calorimetry (ITC), it was found that the interactions were mostly enthalpically driven. This is in line with the positive ΔC m value (+880 cal mol-1), indicating the role of hydrophilic interactions in the complex formation, with the exposure of hydrophobic residues to the solvent, which was firmly supported by the 8-anilino-1-naphthalene sulfonate (ANS) binding study. The stoichiometry (N) value in ITC suggests the multiple insulin molecules binding to the protamine chain, which is consistent with the picture of the condensation of insulin in the presence of protamine. Atomic force microscopy (AFM) suggested the formation of a heterogeneous Zn-insulin-protamine complex. In fluorescence, Zn-insulin experiences strong Tyr quenching, suggesting that the location of the protamine-binding site is near Tyr, which is also supported by the molecular docking study. Since Tyr is critical in the stabilization of insulin self-assembly, its interaction with protamine may impair insulin's self-association ability and thermodynamic stability while at the same time promoting its flexible conformation desired for better biological activity.
Collapse
|
13
|
Maikawa CL, Nguyen LT, Mann JL, Appel EA. Formulation Excipients and Their Role in Insulin Stability and Association State in Formulation. Pharm Res 2022; 39:2721-2728. [PMID: 35978148 PMCID: PMC9633423 DOI: 10.1007/s11095-022-03367-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
While excipients are often overlooked as the "inactive" ingredients in pharmaceutical formulations, they often play a critical role in protein stability and absorption kinetics. Recent work has identified an ultrafast absorbing insulin formulation that is the result of excipient modifications. Specifically, the insulin monomer can be isolated by replacing zinc and the phenolic preservative metacresol with phenoxyethanol as an antimicrobial agent and an amphiphilic acrylamide copolymer excipient for stability. A greater understanding is needed of the interplay between excipients, insulin association state, and stability in order to optimize this formulation. Here, we formulated insulin with different preservatives and stabilizing excipient concentrations using both insulin lispro and regular human insulin and assessed the insulin association states using analytical ultracentrifugation as well as formulation stability. We determined that phenoxyethanol is required to eliminate hexamers and promote a high monomer content even in a zinc-free lispro formulation. There is also a concentration dependent relationship between the concentration of polyacrylamide-based copolymer excipient and insulin stability, where a concentration greater than 0.1 g/mL copolymer is required for a mostly monomeric zinc-free lispro formulation to achieve stability exceeding that of Humalog in a stressed aging assay. Further, we determined that under the formulation conditions tested zinc-free regular human insulin remains primarily hexameric and is not at this time a promising candidate for rapid-acting formulations.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, 94305, USA
| | - Leslee T Nguyen
- Department of Biochemistry, Stanford University, Stanford, 94305, USA
| | - Joseph L Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, 94305, USA. .,Department of Materials Science & Engineering, Stanford University, Stanford, 94305, USA. .,Department of Pediatrics (Endocrinology), Stanford University, Stanford, 94305, USA. .,ChEM-H Institute, Stanford University, Stanford, CA, 94305, USA. .,Woods Institute for the Environment, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Khadria A, Paavola CD, Maslov K, Valenzuela FA, Sperry AE, Cox AL, Cao R, Shi J, Brown-Augsburger PL, Lozano E, Blankenship RL, Majumdar R, Bradley SA, Beals JM, Oladipupo SS, Wang LV. Photoacoustic imaging reveals mechanisms of rapid-acting insulin formulations dynamics at the injection site. Mol Metab 2022; 62:101522. [PMID: 35671972 PMCID: PMC9207296 DOI: 10.1016/j.molmet.2022.101522] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Ultra-rapid insulin formulations control postprandial hyperglycemia; however, inadequate understanding of injection site absorption mechanisms is limiting further advancement. We used photoacoustic imaging to investigate the injection site dynamics of dye-labeled insulin lispro in the Humalog® and Lyumjev® formulations using the murine ear cutaneous model and correlated it with results from unlabeled insulin lispro in pig subcutaneous injection model. METHODS We employed dual-wavelength optical-resolution photoacoustic microscopy to study the absorption and diffusion of the near-infrared dye-labeled insulin lispro in the Humalog and Lyumjev formulations in mouse ears. We mathematically modeled the experimental data to calculate the absorption rate constants and diffusion coefficients. We studied the pharmacokinetics of the unlabeled insulin lispro in both the Humalog and Lyumjev formulations as well as a formulation lacking both the zinc and phenolic preservative in pigs. The association state of insulin lispro in each of the formulations was characterized using SV-AUC and NMR spectroscopy. RESULTS Through experiments using murine and swine models, we show that the hexamer dissociation rate of insulin lispro is not the absorption rate-limiting step. We demonstrated that the excipients in the Lyumjev formulation produce local tissue expansion and speed both insulin diffusion and microvascular absorption. We also show that the diffusion of insulin lispro at the injection site drives its initial absorption; however, the rate at which the insulin lispro crosses the blood vessels is its overall absorption rate-limiting step. CONCLUSIONS This study provides insights into injection site dynamics of insulin lispro and the impact of formulation excipients. It also demonstrates photoacoustic microscopy as a promising tool for studying protein therapeutics. The results from this study address critical questions around the subcutaneous behavior of insulin lispro and the formulation excipients, which could be useful to make faster and better controlled insulin formulations in the future.
Collapse
Affiliation(s)
- Anjul Khadria
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Chad D Paavola
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Konstantin Maslov
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Francisco A Valenzuela
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrea E Sperry
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Amy L Cox
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Rui Cao
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Junhui Shi
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Emmanuel Lozano
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ross L Blankenship
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ranajoy Majumdar
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Scott A Bradley
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - John M Beals
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Biotechnology Center, San Diego, CA, 92121, USA.
| | - Sunday S Oladipupo
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA; Caltech Optical Imaging Laboratory, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
15
|
Xiong X, Blakely A, Kim JH, Menting JG, Schäfer IB, Schubert HL, Agrawal R, Gutmann T, Delaine C, Zhang YW, Artik GO, Merriman A, Eckert D, Lawrence MC, Coskun Ü, Fisher SJ, Forbes BE, Safavi-Hemami H, Hill CP, Chou DHC. Symmetric and asymmetric receptor conformation continuum induced by a new insulin. Nat Chem Biol 2022; 18:511-519. [PMID: 35289328 PMCID: PMC9248236 DOI: 10.1038/s41589-022-00981-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/24/2022] [Indexed: 02/02/2023]
Abstract
Cone snail venoms contain a wide variety of bioactive peptides, including insulin-like molecules with distinct structural features, binding modes and biochemical properties. Here, we report an active humanized cone snail venom insulin with an elongated A chain and a truncated B chain, and use cryo-electron microscopy (cryo-EM) and protein engineering to elucidate its interactions with the human insulin receptor (IR) ectodomain. We reveal how an extended A chain can compensate for deletion of B-chain residues, which are essential for activity of human insulin but also compromise therapeutic utility by delaying dissolution from the site of subcutaneous injection. This finding suggests approaches to developing improved therapeutic insulins. Curiously, the receptor displays a continuum of conformations from the symmetric state to a highly asymmetric low-abundance structure that displays coordination of a single humanized venom insulin using elements from both of the previously characterized site 1 and site 2 interactions.
Collapse
Affiliation(s)
- Xiaochun Xiong
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA.,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Alan Blakely
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Jin Hwan Kim
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - John G Menting
- WEHI, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Ingmar B Schäfer
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Heidi L Schubert
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Rahul Agrawal
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Theresia Gutmann
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Carlie Delaine
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Yi Wolf Zhang
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA.,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Gizem Olay Artik
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Membrane Biochemistry and Lipid Research, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Allanah Merriman
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Debbie Eckert
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Michael C Lawrence
- WEHI, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Ünal Coskun
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Membrane Biochemistry and Lipid Research, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Simon J Fisher
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.,Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Briony E Forbes
- Discipline of Medical Biochemistry and Cell Biology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Helena Safavi-Hemami
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA. .,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Christopher P Hill
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| | - Danny Hung-Chieh Chou
- Department of Pediatrics, Division of Endocrinology and Diabetes, Stanford University, Stanford, CA, USA. .,Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
16
|
Mukherjee S, Acharya S, Mondal S, Banerjee P, Bagchi B. Structural Stability of Insulin Oligomers and Protein Association-Dissociation Processes: Free Energy Landscape and Universal Role of Water. J Phys Chem B 2021; 125:11793-11811. [PMID: 34674526 DOI: 10.1021/acs.jpcb.1c05811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Association and dissociation of proteins are important biochemical events. In this Feature Article, we analyze the available studies of these processes for insulin oligomers in aqueous solution. We focus on the solvation of the insulin monomer in water, stability and dissociation of its dimer, and structural integrity of the hexamer. The intricate role of water in solvation of the dimer- and hexamer-forming surfaces, in long-range interactions between the monomers and the stability of the oligomers, is discussed. Ten water molecules inside the central cavity stabilize the structure of the insulin hexamer. We discuss how different order parameters can be used to understand the dissociation of the insulin dimer. The calculation of the rate using a recently computed multidimensional free energy provides considerable insight into the interplay between protein and water dynamics.
Collapse
Affiliation(s)
- Saumyak Mukherjee
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
| | - Subhajit Acharya
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
| | - Sayantan Mondal
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
| | - Puja Banerjee
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
| | - Biman Bagchi
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
17
|
Jarosinski MA, Dhayalan B, Chen YS, Chatterjee D, Varas N, Weiss MA. Structural principles of insulin formulation and analog design: A century of innovation. Mol Metab 2021; 52:101325. [PMID: 34428558 PMCID: PMC8513154 DOI: 10.1016/j.molmet.2021.101325] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The discovery of insulin in 1921 and its near-immediate clinical use initiated a century of innovation. Advances extended across a broad front, from the stabilization of animal insulin formulations to the frontiers of synthetic peptide chemistry, and in turn, from the advent of recombinant DNA manufacturing to structure-based protein analog design. In each case, a creative interplay was observed between pharmaceutical applications and then-emerging principles of protein science; indeed, translational objectives contributed to a growing molecular understanding of protein structure, aggregation and misfolding. SCOPE OF REVIEW Pioneering crystallographic analyses-beginning with Hodgkin's solving of the 2-Zn insulin hexamer-elucidated general features of protein self-assembly, including zinc coordination and the allosteric transmission of conformational change. Crystallization of insulin was exploited both as a step in manufacturing and as a means of obtaining protracted action. Forty years ago, the confluence of recombinant human insulin with techniques for site-directed mutagenesis initiated the present era of insulin analogs. Variant or modified insulins were developed that exhibit improved prandial or basal pharmacokinetic (PK) properties. Encouraged by clinical trials demonstrating the long-term importance of glycemic control, regimens based on such analogs sought to resemble daily patterns of endogenous β-cell secretion more closely, ideally with reduced risk of hypoglycemia. MAJOR CONCLUSIONS Next-generation insulin analog design seeks to explore new frontiers, including glucose-responsive insulins, organ-selective analogs and biased agonists tailored to address yet-unmet clinical needs. In the coming decade, we envision ever more powerful scientific synergies at the interface of structural biology, molecular physiology and therapeutics.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Nicolás Varas
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA; Department of Chemistry, Indiana University, Bloomington, 47405, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, 47907, IN, USA.
| |
Collapse
|
18
|
Jensen GV, Rosenmejer KR, Huda P, Arleth L. Oligomerization of Pharmaceutically Relevant Insulin Analogues for Varying Concentration and Salinity Revealed by Small-Angle X-ray Scattering. Mol Pharm 2021; 18:3272-3280. [PMID: 34351780 DOI: 10.1021/acs.molpharmaceut.1c00164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two different insulin analogues, insulin degludec and lithocholyl insulin, were studied by small-angle X-ray scattering with respect to their self-assembly and interactions in solution at different concentrations of insulin and salt, NaCl. Very different behavior was observed for the two. Insulin degludec, linked to a hexadecanedioic acid, consistently formed di-hexamers, without any further oligomeric growth upon screening of electrostatic repulsions, indicating a stable di-hexamer unit without further oligomerization, as expected in the presence of phenol. The other insulin analogue, linked to the sterol lithocholic acid, formed n-hexamers with n ranging from 1 to 15, increasing with NaCl concentration and insulin concentration, indicating attractive forces in competition with the electrostatic repulsion and solution entropy. At the highest concentration of insulin and NaCl, a liquid crystal phase was observed, which has not previously been identified, featuring a quadratic structure organized into layers, which might hold interesting properties for pharmaceutical applications.
Collapse
Affiliation(s)
- Grethe V Jensen
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Katrine R Rosenmejer
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Pie Huda
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Lise Arleth
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| |
Collapse
|
19
|
Salahuddin P, Khan RH, Furkan M, Uversky VN, Islam Z, Fatima MT. Mechanisms of amyloid proteins aggregation and their inhibition by antibodies, small molecule inhibitors, nano-particles and nano-bodies. Int J Biol Macromol 2021; 186:580-590. [PMID: 34271045 DOI: 10.1016/j.ijbiomac.2021.07.056] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
Protein misfolding and aggregation can be induced by a wide variety of factors, such as dominant disease-associated mutations, changes in the environmental conditions (pH, temperature, ionic strength, protein concentration, exposure to transition metal ions, exposure to toxins, posttranslational modifications including glycation, phosphorylation, and sulfation). Misfolded intermediates interact with similar intermediates and progressively form dimers, oligomers, protofibrils, and fibrils. In amyloidoses, fibrillar aggregates are deposited in the tissues either as intracellular inclusion or extracellular plaques (amyloid). When such proteinaceous deposit occurs in the neuronal cells, it initiates degeneration of neurons and consequently resulting in the manifestation of various neurodegenerative diseases. Several different types of molecules have been designed and tested both in vitro and in vivo to evaluate their anti-amyloidogenic efficacies. For instance, the native structure of a protein associated with amyloidosis could be stabilized by ligands, antibodies could be used to remove plaques, oligomer-specific antibody A11 could be used to remove oligomers, or prefibrillar aggregates could be removed by affibodies. Keeping the above views in mind, in this review we have discussed protein misfolding and aggregation, mechanisms of protein aggregation, factors responsible for aggregations, and strategies for aggregation inhibition.
Collapse
Affiliation(s)
- Parveen Salahuddin
- DISC, Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India.
| | - Mohammad Furkan
- Interdisciplinary Biotechnology Unit, A.M.U., Aligarh 202002, India
| | - Vladimir N Uversky
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Institutskaya Str., 7, Pushchino, Moscow region 142290, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zeyaul Islam
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, P.O Box 5825, Doha, Qatar
| | - Munazza Tamkeen Fatima
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
20
|
Rege NK, Liu M, Yang Y, Dhayalan B, Wickramasinghe NP, Chen YS, Rahimi L, Guo H, Haataja L, Sun J, Ismail-Beigi F, Phillips NB, Arvan P, Weiss MA. Evolution of insulin at the edge of foldability and its medical implications. Proc Natl Acad Sci U S A 2020; 117:29618-29628. [PMID: 33154160 PMCID: PMC7703552 DOI: 10.1073/pnas.2010908117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Proteins have evolved to be foldable, and yet determinants of foldability may be inapparent once the native state is reached. Insight has emerged from studies of diseases of protein misfolding, exemplified by monogenic diabetes mellitus due to mutations in proinsulin leading to endoplasmic reticulum stress and β-cell death. Cellular foldability of human proinsulin requires an invariant Phe within a conserved crevice at the receptor-binding surface (position B24). Any substitution, even related aromatic residue TyrB24, impairs insulin biosynthesis and secretion. As a seeming paradox, a monomeric TyrB24 insulin analog exhibits a native-like structure in solution with only a modest decrement in stability. Packing of TyrB24 is similar to that of PheB24, adjoining core cystine B19-A20 to seal the core; the analog also exhibits native self-assembly. Although affinity for the insulin receptor is decreased ∼20-fold, biological activities in cells and rats were within the range of natural variation. Together, our findings suggest that the invariance of PheB24 among vertebrate insulins and insulin-like growth factors reflects an essential role in enabling efficient protein folding, trafficking, and secretion, a function that is inapparent in native structures. In particular, we envision that the para-hydroxyl group of TyrB24 hinders pairing of cystine B19-A20 in an obligatory on-pathway folding intermediate. The absence of genetic variation at B24 and other conserved sites near this disulfide bridge-excluded due to β-cell dysfunction-suggests that insulin has evolved to the edge of foldability. Nonrobustness of a protein's fitness landscape underlies both a rare monogenic syndrome and "diabesity" as a pandemic disease of civilization.
Collapse
Affiliation(s)
- Nischay K Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 300052 Tianjin, China
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Yanwu Yang
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Balamurugan Dhayalan
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Yen-Shan Chen
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Leili Rahimi
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Huan Guo
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Jinhong Sun
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106;
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
21
|
Pitt JP, McCarthy OM, Hoeg-Jensen T, Wellman BM, Bracken RM. Factors Influencing Insulin Absorption Around Exercise in Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:573275. [PMID: 33193089 PMCID: PMC7609903 DOI: 10.3389/fendo.2020.573275] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022] Open
Abstract
International charities and health care organizations advocate regular physical activity for health benefit in people with type 1 diabetes. Clinical expert and international diabetes organizations' position statements support the management of good glycemia during acute physical exercise by adjusting exogenous insulin and/or carbohydrate intake. Yet research has detailed, and patients frequently report, variable blood glucose responses following both the same physical exercise session and insulin to carbohydrate alteration. One important source of this variability is insulin delivery to the circulation. With modern insulin analogs, it is important to understand how different insulins, their delivery methods, and inherent physiological factors, influence the reproducibility of insulin absorption from the injection site into circulation. Furthermore, contrary to the adaptive pancreatic response to exercise in the person without diabetes, the physiological and metabolic shifts with exercise may increase circulating insulin concentrations that may contribute to exercise-related hyperinsulinemia and consequent hypoglycemia. Thus, a furthered understanding of factors underpinning insulin delivery may offer more confidence for healthcare professionals and patients when looking to improve management of glycemia around exercise.
Collapse
Affiliation(s)
- Jason P. Pitt
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Olivia M. McCarthy
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Thomas Hoeg-Jensen
- Diabetes Peptide and Protein Chemistry, Novo Nordisk A/S, Maaloev, Denmark
| | - Benjamin M. Wellman
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Richard M. Bracken
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| |
Collapse
|
22
|
Santra S, Jana M. Insights into the Sensitivity of Arginine Concentration to Preserve the Folded Form of Insulin Monomer under Thermal Stress. J Chem Inf Model 2020; 60:3105-3119. [PMID: 32479724 DOI: 10.1021/acs.jcim.0c00006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Arginine, although popularly known as aggregation suppressor additive, has been found to quench proteins' structure and function by destabilizing their conformations. Driven by such controversial evidence, in this work we performed a series of atomistic molecular dynamics simulations of insulin monomer, a biologically active hormone protein, in arginine solution of varying concentrations (0.5, 1, and 2 M) at ambient and elevated temperature (400 K) to explore the arginine concentration driven structure-based stability of the protein. Our study reveals that the flexibility of the protein's structure is dependent on the arginine concentration, and among all the used solutions, 2 M arginine, a "neutral crowder" that mimics the cellular environment, can preserve the native folded form of the protein at ambient temperature in an excellent manner. Further, while the protein unfolds at 400 K in pure water, this solution worked satisfactorily to preserve the protein's folded conformation more firmly than the other solutions. The replica-exchange MD of insulin in 2 M arginine solution further supports the fact. In this aspect an important issue in molecular pharmacology is to identify and recognize the physical origin of the stability of a protein, i.e, in this case, how arginine directs the conformational flexibility of the protein and preserves its native folded form. We identified that the exclusion of arginine from the protein surface increases the local structuration of water around the protein, thereby preserving its "biological water" layer, and makes the protein more hydrated at 2 M concentration as compared to the other arginine solutions. Additionally, our microscopic investigation on the interactions of the protein-solvation layer revealed that the structural heterogeneity of the protein surface, arising from the differential physicochemical nature of the amino acid residues, controls the favorable formation of sluggish water-arginine mixed solvation layer at higher arginine concentration that helps the protein to maintain its structural rigidity. Importantly, apart from the protein-solvent hydrogen-bonding interactions, the anion-pi interactions, established between the carboxyl group of arginine and the aromatic amino acid residues of insulin, were recognized to facilitate the protein to maintain its native folded form at the experimental temperatures.
Collapse
Affiliation(s)
- Santanu Santra
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela-769008, India
| | - Madhurima Jana
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela-769008, India
| |
Collapse
|
23
|
Hu Y, Arora J, Joshi SB, Esfandiary R, Middaugh CR, Weis DD, Volkin DB. Characterization of Excipient Effects on Reversible Self-Association, Backbone Flexibility, and Solution Properties of an IgG1 Monoclonal Antibody at High Concentrations: Part 1. J Pharm Sci 2020; 109:340-352. [DOI: 10.1016/j.xphs.2019.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/13/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022]
|
24
|
Desmond JL, Koner D, Meuwly M. Probing the Differential Dynamics of the Monomeric and Dimeric Insulin from Amide-I IR Spectroscopy. J Phys Chem B 2019; 123:6588-6598. [PMID: 31318551 DOI: 10.1021/acs.jpcb.9b04628] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The monomer-dimer equilibrium for insulin is one of the essential steps in forming the receptor-binding competent monomeric form of the hormone. Despite this importance, the thermodynamic stability, in particular for modified insulins, is quite poorly understood, in part, due to experimental difficulties. This work explores one- and two-dimensional infrared spectroscopy in the range of the amide-I band for the hydrated monomeric and dimeric wild-type hormone. It is found that for the monomer the frequency fluctuation correlation function (FFCF) and the one-dimensional infrared spectra are position sensitive. The spectra of the -CO probes at the dimerization interface (residues Phe24, Phe25, and Tyr26) split and indicate an asymmetry despite the overall (formal) point symmetry of the dimer structure. Also, the decay times of the FFCF for the same -CO probe in the monomer and the dimer can differ by up to 1 order of magnitude, for example, for residue PheB24, which is solvent exposed for the monomer but at the interface for the dimer. The spectroscopic shifts correlate approximately with the average number of hydration waters and the magnitude of the FFCF at time zero. However, this correlation is only qualitative due to the heterogeneous and highly dynamical environment. Based on density functional theory calculations, the dominant contribution for solvent-exposed -CO is found to arise from the surrounding water (∼75%), whereas the protein environment contributes considerably less. The results suggest that infrared spectroscopy is a positionally sensitive probe of insulin dimerization, in particular in conjunction with isotopic labeling of the probe.
Collapse
Affiliation(s)
- Jasmine L Desmond
- Department of Chemistry , University of Basel , Klingelbergstrasse 80 , 4056 Basel , Switzerland
| | - Debasish Koner
- Department of Chemistry , University of Basel , Klingelbergstrasse 80 , 4056 Basel , Switzerland
| | - Markus Meuwly
- Department of Chemistry , University of Basel , Klingelbergstrasse 80 , 4056 Basel , Switzerland
| |
Collapse
|
25
|
Protein misfolding, aggregation and mechanism of amyloid cytotoxicity: An overview and therapeutic strategies to inhibit aggregation. Int J Biol Macromol 2019; 134:1022-1037. [PMID: 31128177 DOI: 10.1016/j.ijbiomac.2019.05.109] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/18/2019] [Accepted: 05/18/2019] [Indexed: 12/18/2022]
Abstract
Protein and peptides are converted from their soluble forms into highly ordered fibrillar aggregates under various conditions inside the cell. Such transitions confer diverse neurodegenerative diseases including Alzheimer's disease, Huntington's disease Prion's disease, Parkinson's disease, polyQ and share abnormal folding of potentially cytotoxic protein species linked with degeneration and death of precise neuronal populations. Presently, major advances are made to understand and get detailed insight into the structural basis and mechanism of amyloid formation, cytotoxicity and therapeutic approaches to combat them. Here we highlight classifies and summarizes the detailed overview of protein misfolding and aggregation at their molecular level including the factors that promote protein aggregation under in vivo and in vitro conditions. In addition, we describe the recent technologies that aid the characterization of amyloid aggregates along with several models that might be responsible for amyloid induced cytotoxicity to cells. Overview on the inhibition of amyloidosis by targeting different small molecules (both natural and synthetic origin) have been also discussed, that provides important approaches to identify novel targets and develop specific therapeutic strategies to combat protein aggregation related neurodegenerative diseases.
Collapse
|
26
|
Zhang R, Zhang N, Mohri M, Wu L, Eckert T, Krylov VB, Antosova A, Ponikova S, Bednarikova Z, Markart P, Günther A, Norden B, Billeter M, Schauer R, Scheidig AJ, Ratha BN, Bhunia A, Hesse K, Enani MA, Steinmeyer J, Petridis AK, Kozar T, Gazova Z, Nifantiev NE, Siebert HC. Nanomedical Relevance of the Intermolecular Interaction Dynamics-Examples from Lysozymes and Insulins. ACS OMEGA 2019; 4:4206-4220. [PMID: 30847433 PMCID: PMC6398350 DOI: 10.1021/acsomega.8b02471] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/28/2018] [Indexed: 06/01/2023]
Abstract
Insulin and lysozyme share the common features of being prone to aggregate and having biomedical importance. Encapsulating lysozyme and insulin in micellar nanoparticles probably would prevent aggregation and facilitate oral drug delivery. Despite the vivid structural knowledge of lysozyme and insulin, the environment-dependent oligomerization (dimer, trimer, and multimer) and associated structural dynamics remain elusive. The knowledge of the intra- and intermolecular interaction profiles has cardinal importance for the design of encapsulation protocols. We have employed various biophysical methods such as NMR spectroscopy, X-ray crystallography, Thioflavin T fluorescence, and atomic force microscopy in conjugation with molecular modeling to improve the understanding of interaction dynamics during homo-oligomerization of lysozyme (human and hen egg) and insulin (porcine, human, and glargine). The results obtained depict the atomistic intra- and intermolecular interaction details of the homo-oligomerization and confirm the propensity to form fibrils. Taken together, the data accumulated and knowledge gained will further facilitate nanoparticle design and production with insulin or lysozyme-related protein encapsulation.
Collapse
Affiliation(s)
- Ruiyan Zhang
- Institute
of Biopharmaceutical Research, Liaocheng
University, Liaocheng 252059, P. R. China
- RI-B-NT
Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148 Kiel, Germany
- Institute
of Zoology, Department of Structural Biology, Christian-Albrechts-University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Ning Zhang
- Institute
of Biopharmaceutical Research, Liaocheng
University, Liaocheng 252059, P. R. China
| | - Marzieh Mohri
- RI-B-NT
Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148 Kiel, Germany
| | - Lisha Wu
- Department
of Chemical and Biological Engineering, Chalmers University of Technology, 41296 Gothenburg, Sweden
| | - Thomas Eckert
- Department
of Chemistry and Biology, University of
Applied Sciences Fresenius, Limburger Str. 2, 65510 Idstein, Germany
- Institut
für Veterinärphysiolgie und Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Gießen, Frankfurter Str. 100, 35392 Gießen, Germany
| | - Vadim B. Krylov
- Laboratory
of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Andrea Antosova
- Department
of Biophysics Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001 Kosice, Slovakia
| | - Slavomira Ponikova
- Department
of Biophysics Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001 Kosice, Slovakia
| | - Zuzana Bednarikova
- Department
of Biophysics Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001 Kosice, Slovakia
| | - Philipp Markart
- Medical
Clinic II, Justus-Liebig-University, Klinikstraße 33, 35392 Giessen, Germany
- Pneumology,
Heart-Thorax-Center Fulda, Pacelliallee 4, 36043 Fulda, Germany
| | - Andreas Günther
- Medical
Clinic II, Justus-Liebig-University, Klinikstraße 33, 35392 Giessen, Germany
| | - Bengt Norden
- Department
of Chemical and Biological Engineering, Chalmers University of Technology, 41296 Gothenburg, Sweden
| | - Martin Billeter
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 40530 Gothenburg, Sweden
| | - Roland Schauer
- Institute
of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Axel J. Scheidig
- Institute
of Zoology, Department of Structural Biology, Christian-Albrechts-University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Bhisma N. Ratha
- Biomolecular
NMR and Drug Design Laboratory, Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), 700054 Kolkata, India
| | - Anirban Bhunia
- Biomolecular
NMR and Drug Design Laboratory, Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), 700054 Kolkata, India
| | - Karsten Hesse
- Tierarztpraxis
Dr. Karsten Hesse, Rathausstraße
16, 35460 Stauffenberg, Germany
| | - Mushira Abdelaziz Enani
- Infectious
Diseases Division, Department of Medicine, King Fahad Medical City, P.O. Box 59046, 11525 Riyadh, Kingdom of Saudi
Arabia
| | - Jürgen Steinmeyer
- Laboratory
for Experimental Orthopaedics, Department of Orthopaedics, Justus-Liebig-University, Paul-Meimberg-Str. 3, D-35392 Giessen, Germany
| | - Athanasios K. Petridis
- Neurochirurgische
Klinik, Universität Düsseldorf, Geb. 11.54, Moorenstraße 5, 40255 Düsseldorf, Germany
| | - Tibor Kozar
- Center
for Interdisciplinary Biosciences, TIP-UPJS, Jesenna 5, 04001 Kosice, Slovakia
| | - Zuzana Gazova
- Department
of Biophysics Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001 Kosice, Slovakia
| | - Nikolay E. Nifantiev
- Laboratory
of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russian Federation
| | - Hans-Christian Siebert
- RI-B-NT
Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148 Kiel, Germany
| |
Collapse
|
27
|
Weiss MA, Lawrence MC. A thing of beauty: Structure and function of insulin's "aromatic triplet". Diabetes Obes Metab 2018; 20 Suppl 2:51-63. [PMID: 30230175 PMCID: PMC6159917 DOI: 10.1111/dom.13402] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/30/2022]
Abstract
The classical crystal structure of insulin was determined in 1969 by D.C. Hodgkin et al. following a 35-year program of research. This structure depicted a hexamer remarkable for its self-assembly as a zinc-coordinated trimer of dimer. Prominent at the dimer interface was an "aromatic triplet" of conserved residues at consecutive positions in the B chain: PheB24 , PheB25 and TyrB26 . The elegance of this interface inspired the Oxford team to poetry: "A thing of beauty is a joy forever" (John Keats as quoted by Blundell, T.L., et al. Advances in Protein Chemistry 26:279-286 [1972]). Here, we revisit this aromatic triplet in light of recent advances in the structural biology of insulin bound as a monomer to fragments of the insulin receptor. Such co-crystal structures have defined how these side chains pack at the primary hormone-binding surface of the receptor ectodomain. On receptor binding, the B-chain β-strand (residues B24-B28) containing the aromatic triplet detaches from the α-helical core of the hormone. Whereas TyrB26 lies at the periphery of the receptor interface and may functionally be replaced by a diverse set of substitutions, PheB24 and PheB25 engage invariant elements of receptor domains L1 and αCT. These critical contacts were anticipated by the discovery of diabetes-associated mutations at these positions by Donald Steiner et al. at the University of Chicago. Conservation of PheB24 , PheB25 and TyrB26 among vertebrate insulins reflects the striking confluence of structure-based evolutionary constraints: foldability, protective self-assembly and hormonal activity.
Collapse
Affiliation(s)
- Michael A. Weiss
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202 USA
| | - Michael C. Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, AUSTRALIA
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, AUSTRALIA
| |
Collapse
|
28
|
Rege NK, Wickramasinghe NP, Tustan AN, Phillips NFB, Yee VC, Ismail-Beigi F, Weiss MA. Structure-based stabilization of insulin as a therapeutic protein assembly via enhanced aromatic-aromatic interactions. J Biol Chem 2018; 293:10895-10910. [PMID: 29880646 PMCID: PMC6052209 DOI: 10.1074/jbc.ra118.003650] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
Key contributions to protein structure and stability are provided by weakly polar interactions, which arise from asymmetric electronic distributions within amino acids and peptide bonds. Of particular interest are aromatic side chains whose directional π-systems commonly stabilize protein interiors and interfaces. Here, we consider aromatic-aromatic interactions within a model protein assembly: the dimer interface of insulin. Semi-classical simulations of aromatic-aromatic interactions at this interface suggested that substitution of residue TyrB26 by Trp would preserve native structure while enhancing dimerization (and hence hexamer stability). The crystal structure of a [TrpB26]insulin analog (determined as a T3Rf3 zinc hexamer at a resolution of 2.25 Å) was observed to be essentially identical to that of WT insulin. Remarkably and yet in general accordance with theoretical expectations, spectroscopic studies demonstrated a 150-fold increase in the in vitro lifetime of the variant hexamer, a critical pharmacokinetic parameter influencing design of long-acting formulations. Functional studies in diabetic rats indeed revealed prolonged action following subcutaneous injection. The potency of the TrpB26-modified analog was equal to or greater than an unmodified control. Thus, exploiting a general quantum-chemical feature of protein structure and stability, our results exemplify a mechanism-based approach to the optimization of a therapeutic protein assembly.
Collapse
Affiliation(s)
| | | | - Alisar N Tustan
- Medicine, Case Western Reserve University, Cleveland, Ohio 44106 and
| | | | | | | | - Michael A Weiss
- From the Departments of Biochemistry and
- the Department of Biochemistry, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
29
|
Raghunathan S, El Hage K, Desmond JL, Zhang L, Meuwly M. The Role of Water in the Stability of Wild-type and Mutant Insulin Dimers. J Phys Chem B 2018; 122:7038-7048. [DOI: 10.1021/acs.jpcb.8b04448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Shampa Raghunathan
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Krystel El Hage
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Jasmine L. Desmond
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Lixian Zhang
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Markus Meuwly
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| |
Collapse
|
30
|
Candido R, Wyne K, Romoli E. A Review of Basal-Bolus Therapy Using Insulin Glargine and Insulin Lispro in the Management of Diabetes Mellitus. Diabetes Ther 2018; 9:927-949. [PMID: 29654514 PMCID: PMC5984925 DOI: 10.1007/s13300-018-0422-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Basal-bolus therapy (BBT) refers to the combination of a long-acting basal insulin with a rapid-acting insulin at mealtimes. Basal insulin glargine 100 U/mL and prandial insulin lispro have been available for many years and there is a substantial evidence base to support the efficacy and safety of these agents when they are used in BBT or basal-plus therapy for patients with type 1 or type 2 diabetes mellitus (T1DM, T2DM). With the growing availability of alternative insulins for use in such regimens, it seems timely to review the data regarding BBT with insulin glargine 100 U/mL and insulin lispro. In patients with T1DM, BBT with insulin glargine plus insulin lispro provides similar or better glycemic control and leads to less nocturnal hypoglycemia compared to BBT using human insulin as the basal and/or prandial component, and generally provides similar glycemic control and rates of severe hypoglycemia to those achieved with insulin lispro administered by continuous subcutaneous insulin infusion (CSII). Studies evaluating BBT with insulin glargine plus insulin lispro in patients with T2DM also demonstrate the efficacy and safety of these insulins. Available data suggest that BBT with insulin glargine and insulin lispro provides similar levels of efficacy and safety in pediatric and adult populations with T1DM and in adult patients and those aged more than 65 years with T2DM. These insulin preparations also appear to be safe and effective for controlling T2DM in people of different ethnicities and in patients with T1DM or T2DM and comorbidities. FUNDING Eli Lilly and Company.
Collapse
Affiliation(s)
- Riccardo Candido
- Diabetes Centre District 3, Azienda Sanitaria Universitaria Integrata di Trieste, Via Puccini 48/50, 34100, Trieste, Italy.
| | - Kathleen Wyne
- The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Ester Romoli
- Eli Lilly Italia SPA, via A. Gramsci 731/733, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
31
|
Sitkowski J, Bocian W, Bednarek E, Urbańczyk M, Koźmiński W, Borowicz P, Płucienniczak G, Łukasiewicz N, Sokołowska I, Kozerski L. Insight into human insulin aggregation revisited using NMR derived translational diffusion parameters. JOURNAL OF BIOMOLECULAR NMR 2018; 71:101-114. [PMID: 29948440 DOI: 10.1007/s10858-018-0197-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
The NMR derived translational diffusion coefficients were performed on unlabeled and uniformly labeled 13C,15N human insulin in water, both in neat, with zinc ions only, and in pharmaceutical formulation, containing only m-cresol as phenolic ligand, glycerol and zinc ions. The results show the dominant role of the pH parameter and the concentration on aggregation. The diffusion coefficient Dav was used for monitoring the overall average state of oligomeric ensemble in solution. The analysis of the experimental data of diffusion measurements, using the direct exponential curve resolution algorithm (DECRA) allows suggesting the two main components of the oligomeric ensemble. The 3D HSQC-iDOSY, (diffusion ordered HSQC) experiments performed on 13C, 15N-fully labeled insulin at the two pH values, 4 and 7.5, allow for the first time a more detailed experimental observation of individual components in the ensemble. The discussion involves earlier static and dynamic laser light scattering experiments and recent NMR derived translational diffusion results. The results bring new informations concerning the preparation of pharmaceutical formulation and in particular a role of Zn2+ ions. They also will enable better understanding and unifying the results of studies on insulin misfolding effects performed in solution by diverse physicochemical methods at different pH and concentration.
Collapse
Affiliation(s)
- Jerzy Sitkowski
- National Medicines Institute, Chełmska 30, 00-725, Warsaw, Poland
| | - Wojciech Bocian
- National Medicines Institute, Chełmska 30, 00-725, Warsaw, Poland
| | | | - Mateusz Urbańczyk
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089, Warsaw, Poland
| | - Wiktor Koźmiński
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089, Warsaw, Poland
| | - Piotr Borowicz
- Institute of Biotechnology and Antibiotics, Starościńska 5, 02-516, Warsaw, Poland
| | | | - Natalia Łukasiewicz
- Institute of Biotechnology and Antibiotics, Starościńska 5, 02-516, Warsaw, Poland
| | - Iwona Sokołowska
- Institute of Biotechnology and Antibiotics, Starościńska 5, 02-516, Warsaw, Poland
| | - Lech Kozerski
- National Medicines Institute, Chełmska 30, 00-725, Warsaw, Poland.
| |
Collapse
|
32
|
Adams GG, Meal A, Morgan PS, Alzahrani QE, Zobel H, Lithgo R, Kok MS, Besong DTM, Jiwani SI, Ballance S, Harding SE, Chayen N, Gillis RB. Characterisation of insulin analogues therapeutically available to patients. PLoS One 2018; 13:e0195010. [PMID: 29596514 PMCID: PMC5875863 DOI: 10.1371/journal.pone.0195010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
The structure and function of clinical dosage insulin and its analogues were assessed. This included ‘native insulins’ (human recombinant, bovine, porcine), ‘fast-acting analogues’ (aspart, glulisine, lispro) and ‘slow-acting analogues’ (glargine, detemir, degludec). Analytical ultracentrifugation, both sedimentation velocity and equilibrium experiments, were employed to yield distributions of both molar mass and sedimentation coefficient of all nine insulins. Size exclusion chromatography, coupled to multi-angle light scattering, was also used to explore the function of these analogues. On ultracentrifugation analysis, the insulins under investigation were found to be in numerous conformational states, however the majority of insulins were present in a primarily hexameric conformation. This was true for all native insulins and two fast-acting analogues. However, glargine was present as a dimer, detemir was a multi-hexameric system, degludec was a dodecamer (di-hexamer) and glulisine was present as a dimer-hexamer-dihexamer system. However, size-exclusion chromatography showed that the two hexameric fast-acting analogues (aspart and lispro) dissociated into monomers and dimers due to the lack of zinc in the mobile phase. This comprehensive study is the first time all nine insulins have been characterised in this way, the first time that insulin detemir have been studied using analytical ultracentrifugation and the first time that insulins aspart and glulisine have been studied using sedimentation equilibrium. The structure and function of these clinically administered insulins is of critical importance and this research adds novel data to an otherwise complex functional physiological protein.
Collapse
Affiliation(s)
- Gary G. Adams
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- * E-mail: (GGA); (RBG)
| | - Andrew Meal
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Paul S. Morgan
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Qushmua E. Alzahrani
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- Taif University, Faculty of Science, Taif, Saudi Arabia
| | | | - Ryan Lithgo
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | - M. Samil Kok
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- Department of Food Engineering, Abant Izzet Baysal University, Bolu, Turkey
| | - David T. M. Besong
- Functional Nanomaterials Lab, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Shahwar I. Jiwani
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | | | - Stephen E. Harding
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
| | - Naomi Chayen
- Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Richard B. Gillis
- Faculty of Medicine and Health Sciences, University of Nottingham, Queens Medical Centre, Nottingham, NG7 2UH, United Kingdom
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, School of Biosciences, Sutton Bonington, LE12 5RD, United Kingdom
- * E-mail: (GGA); (RBG)
| |
Collapse
|
33
|
El Hage K, Mondal P, Meuwly M. Free energy simulations for protein ligand binding and stability. MOLECULAR SIMULATION 2018. [DOI: 10.1080/08927022.2017.1416115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Krystel El Hage
- Department of Chemistry, University of Basel , Basel, Switzerland
| | - Padmabati Mondal
- Department of Chemistry, University of Basel , Basel, Switzerland
| | - Markus Meuwly
- Department of Chemistry, University of Basel , Basel, Switzerland
| |
Collapse
|
34
|
A mechanistic insight into protein-ligand interaction, folding, misfolding, aggregation and inhibition of protein aggregates: An overview. Int J Biol Macromol 2018; 106:1115-1129. [DOI: 10.1016/j.ijbiomac.2017.07.185] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 07/28/2017] [Accepted: 07/30/2017] [Indexed: 11/22/2022]
|
35
|
Selivanova OM, Grishin SY, Glyakina AV, Sadgyan AS, Ushakova NI, Galzitskaya OV. Analysis of Insulin Analogs and the Strategy of Their Further Development. BIOCHEMISTRY (MOSCOW) 2018; 83:S146-S162. [DOI: 10.1134/s0006297918140122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/09/2017] [Indexed: 08/30/2023]
|
36
|
Glidden MD, Aldabbagh K, Phillips NB, Carr K, Chen YS, Whittaker J, Phillips M, Wickramasinghe NP, Rege N, Swain M, Peng Y, Yang Y, Lawrence MC, Yee VC, Ismail-Beigi F, Weiss MA. An ultra-stable single-chain insulin analog resists thermal inactivation and exhibits biological signaling duration equivalent to the native protein. J Biol Chem 2017; 293:47-68. [PMID: 29114035 DOI: 10.1074/jbc.m117.808626] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Indexed: 12/12/2022] Open
Abstract
Thermal degradation of insulin complicates its delivery and use. Previous efforts to engineer ultra-stable analogs were confounded by prolonged cellular signaling in vivo, of unclear safety and complicating mealtime therapy. We therefore sought an ultra-stable analog whose potency and duration of action on intravenous bolus injection in diabetic rats are indistinguishable from wild-type (WT) insulin. Here, we describe the structure, function, and stability of such an analog, a 57-residue single-chain insulin (SCI) with multiple acidic substitutions. Cell-based studies revealed native-like signaling properties with negligible mitogenic activity. Its crystal structure, determined as a novel zinc-free hexamer at 2.8 Å, revealed a native insulin fold with incomplete or absent electron density in the C domain; complementary NMR studies are described in the accompanying article. The stability of the analog (ΔGU 5.0(±0.1) kcal/mol at 25 °C) was greater than that of WT insulin (3.3(±0.1) kcal/mol). On gentle agitation, the SCI retained full activity for >140 days at 45 °C and >48 h at 75 °C. These findings indicate that marked resistance to thermal inactivation in vitro is compatible with native duration of activity in vivo Further, whereas WT insulin forms large and heterogeneous aggregates above the standard 0.6 mm pharmaceutical strength, perturbing the pharmacokinetic properties of concentrated formulations, dynamic light scattering, and size-exclusion chromatography revealed only limited SCI self-assembly and aggregation in the concentration range 1-7 mm Such a combination of favorable biophysical and biological properties suggests that SCIs could provide a global therapeutic platform without a cold chain.
Collapse
Affiliation(s)
- Michael D Glidden
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Khadijah Aldabbagh
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yen-Shan Chen
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Manijeh Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | | | - Nischay Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Mamuni Swain
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yi Peng
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yanwu Yang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vivien C Yee
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
37
|
Evaluation of pharmacokinetic model designs for subcutaneous infusion of insulin aspart. J Pharmacokinet Pharmacodyn 2017; 44:477-489. [DOI: 10.1007/s10928-017-9535-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/11/2017] [Indexed: 10/19/2022]
|
38
|
Moroder L, Musiol HJ. Insulin - von seiner Entdeckung bis zur industriellen Synthese moderner Insulin-Analoga. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201702493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Luis Moroder
- Bioorganische Chemie; Max-Planck-Institut für Biochemie; Am Klopferspitz 18 82152 Martinsried Deutschland
| | - Hans-Jürgen Musiol
- Bioorganische Chemie; Max-Planck-Institut für Biochemie; Am Klopferspitz 18 82152 Martinsried Deutschland
| |
Collapse
|
39
|
Moroder L, Musiol HJ. Insulin-From its Discovery to the Industrial Synthesis of Modern Insulin Analogues. Angew Chem Int Ed Engl 2017; 56:10656-10669. [DOI: 10.1002/anie.201702493] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Luis Moroder
- Bioorganic Chemistry; Max-Planck Institute of Biochemistry; Am Klopferspitz 18 82152 Martinsried Germany
| | - Hans-Jürgen Musiol
- Bioorganic Chemistry; Max-Planck Institute of Biochemistry; Am Klopferspitz 18 82152 Martinsried Germany
| |
Collapse
|
40
|
Lieblich SA, Fang KY, Cahn JKB, Rawson J, LeBon J, Ku HT, Tirrell DA. 4S-Hydroxylation of Insulin at ProB28 Accelerates Hexamer Dissociation and Delays Fibrillation. J Am Chem Soc 2017; 139:8384-8387. [PMID: 28598606 PMCID: PMC5812673 DOI: 10.1021/jacs.7b00794] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Daily injections of insulin provide lifesaving benefits to millions of diabetics. But currently available prandial insulins are suboptimal: The onset of action is delayed by slow dissociation of the insulin hexamer in the subcutaneous space, and insulin forms amyloid fibrils upon storage in solution. Here we show, through the use of noncanonical amino acid mutagenesis, that replacement of the proline residue at position 28 of the insulin B-chain (ProB28) by (4S)-hydroxyproline (Hzp) yields an active form of insulin that dissociates more rapidly, and fibrillates more slowly, than the wild-type protein. Crystal structures of dimeric and hexameric insulin preparations suggest that a hydrogen bond between the hydroxyl group of Hzp and a backbone amide carbonyl positioned across the dimer interface may be responsible for the altered behavior. The effects of hydroxylation are stereospecific; replacement of ProB28 by (4R)-hydroxyproline (Hyp) causes little change in the rates of fibrillation and hexamer disassociation. These results demonstrate a new approach that fuses the concepts of medicinal chemistry and protein design, and paves the way to further engineering of insulin and other therapeutic proteins.
Collapse
Affiliation(s)
- Seth A. Lieblich
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Katharine Y. Fang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jackson K. B. Cahn
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
- Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jeanne LeBon
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - H. Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
- Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA
| | - David A. Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
41
|
Xie J, Lu Y, Wang W, Zhu H, Wang Z, Cao Z. Simple Protein Modification Using Zwitterionic Polymer to Mitigate the Bioactivity Loss of Conjugated Insulin. Adv Healthc Mater 2017; 6. [PMID: 28337855 DOI: 10.1002/adhm.201601428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/28/2017] [Indexed: 12/17/2022]
Abstract
Polymer-protein conjugation has been extensively explored toward a better protein drug with improved pharmacokinetics. However, a major problem with polymer-protein conjugation is that the polymers drastically reduce the bioactivity of the modified protein. There is no perfect solution to prevent the bioactivity loss, no matter the polymer is conjugated in a non-site specific way, or a more complex site-specific procedure. Here the authors report for the first time that when zwitterionic carboxybetaine polymer (PCB) is conjugated to insulin through simple conventional coupling chemistry. The resulting PCB-insulin does not show a significant reduction of in vitro bioactivity. The obtained PCB-insulin shows two significant advantages as a novel pharmaceutical agent. First, its therapeutic performance is remarkable. For PCB-insulin, there is a 24% increase of in vivo pharmacological activity of lowering blood glucose compared with native insulin. Such uncommonly seen increase has rarely been reported and is expected to be due to both the improved pharmacokinetics and retained bioactivity of PCB-insulin. Second, the production is simple from manufacturing standpoints. Conjugation procedure involves only one-step coupling reaction without complex site-specific linkage technique. The synthesized PCB-insulin conjugates do not require chromatographic separation to purify and obtain particular isoforms.
Collapse
Affiliation(s)
- Jinbing Xie
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| | - Yang Lu
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| | - Wei Wang
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| | - Hui Zhu
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| | - Zhigang Wang
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science; Wayne State University; Detroit MA 48202 USA
| |
Collapse
|
42
|
Mroz PA, Perez-Tilve D, Liu F, Mayer JP, DiMarchi RD. Native Design of Soluble, Aggregation-Resistant Bioactive Peptides: Chemical Evolution of Human Glucagon. ACS Chem Biol 2016; 11:3412-3420. [PMID: 27797473 DOI: 10.1021/acschembio.6b00923] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Peptide-based therapeutics commonly suffer from biophysical properties that compromise pharmacology and medicinal use. Structural optimization of the primary sequence is the usual route to address such challenges while trying to maintain as much native character and avoiding introduction of any foreign element that might evoke an immunological response. Glucagon serves a seminal physiological role in buffering against hypoglycemia, but its low aqueous solubility, chemical instability, and propensity to self-aggregate severely complicate its medicinal use. Selective amide bond replacement with metastable ester bonds is a preferred approach to the preparation of peptides with biophysical properties that otherwise inhibit synthesis. We have recruited such chemistry in the design and development of unique glucagon prodrugs that have physical properties suitable for medicinal use and yet rapidly convert to native hormone upon exposure to slightly alkaline pH. These prodrugs demonstrate in vitro and in vivo pharmacology when formulated in physiological buffers that are nearly identical to native hormone when solubilized in conventional dilute hydrochloric acid. This approach provides the best of both worlds, where the pro-drug delivers chemical properties supportive of aqueous formulation and the native biological properties.
Collapse
Affiliation(s)
- Piotr A. Mroz
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Diego Perez-Tilve
- Department
of Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - Fa Liu
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - John P. Mayer
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - Richard D. DiMarchi
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| |
Collapse
|
43
|
Buelke-Sam J, Byrd RA, Hoyt JA, Zimmermann JL. A Reproductive and Developmental Toxicity Study in CD Rats of LY275585, [Lys(B28),Pro(B29)]-Human Insulin. ACTA ACUST UNITED AC 2016. [DOI: 10.3109/10915819409140597] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
LY275585, [Lys(B28),Pro(B29)]-human insulin, was administered daily by subcutaneous injection at doses of 0, 1, 5, or 20 U/kg. Male rats were treated with LY275585 beginning 2 weeks prior to cohabitation and throughout the mating period. Females assigned to the teratology component of the study were treated for 2 weeks prior to cohabitation with the males and through gestation day (GD) 19. These dams were killed on GD 20 and uterine and fetal examinations were performed. Female rats assigned to the delivery component were treated for 2 weeks prior to cohabitation through postpartum day (PD) 20. Dams were allowed to deliver and maintain their progeny through a 21-day lactation period. After weaning, 1 pup/sex/litter was assigned to the F1 generation, and these animals received no treatment. Survival, growth, behavior, and reproductive performance were evaluated, and reproductive organs were collected for histological evaluation. Treatment of F0 male and female rats with LY275585 resulted in isolated incidences of severe hypoglycemia at 5 and 20 U/kg/day and some modest changes in food consumption and body weight measures at all treatment levels. These changes were anticipated and attributed to the pharmacology of this compound. Mating and fertility of the F0 animals were unaffected by treatment. While slight decreases in fetal body weights and increased fetal runts/litter were observed in the 20-U/kg/day group, PD 1 progeny weights were not affected in the delivery component, and there was no indication of teratogenicity in this study. There were no remarkable treatment-related effects on offspring growth patterns, survival, or reproductive performance, but the F1 animals from the 20-U/kg/day treatment-derived group were more reactive than controls in the startle habituation test. Thus, F0 parental toxicity, related to the hypoglycemic action of LY275585, was found at all doses. A dose of 5 U/kg/day was considered a no-adverse-effect level for developmental toxicity. There were no remarkable effects of LY275585 treatment on F0 or F1 generation reproductive performance at 20 U/kg/day, the highest dose tested in this study.
Collapse
Affiliation(s)
- J. Buelke-Sam
- P.O. Box 708, Lilly Research Laboratories, Eli Lilly & Company, Greenfield, IN 46140, U.S.A
| | - R. A. Byrd
- P.O. Box 708, Lilly Research Laboratories, Eli Lilly & Company, Greenfield, IN 46140, U.S.A
| | - J. A. Hoyt
- P.O. Box 708, Lilly Research Laboratories, Eli Lilly & Company, Greenfield, IN 46140, U.S.A
| | - J. L. Zimmermann
- P.O. Box 708, Lilly Research Laboratories, Eli Lilly & Company, Greenfield, IN 46140, U.S.A
| |
Collapse
|
44
|
Chaturvedi SK, Siddiqi MK, Alam P, Khan RH. Protein misfolding and aggregation: Mechanism, factors and detection. Process Biochem 2016. [DOI: 10.1016/j.procbio.2016.05.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Abstract
Diabetes is associated with microvascular and macrovascular complications leading to significant morbidity and mortality. Glycaemic control is important to prevent and delay the progression of these complications. An ideal insulin regimen in patients with diabetes would mirror the 24-hour insulin profile of a non-diabetic person and thereby prevent hyperglycaemia without inducing hypoglycaemia. This has, until recently, proved difficult to reproduce by regular subcutaneous insulin injections due to the inherent pharmacokinetic properties of the available insulins. Normoglycaemia was rarely achieved without hypoglycaemia compromising the quality of patients' lives. The advent of the new long- and short-acting insulin analogues are expected to both improve glycaemic control leading to a reduction in diabetes-related complications and reduce the incidence of hypoglycaemia thereby offering patients a better lifestyle.
Collapse
Affiliation(s)
- Bushra Ahmad
- Department of Diabetes, Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, OX3 7LJ, UK,
| |
Collapse
|
46
|
Zimmermann JL, Truex LL. 12-Month Chronic Toxicity Study of LY275585 (Human Insulin Analog) Administered Subcutaneously to Fischer 344 Rats. Int J Toxicol 2016. [DOI: 10.1080/109158197226946] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The monocomponent insulin analog LY275585 was administered to Fischer 344 rats in daily subcutaneous doses of 0, 20, or 200 U/kg for 12 months. Each treatment group consisted of 30 males and 30 females, and the vehicle control group consisted of 35 of each sex. Changes related to the pharmacologic activity of LY275585 included an increase in body weight, body weight gain, food consumption, and efficacy of food utilization in male and female rats given 200 U/kg. Pharmacologically related clinical chemistry effects occurring at 200 U/kg were an increase in mean serum glucose (24 h postdose) of males and females, a slight decrease in mean serum triglyceride of females, and decreased mean serum cholesterol of males and females. Fewer effects occurred in low-dose rats (20 U/kg) and included an increase of efficiency of food utilization only in female rats, a slight decrease in mean serum triglyceride in females, and decreased mean serum cholesterol in low-dose males and females. Three male and two female rats administered 200 U/kg died during the treatment period. Death of one male was attributed to hypoglycemia. Cause of death of four other high-dose rats was either not apparent, or the result of a variety of intercurrent disease processes unrelated to treatment. Survival in the low-dose (20 U/kg) and control groups was similar; one rat in each group died prior to treatment termination. The only compound-related lesion associated with treatment was a non-dose-related enhancement of the degree and incidence of inflammation at the site of subcutaneous injection. There were no compound-related changes in background lesions including neoplasms, or in hematologic or urinalysis values. In a comparison study, pharmacokinetic profiles of LY275585 and serum glucose values were determined in groups of 30 male and female rats administered single doses of 20 or 200 U/kg. Plasma levels of LY275585 peaked after 30 min, were dose related, and were similar in male and female rats. The hypoglycemia produced in rats receiving a single dose of 20 U/kg was maximal, suggesting saturation of the insulin receptor sites at this dose. The dose of 200 U/kg of LY275585 was, therefore, a supramaximal pharmacological dose when compared to an average daily human dose of 0.5 U/kg/ d. It is concluded that no toxicologically significant changes, other than moderate injection site inflammation, occurred in Fischer 344 rats treated with LY275585 at doses of 20 and 200 U/kg for 12 months.
Collapse
Affiliation(s)
- J. L. Zimmermann
- Lilly Research Laboratories, P.O. Box 708, Greenfield, IN 46140, USA
| | - L. L. Truex
- Lilly Research Laboratories, P.O. Box 708, Greenfield, IN 46140, USA
| |
Collapse
|
47
|
Tay T, Agius C, Hamilton R, Bodle J, Rockman S. Investigation into alternative testing methodologies for characterization of influenza virus vaccine. Hum Vaccin Immunother 2016; 11:1673-84. [PMID: 26090618 PMCID: PMC4514297 DOI: 10.1080/21645515.2015.1034914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The objective of this study was to explore various testing methodologies suitable for characterizing sedimented or agglomerated material. To model this, bioCSL's split influenza virus vaccine, Fluvax® was utilized. The investigation was conducted on 5 dispensed lots of commercially manufactured vaccine, formulated for the 2013 Southern Hemisphere season. Vaccine syringes were initially inspected by visual tests; the material was then aseptically pooled for characterization assessment by microscopy and several agglomeration assays. All syringes passed bioCSL's description test where any fine or large sized particles of sediment observed in the vaccine were resuspended upon shaking; inverted light microscopy verified that the sediment morphology was consistent with influenza vaccine. Electron microscopic examination of pooled vaccine material demonstrated the presence of typical influenza structures including split virus, virosomes, whole virus particles and agglomerates. An optical density turbidity assay revealed relatively high protein recoveries in the vaccine supernatant post-centrifugation treatment, thus indicative of a well-dispersed vaccine formulation. This was corroborated by particle sizing analysis using dynamic light scattering which generated reproducible volume particle size distributions of a polydisperse nature. Ultraviolet-visible absorbance profiles further confirmed the presence of some agglomerated material. Data from all methods demonstrated consistent results between all batches of vaccine. Therefore, this investigation revealed the suitability and usefulness of the various methodologies in characterizing the appearance of agglomerated vaccine material. It is suggested that such methods may be applicable and beneficial for the development of a wider spectrum of heterogeneous and agglomerated formulations to provide safe, efficacious and superior quality biopharmaceutical products.
Collapse
Key Words
- AI, Agglomeration index
- ANOVA, Analysis of variance
- AUC, Analytical ultracentrifugation
- DLS, Dynamic light scattering
- EM, Electron microscopy
- EP, European Pharmacopoeia
- FDA, Food and Drug Administration
- FFF, Field flow fractionation
- HA, Hemagglutinin
- ILM, Inverted light microscopy
- IVV, Influenza virus vaccine
- NA, Neuraminidase
- OD, Optical density
- ODT, Optical density turbidity
- PSD, Particle size distribution
- QbD, Quality by Design
- RCF, Relative centrifugal force
- RI, Refractive index
- SEC, Size exclusion chromatography
- SH, Southern Hemisphere
- SLS, Static light scattering
- TEM, Transmission electron microscope
- TGA, Therapeutic Goods Administration
- TIV, Trivalent influenza vaccine
- UV, Ultraviolet
- biopharmaceuticals
- characterization methods
- dispersed formulation
- influenza vaccine
- protein agglomeration
- sedimentation
Collapse
Affiliation(s)
- Tracy Tay
- a Influenza Research and Development; bioCSL Pty Ltd. ; Parkville , Victoria , Australia
| | | | | | | | | |
Collapse
|
48
|
Geoghegan JC, Fleming R, Damschroder M, Bishop SM, Sathish HA, Esfandiary R. Mitigation of reversible self-association and viscosity in a human IgG1 monoclonal antibody by rational, structure-guided Fv engineering. MAbs 2016; 8:941-50. [PMID: 27050875 DOI: 10.1080/19420862.2016.1171444] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Undesired solution behaviors such as reversible self-association (RSA), high viscosity, and liquid-liquid phase separation can introduce substantial challenges during development of monoclonal antibody formulations. Although a global mechanistic understanding of RSA (i.e., native and reversible protein-protein interactions) is sufficient to develop robust formulation controls, its mitigation via protein engineering requires knowledge of the sites of protein-protein interactions. In the study reported here, we coupled our previous hydrogen-deuterium exchange mass spectrometry findings with structural modeling and in vitro screening to identify the residues responsible for RSA of a model IgG1 monoclonal antibody (mAb-C), and rationally engineered variants with improved solution properties (i.e., reduced RSA and viscosity). Our data show that mutation of either solvent-exposed aromatic residues within the heavy and light chain variable regions or buried residues within the heavy chain/light chain interface can significantly mitigate RSA and viscosity by reducing the IgG's surface hydrophobicity. The engineering strategy described here highlights the utility of integrating complementary experimental and in silico methods to identify mutations that can improve developability, in particular, high concentration solution properties, of candidate therapeutic antibodies.
Collapse
Affiliation(s)
- James C Geoghegan
- a Department of Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Ryan Fleming
- a Department of Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Melissa Damschroder
- a Department of Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Steven M Bishop
- b Department of Formulation Sciences , MedImmune , Gaithersburg , MD , USA
| | - Hasige A Sathish
- b Department of Formulation Sciences , MedImmune , Gaithersburg , MD , USA
| | - Reza Esfandiary
- b Department of Formulation Sciences , MedImmune , Gaithersburg , MD , USA
| |
Collapse
|
49
|
Esfandiary R, Parupudi A, Casas-Finet J, Gadre D, Sathish H. Mechanism of Reversible Self-Association of a Monoclonal Antibody: Role of Electrostatic and Hydrophobic Interactions. J Pharm Sci 2015; 104:577-86. [DOI: 10.1002/jps.24237] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 02/06/2023]
|
50
|
Handorf AM, Sollinger HW, Alam T. Genetic Engineering of Surrogate <i>β</i> Cells for Treatment of Type 1 Diabetes Mellitus. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/jdm.2015.54037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|