1
|
Zhang Y, Austin MJ, Chou DHC. Insulin Stabilization Designs for Enhanced Therapeutic Efficacy and Accessibility. Acc Chem Res 2024. [PMID: 39466175 DOI: 10.1021/acs.accounts.4c00500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
ConspectusInsulin has remained indispensable in the treatment of diabetes since it was first discovered in 1921. Unlike small molecular drugs, insulin and other protein drugs are prone to degradation when exposed to elevated temperatures, mechanical agitation during transportation, and prolonged storage periods. Therefore, strict cold-chain management is crucial for the insulin supply, requiring significant resources, which can limit the access to insulin, particularly in low-income areas. Moreover, although insulin formulations have advanced tremendously in the last century, insulin treatment still imposes a challenging regimen and provides suboptimal outcomes for the majority of patients. There is an increasing focus on pursuing improved pharmacology, specifically on safer, more user-friendly insulin therapies that minimize the self-management burden. These challenges underscore the need for developing novel insulin formulations with improved stability that are compatible with advanced insulin therapy.Insulin stabilization can be achieved through either chemical modification of insulin or formulation component design. Inspired by insulin-like peptides from invertebrates, we have developed novel stable insulin analogs based on a fundamental understanding of the insulin receptor engagement for insulin bioactivity. We created a novel four-disulfide insulin analog with high aggregation stability and potency by introducing a fourth disulfide bond between a C-terminal extended insulin A-chain and residues near the C-terminus of the B-chain. In an effort to stabilize insulin in its monomeric state to develop ultrafast-acting insulin with rapid absorption upon injection, we have developed a series of structurally miniaturized yet fully active insulin analogs that do not form dimers due to the lack of the canonical B-chain C-terminal octapeptide. Additionally, our study provided strategies for expanding the scope of cucurbit[7]uril (CB[7])-assisted insulin stabilization by engineering safe and biodegradable CB[7]-zwitterionic polypeptide excipients. We also explored insulin N-terminal substitution methods to achieve pH-dependent insulin stabilization without prolonging the duration of action.This Account describes our exploration of engineering stable insulin analogs and formulation design strategies for stabilizing insulin in aqueous solutions. Beyond conventional stabilization strategies for insulin injections, the unmet challenges and recent innovations in insulin stabilization are discussed, addressing the growing demand for alternative, less invasive routes of insulin administration. Additionally, we aim to provide a thorough overview of insulin stabilization from the perspective of commercially available insulin drugs and common pharmaceutical engineering practices in the industry. We also highlight unresolved insulin stabilization challenges and ongoing research strategies. We anticipate that further emphasis on collective efforts of protein engineering, pharmaceutical formulation design, and drug delivery will inform the development of stable and advanced insulin therapy.
Collapse
Affiliation(s)
- Yanxian Zhang
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Maxwell Jack Austin
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Danny Hung-Chieh Chou
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
2
|
Bian H, Shao X, Cai W, Fu H. Understanding the Reversible Binding of a Multichain Protein-Protein Complex through Free-Energy Calculations. J Phys Chem B 2024; 128:3598-3604. [PMID: 38574232 DOI: 10.1021/acs.jpcb.4c00519] [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: 04/06/2024]
Abstract
We demonstrate that the binding affinity of a multichain protein-protein complex, insulin dimer, can be accurately predicted using a streamlined route of standard binding free-energy calculations. We find that chains A and C, which do not interact directly during binding, stabilize the insulin monomer structures and reduce the binding affinity of the two monomers, therefore enabling their reversible association. Notably, we confirm that although classical methods can estimate the binding affinity of the insulin dimer, conventional molecular dynamics, enhanced sampling algorithms, and classical geometrical routes of binding free-energy calculations may not fully capture certain aspects of the role played by the noninteracting chains in the binding dynamics. Therefore, this study not only elucidates the role of noninteracting chains in the reversible binding of the insulin dimer but also offers a methodological guide for investigating the reversible binding of multichain protein-protein complexes utilizing streamlined free-energy calculations.
Collapse
Affiliation(s)
- Hengwei Bian
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
- School of Materials Science and Engineering, Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin 300350, China
| |
Collapse
|
3
|
Pujahari SR, Purusottam RN, Mali PS, Sarkar S, Khaneja N, Vajpai N, Kumar A. Exploring the Higher Order Structure and Conformational Transitions in Insulin Microcrystalline Biopharmaceuticals by Proton-Detected Solid-State Nuclear Magnetic Resonance at Natural Abundance. Anal Chem 2024; 96:4756-4763. [PMID: 38326990 DOI: 10.1021/acs.analchem.3c04040] [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: 02/09/2024]
Abstract
The integrity of a higher order structure (HOS) is an essential requirement to ensure the efficacy, stability, and safety of protein therapeutics. Solution-state nuclear magnetic resonance (NMR) occupies a unique niche as one of the most promising methods to access atomic-level structural information on soluble biopharmaceutical formulations. Another major class of drugs is poorly soluble, such as microcrystalline suspensions, which poses significant challenges for the characterization of the active ingredient in its native state. Here, we have demonstrated a solid-state NMR method for HOS characterization of biopharmaceutical suspensions employing a selective excitation scheme under fast magic angle spinning (MAS). The applicability of the method is shown on commercial insulin suspensions at natural isotopic abundance. Selective excitation aided with proton detection and non-uniform sampling (NUS) provides improved sensitivity and resolution. The enhanced resolution enabled us to demonstrate the first experimental evidence of a phenol-escaping pathway in insulin, leading to conformational transitions to different hexameric states. This approach has the potential to serve as a valuable means for meticulously examining microcrystalline biopharmaceutical suspensions, which was previously not attainable in their native formulation states and can be seamlessly extended to other classes of biopharmaceuticals such as mAbs and other microcrystalline proteins.
Collapse
Affiliation(s)
- Soumya Ranjan Pujahari
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| | - Rudra N Purusottam
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| | - Pramod S Mali
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| | - Sambeda Sarkar
- System and Control Engineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| | - Navin Khaneja
- System and Control Engineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| | - Navratna Vajpai
- Biocon Biologics Limited, Biocon SEZ, Plot No. 2 & 3, Phase IV-B.I.A, Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai Mumbai 400076, India
| |
Collapse
|
4
|
Begum S, Parvej H, Dalui R, Paul S, Maity S, Sepay N, Afzal M, Chandra Halder U. Structural modulation of insulin by hydrophobic and hydrophilic molecules. RSC Adv 2023; 13:34097-34106. [PMID: 38019994 PMCID: PMC10662218 DOI: 10.1039/d3ra06647a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
In the bloodstream, insulin interacts with various kinds of molecules, which can alter its structure and modulate its function. In this work, we have synthesized two molecules having extremely hydrophilic and hydrophobic side chains. The effects of hydrophilic and hydrophobic molecules on the binding with insulin have been investigated through a multi-spectroscopic approach. We found that hydrophilic molecules have a slightly higher binding affinity towards insulin. Insulin can bind with the hydrophilic molecules as it binds glucose. The high insulin binding affinity of a hydrophobic molecule indicates its dual nature. The hydrophobic molecule binds at the hydrophobic pocket of the insulin surface, where hydrophilic molecules interact at the polar surface of the insulin. Such binding with the hydrophobic molecule perturbs strongly the secondary structure of the insulin much more in comparison to hydrophilic molecules. Therefore, the stability of insulin decreases in the presence of hydrophobic molecules.
Collapse
Affiliation(s)
- Shahnaz Begum
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Hasan Parvej
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Ramkrishna Dalui
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Swarnali Paul
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Sanhita Maity
- Department of Chemistry, Jadavpur University Kolkata-700032 India
| | - Nayim Sepay
- Department of Chemistry, Lady Brabourne College Kolkata-700017 India
| | - Mohd Afzal
- Department of Chemistry, College of Science, King Saud University Riyadh 11451 Saudi Arabia
| | | |
Collapse
|
5
|
Supramolecular approaches for insulin stabilization without prolonged duration of action. Acta Pharm Sin B 2023; 13:2281-2290. [DOI: 10.1016/j.apsb.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/05/2022] [Accepted: 11/08/2022] [Indexed: 01/13/2023] Open
|
6
|
Mohamed OA, Samir S, Omar H, Hassan EA, Abdelazeem E. Lab-scale Preparation of Recombinant Human Insulin-like Growth Factor-1 in Escherichia coli and its Potential Safety on Normal Human Lung Cell Line. Recent Pat Biotechnol 2022; 16:266-280. [PMID: 35418294 DOI: 10.2174/1872208316666220412105822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/30/2022] [Accepted: 03/03/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Insulin-like growth factor-1 (IGF-1) is structurally similar to insulin and acts as an endocrine hormone secreted by the liver. OBJECTIVE Production of recombinant human IGF-1 (rhIGF-1) in Escherichia coli (E.coli) and evaluation of its proliferation stimulatory activity. METHODS hIGF-1 gene cloned into pBSK (+) simple vector was transformed into TOP 10 chemically competent cells of E. coli. Polymerase chain reaction (PCR) was achieved using specific hIGF-1 gene primers to confirm the successful transformation. To express the rhIGF-1 in E. coli (Rosetta (DE3) pLysS); the hIGF-1 gene was cloned into the pET-15b expression vector and then the recombinant pET-15b/IGF-1 vector was transformed into a chemically prepared competent expression bacterial cells; Rosetta (DE3) pLysS. The rhIGF-1 was expressed as insoluble aggregates called inclusion bodies (IBs) using a 2 mM Isopropyl β-D-1-thiogalactopyranoside (IPTG) inducer. IBs were solubilized in a denatured form using 6 M guanidinium hydrochloride (GdmCl), followed by in vitro protein refolding using the rapid dilution method. The refolded hIGF-1 was purified using the HiTrap- ANX anion exchange column. Western blot and ELISA using rabbit polyvalent anti-hIGF- 1 were performed to confirm the protein antigenic identity. Cell proliferation activity of rhIGF-1 was testified on normal human lung cell line (WI-38). RESULTS rhIGF-1 was purified from the HiTrap-ANX column at a concentration of 300 μg/ml. Western blot showed a single 7.6 kDa band obtained in the induced Rosetta (DE3) pLYsS. ELISA confirmed the molecular identity of the rhIGF-1 epitope, the concentration of purified rhIGF-1 obtained from the ELISA standard curve using rhIGF-1 reference protein as a standard was 300 μg/ml, and activity on WI-38 cells was 2604.17I U/mg. CONCLUSION Biologically active native rhIGF-1 protein was successfully expressed. Patents related to the preparation of IGF-1 were mentioned along the text.
Collapse
Affiliation(s)
- Omnia A Mohamed
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Safia Samir
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Hanan Omar
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Ekrami A Hassan
- Biochemistry Department, Faculty of Science, Ain-Shams University, Cairo, Egypt
| | - Eman Abdelazeem
- Biochemistry Department, Faculty of Science, Ain-Shams University, Cairo, Egypt
| |
Collapse
|
7
|
Pourhosseini PS, Ghasemitabesh R, Pirhaghi M, Fayazzadeh S, Saboury AA, Najafi F. Urethane-containing cationic gemini surfactants with amphiphilic tails: novel near-neutral protein carriers with minor effects on insulin structure. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Das A, Shah M, Saraogi I. Molecular Aspects of Insulin Aggregation and Various Therapeutic Interventions. ACS BIO & MED CHEM AU 2022; 2:205-221. [PMID: 37101572 PMCID: PMC10114644 DOI: 10.1021/acsbiomedchemau.1c00054] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Protein aggregation leading to the formation of amyloid fibrils has various adverse effects on human health ranging from fatigue and numbness to organ failure and death in extreme cases. Insulin, a peptide hormone commonly used to treat diabetes, undergoes aggregation at the site of repeated injections in diabetic patients as well as during its industrial production and transport. The reduced bioavailability of insulin due to aggregation hinders the proper control of glucose levels in diabetic patients. Thus, it is necessary to develop rational approaches for inhibiting insulin aggregation, which in turn requires a detailed understanding of the mechanism of fibrillation. Given the relative simplicity of insulin and ease of access, insulin has also served as a model system for studying amyloids. Approaches to inhibit insulin aggregation have included the use of natural molecules, synthetic peptides or small molecules, and bacterial chaperone machinery. This review focuses on insulin aggregation with an emphasis on its mechanism, the structural features of insulin fibrils, and the reported inhibitors that act at different stages in the aggregation pathway. We discuss molecules that can serve as leads for improved inhibitors for use in commercial insulin formulations. We also discuss the aggregation propensity of fast- and slow-acting insulin biosimilars, commonly administered to diabetic patients. The development of better insulin aggregation inhibitors and insights into their mechanism of action will not only aid diabetic therapies, but also enhance our knowledge of protein amyloidosis.
Collapse
Affiliation(s)
- Anirban Das
- Department
of Chemistry and Department of Biological Sciences, Indian
Institute of Science Education and Research
Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Mosami Shah
- Department
of Chemistry and Department of Biological Sciences, Indian
Institute of Science Education and Research
Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Ishu Saraogi
- Department
of Chemistry and Department of Biological Sciences, Indian
Institute of Science Education and Research
Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| |
Collapse
|
9
|
Iyer G, Dyawanapelly S, Jain R, Dandekar P. An overview of oral insulin delivery strategies (OIDS). Int J Biol Macromol 2022; 208:565-585. [PMID: 35346680 DOI: 10.1016/j.ijbiomac.2022.03.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Despite tremendous efforts, the world continues its fight against the common chronic disease-diabetes. Diabetes is caused by elevated glucose levels in the blood, which can lead to several complications like glaucoma, cataract, kidney failure, diabetic ketoacidosis, heart attack, and stroke. According to recent statistics, China, India, and the US rank at the top three positions with regards to the number of patients affected by diabetes. Ever since its discovery, insulin is one of the major therapeutic molecules that is used to control the disease in the diabetic population, worldwide. The most common route of insulin administration has been the subcutaneous route. However, the limitations associated with this route have motivated global efforts to explore alternative strategies to deliver insulin, including pulmonary, transdermal, nasal, rectal, buccal, and oral routes. Oral insulin delivery is the most convenient and patient-centered route. However, the oral route is also associated with numerous drawbacks that present significant challenges to the scientific fraternity. The human physiological system acts as a formidable barrier to insulin, limiting its bioavailability. The present review covers the major barriers against oral insulin delivery and explains formulation strategies that have been adopted to overcome these barriers. The review focuses on oral insulin delivery strategies (OIDS) for increasing the bioavailability of oral insulin, including nanoparticles, microparticles, nano-in-microparticles, hydrogels, tablets, capsules, intestinal patches, and use of ionic liquids. It also highlights some of the notable recent advancements and clinical trials in oral insulin delivery. This formulation based OIDS may significantly improve patient compliance in the treatment of diabetes.
Collapse
Affiliation(s)
- Gayatri Iyer
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India
| | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Matunga, Mumbai 400019, India.
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India.
| |
Collapse
|
10
|
Sen S, Ali R, Onkar A, Ganesh S, Verma S. Strategies for interference of insulin fibrillogenesis: challenges and advances. Chembiochem 2022; 23:e202100678. [PMID: 35025120 DOI: 10.1002/cbic.202100678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Indexed: 11/10/2022]
Abstract
The discovery of insulin came up with very high hopes for diabetic patients. In the year 2021, the world celebrated the 100 th anniversary of the discovery of this vital hormone. However, external use of insulin is highly affected by its aggregating tendency that occurs during its manufacturing, transportation, and improper handling which ultimately leads its pharmaceutically and biologically ineffective form. In this review, we aim to discuss the various approaches used for decelerating insulin aggregation which results in the enhancement of its overall structural stability and usage. The approaches that are discussed are broadly classified as either a measure through excipient additions or by intrinsic modifications in the insulin native structure.
Collapse
Affiliation(s)
- Shantanu Sen
- Indian Institute of Technology Kanpur, Chemistry, INDIA
| | - Rafat Ali
- Indian Institute of Technology Kanpur, Chemistry, Room No 131 Lab No2, CESE department IIT Kanpur, 208016, Kanpur, INDIA
| | - Akanksha Onkar
- Indian Institute of Technology Kanpur, Biological Sciences and Bioengineering, INDIA
| | - Subramaniam Ganesh
- Indian Institute of Technology Kanpur, Biological Sciences and Bioengineering, INDIA
| | - Sandeep Verma
- Indian Institute of Technology-Kanpur, Department of Chemistry, IIT-Kanpur, 208016, Kanpur, INDIA
| |
Collapse
|
11
|
Kjeldsen TB, Hubálek F, Hjørringgaard CU, Tagmose TM, Nishimura E, Stidsen CE, Porsgaard T, Fledelius C, Refsgaard HHF, Gram-Nielsen S, Naver H, Pridal L, Hoeg-Jensen T, Jeppesen CB, Manfè V, Ludvigsen S, Lautrup-Larsen I, Madsen P. Molecular Engineering of Insulin Icodec, the First Acylated Insulin Analog for Once-Weekly Administration in Humans. J Med Chem 2021; 64:8942-8950. [PMID: 33944562 DOI: 10.1021/acs.jmedchem.1c00257] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Here, we describe the molecular engineering of insulin icodec to achieve a plasma half-life of 196 h in humans, suitable for once-weekly subcutaneously administration. Insulin icodec is based on re-engineering of the ultra-long oral basal insulin OI338 with a plasma half-life of 70 h in humans. This systematic re-engineering was accomplished by (1) further increasing the albumin binding by changing the fatty diacid from a 1,18-octadecanedioic acid (C18) to a 1,20-icosanedioic acid (C20) and (2) further reducing the insulin receptor affinity by the B16Tyr → His substitution. Insulin icodec was selected by screening for long intravenous plasma half-life in dogs while ensuring glucose-lowering potency following subcutaneous administration in rats. The ensuing structure-activity relationship resulted in insulin icodec. In phase-2 clinical trial, once-weekly insulin icodec provided safe and efficacious glycemic control comparable to once-daily insulin glargine in type 2 diabetes patients. The structure-activity relationship study leading to insulin icodec is presented here.
Collapse
Affiliation(s)
- Thomas B Kjeldsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - František Hubálek
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | | | - Tina M Tagmose
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Erica Nishimura
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Carsten E Stidsen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Trine Porsgaard
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Christian Fledelius
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Hanne H F Refsgaard
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Sanne Gram-Nielsen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Helle Naver
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Lone Pridal
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Thomas Hoeg-Jensen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Claus Bekker Jeppesen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Valentina Manfè
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Svend Ludvigsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Inger Lautrup-Larsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Peter Madsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| |
Collapse
|
12
|
Jarosinski MA, Dhayalan B, Rege N, Chatterjee D, Weiss MA. 'Smart' insulin-delivery technologies and intrinsic glucose-responsive insulin analogues. Diabetologia 2021; 64:1016-1029. [PMID: 33710398 PMCID: PMC8158166 DOI: 10.1007/s00125-021-05422-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Insulin replacement therapy for diabetes mellitus seeks to minimise excursions in blood glucose concentration above or below the therapeutic range (hyper- or hypoglycaemia). To mitigate acute and chronic risks of such excursions, glucose-responsive insulin-delivery technologies have long been sought for clinical application in type 1 and long-standing type 2 diabetes mellitus. Such 'smart' systems or insulin analogues seek to provide hormonal activity proportional to blood glucose levels without external monitoring. This review highlights three broad strategies to co-optimise mean glycaemic control and time in range: (1) coupling of continuous glucose monitoring (CGM) to delivery devices (algorithm-based 'closed-loop' systems); (2) glucose-responsive polymer encapsulation of insulin; and (3) mechanism-based hormone modifications. Innovations span control algorithms for CGM-based insulin-delivery systems, glucose-responsive polymer matrices, bio-inspired design based on insulin's conformational switch mechanism upon insulin receptor engagement, and glucose-responsive modifications of new insulin analogues. In each case, innovations in insulin chemistry and formulation may enhance clinical outcomes. Prospects are discussed for intrinsic glucose-responsive insulin analogues containing a reversible switch (regulating bioavailability or conformation) that can be activated by glucose at high concentrations.
Collapse
Affiliation(s)
- Mark A Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nischay Rege
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Deepak Chatterjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Chemistry, Indiana University, Bloomington, IN, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
13
|
Crosslinking of human plasma C-reactive protein to human serum albumin via disulfide bond oxidation. Redox Biol 2021; 41:101925. [PMID: 33714740 PMCID: PMC7966873 DOI: 10.1016/j.redox.2021.101925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 01/08/2023] Open
Abstract
Inter- and intra-molecular crosslinks can generate protein dysfunction, and are associated with protein aggregate accumulation in aged and diseased tissues. Crosslinks formed between multiple amino acid side chains can be reversible or irreversible. Disulfides formed either enzymatically, or as a result of oxidant-mediated reactions, are a major class of reversible crosslinks. Whilst these are commonly generated via oxidation of Cys thiol groups, they are also formed by ‘oxidant-mediated thiol-disulfide reactions’ via initial disulfide oxidation to a thiosulfinate or zwitterionic peroxide, and subsequent reaction with another thiol including those on other proteins. This generates new intermolecular protein-protein crosslinks. Here we demonstrate that photooxidation, or reaction with the biological oxidants HOCl and ONOOH, of the single disulfide present in the major human plasma inflammatory protein, C-reactive protein (CRP) can give rise to reversible disulfide bond formation with human serum albumin (HSA). This occurs in an oxidant dose-, or illumination-time-, dependent manner. These CRP-HSA crosslinks are formed both in isolated protein systems, and in fresh human plasma samples containing high, but not low, levels of CRP. The inter-protein crosslinks which involve Cys36 of CRP and Cys34 of HSA, have been detected by both immunoblotting and mass spectrometry (MS). The yield of protein-protein crosslinks depends on the nature and extent of oxidant exposure, and can be reversed by dithiothreitol and tris(2-carboxyethyl)phosphine hydrochloride. These data indicate that oxidation of disulfide bonds in proteins can be a source of novel inter-protein crosslinks, which may help rationalize the accumulation of crosslinked proteins in aged and diseased tissues. C-reactive protein (CRP) is a major acute phase inflammatory protein in human plasma. Oxidation of the single Cys36-Cys97 disulfide in CRP generates reactive intermediates. The oxidized disulfide reacts with Cys34 of human serum albumin to forms a new crosslink. The inter-protein CRP-HSA crosslink has been characterized by immunoblotting and LS-MS/MS. This novel crosslink may be a long-lived plasma marker of inflammation-induced damage.
Collapse
|
14
|
Tombling BJ, Lammi C, Lawrence N, Li J, Arnoldi A, Craik DJ, Wang CK. Engineered EGF-A Peptides with Improved Affinity for Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9). ACS Chem Biol 2021; 16:429-439. [PMID: 33512150 DOI: 10.1021/acschembio.0c00991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The epidermal growth-factor-like domain A (EGF-A) of the low-density lipoprotein (LDL) receptor is a promising lead for therapeutic inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9). However, the clinical potential of EGF-A is limited by its suboptimal affinity for PCSK9. Here, we use phage display to identify EGF-A analogues with extended bioactive segments that have improved affinity for PCSK9. The most potent analogue, TEX-S2_03, demonstrated ∼130-fold improved affinity over the parent domain and had a reduced calcium dependency for efficient PCSK9 binding. Thermodynamic binding analysis suggests the improved affinity of TEX-S2_03 is enthalpically driven, indicating favorable interactions are formed between the extended segment of TEX-S2_03 and the PCSK9 surface. The improved affinity of TEX-S2_03 resulted in increased activity in competition binding assays and more efficient restoration of LDL receptor levels with clearance of extracellular LDL cholesterol in functional cell assays. These results confirm that TEX-S2_03 is a promising therapeutic lead for treating hypercholesterolemia. Many EGF-like domains are involved in disease-related protein-protein interactions; therefore, our strategy for engineering EGF-like domains has the potential to be broadly implemented in EGF-based drug design.
Collapse
Affiliation(s)
- Benjamin J. Tombling
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Carmen Lammi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Nicole Lawrence
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jianqiang Li
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Anna Arnoldi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - David J. Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Conan K. Wang
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
15
|
Kaufmann B, Boulle P, Berthou F, Fournier M, Beran D, Ciglenecki I, Townsend M, Schmidt G, Shah M, Cristofani S, Cavailler P, Foti M, Scapozza L. Heat-stability study of various insulin types in tropical temperature conditions: New insights towards improving diabetes care. PLoS One 2021; 16:e0245372. [PMID: 33534816 PMCID: PMC7857579 DOI: 10.1371/journal.pone.0245372] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 12/29/2020] [Indexed: 01/08/2023] Open
Abstract
Strict storage recommendations for insulin are difficult to follow in hot tropical regions and even more challenging in conflict and humanitarian emergency settings, adding an extra burden to the management of people with diabetes. According to pharmacopeia unopened insulin vials must be stored in a refrigerator (2-8°C), while storage at ambient temperature (25-30°C) is usually permitted for the 4-week usage period during treatment. In the present work we address a critical question towards improving diabetes care in resource poor settings, namely whether insulin is stable and retains biological activity in tropical temperatures during a 4-week treatment period. To answer this question, temperature fluctuations were measured in Dagahaley refugee camp (Northern Kenya) using log tag recorders. Oscillating temperatures between 25 and 37°C were observed. Insulin heat stability was assessed under these specific temperatures which were precisely reproduced in the laboratory. Different commercialized formulations of insulin were quantified weekly by high performance liquid chromatography and the results showed perfect conformity to pharmacopeia guidelines, thus confirming stability over the assessment period (four weeks). Monitoring the 3D-structure of the tested insulin by circular dichroism confirmed that insulin monomer conformation did not undergo significant modifications. The measure of insulin efficiency on insulin receptor (IR) and Akt phosphorylation in hepatic cells indicated that insulin bioactivity of the samples stored at oscillating temperature during the usage period is identical to that of the samples maintained at 2-8°C. Taken together, these results indicate that insulin can be stored at such oscillating ambient temperatures for the usual four-week period of use. This enables the barrier of cold storage during use to be removed, thereby opening up the perspective for easier management of diabetes in humanitarian contexts and resource poor settings.
Collapse
Affiliation(s)
- Beatrice Kaufmann
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | | | - Flavien Berthou
- Faculty of Medicine, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Margot Fournier
- Faculty of Medicine, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - David Beran
- Division of Tropical and Humanitarian Medicine, University of Geneva and Geneva University Hospitals, Geneva, Switzerland
| | - Iza Ciglenecki
- Médecins Sans Frontières Switzerland, Geneva, Switzerland
| | | | | | - Maya Shah
- Médecins Sans Frontières Switzerland, Geneva, Switzerland
| | | | | | - Michelangelo Foti
- Faculty of Medicine, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Kabotso DEK, Smiley D, Mayer JP, Gelfanov VM, Perez-Tilve D, DiMarchi RD, Pohl NLB, Liu F. Addition of Sialic Acid to Insulin Confers Superior Physical Properties and Bioequivalence. J Med Chem 2020; 63:6134-6143. [PMID: 32406685 DOI: 10.1021/acs.jmedchem.0c00266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Native insulin is susceptible to biophysical aggregation and fibril formation, promoted by manual agitation and elevated temperatures. The safety of the drug and its application to alternative forms of administration could be enhanced through the identification of chemical modifications that strengthen its physical stability without compromising its biological properties. Complex polysialic acids (PSAs) exist naturally and provide a means to enhance the physical properties of peptide therapeutics. A set of insulin analogues site-specifically derivatized with sialic acid were prepared in an overall yield of 50-60%. Addition of a single or multiple sialic acids conferred remarkable enhancement to the biophysical stability of human insulin while maintaining its potency. The time to the onset of fibrillation was extended by more than 10-fold relative to that of the native hormone. These results demonstrate that simplified sialic acid conjugates represent a viable alternative to complex natural PSAs in increasing the stability of therapeutic peptides.
Collapse
Affiliation(s)
- Daniel E K Kabotso
- School of Basic and Biomedical Sciences, University of Health and Allied Sciences, PMB 31 Ho, Volta Region, Ghana.,Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - David Smiley
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - John P Mayer
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Vasily M Gelfanov
- Novo Nordisk Indianapolis Research Center, 5225 Exploration Dr., Indianapolis, Indiana 46241, United States
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati-College of Medicine, Cincinnati, Ohio 45267, United States
| | - Richard D DiMarchi
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Nicola L B Pohl
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Fa Liu
- Novo Nordisk Research Center, 530 Fairview Avenue North, Seattle, Washington 98109, United States
| |
Collapse
|
17
|
Frost TS, Jiang L, Zohar Y. Pharmacokinetic Analysis of Epithelial/Endothelial Cell Barriers in Microfluidic Bilayer Devices with an Air-Liquid Interface. MICROMACHINES 2020; 11:mi11050536. [PMID: 32466113 PMCID: PMC7281310 DOI: 10.3390/mi11050536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
As the range of applications of organs-on-chips is broadening, the evaluation of aerosol-based therapies using a lung-on-a-chip model has become an attractive approach. Inhalation therapies are not only minimally invasive but also provide optimal pharmacokinetic conditions for drug absorption. As drug development evolves, it is likely that better screening through use of organs-on-chips can significantly save time and cost. In this work, bio-aerosols of various compounds including insulin were generated using a jet nebulizer. The aerosol flows were driven through microfluidic bilayer devices establishing an air–liquid interface to mimic the blood–air barrier in human small airways. The aerosol flow in the microfluidic devices has been characterized and adjusted to closely match physiological values. The permeability of several compounds, including paracellular and transcellular biomarkers, across epithelial/endothelial cell barriers was measured. Concentration–time plots were established in microfluidic devices with and without cells; the curves were then utilized to extract standard pharmacokinetic parameters such as the area under the curve, maximum concentration, and time to maximum concentration. The cell barrier significantly affected the measured pharmacokinetic parameters, as compound absorption through the barrier decreases with its increasing molecular size. Aerosolizing insulin can lead to the formation of fibrils, prior to its entry to the microfluidic device, with a substantially larger apparent molecular size effectively blocking its paracellular transport. The results demonstrate the advantage of using lung-on-a-chip for drug discovery with applications such as development of novel inhaled therapies.
Collapse
Affiliation(s)
- Timothy S. Frost
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
- Correspondence:
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Yitshak Zohar
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
18
|
Østergaard M, Mishra NK, Jensen KJ. The ABC of Insulin: The Organic Chemistry of a Small Protein. Chemistry 2020; 26:8341-8357. [DOI: 10.1002/chem.202000337] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/15/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Mads Østergaard
- Department of ChemistryUniversity of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg C Denmark
| | - Narendra Kumar Mishra
- Department of ChemistryUniversity of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg C Denmark
| | - Knud J. Jensen
- Department of ChemistryUniversity of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg C Denmark
| |
Collapse
|
19
|
Collier AM, Nemtsova Y, Kuber N, Banach-Petrosky W, Modak A, Sleat DE, Nanda V, Lobel P. Lysosomal protein thermal stability does not correlate with cellular half-life: global observations and a case study of tripeptidyl-peptidase 1. Biochem J 2020; 477:727-745. [PMID: 31957806 PMCID: PMC8442665 DOI: 10.1042/bcj20190874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/15/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Late-infantile neuronal ceroid lipofuscinosis (LINCL) is a neurodegenerative lysosomal storage disorder caused by mutations in the gene encoding the protease tripeptidyl-peptidase 1 (TPP1). Progression of LINCL can be slowed or halted by enzyme replacement therapy, where recombinant human TPP1 is administered to patients. In this study, we utilized protein engineering techniques to increase the stability of recombinant TPP1 with the rationale that this may lengthen its lysosomal half-life, potentially increasing the potency of the therapeutic protein. Utilizing multiple structure-based methods that have been shown to increase the stability of other proteins, we have generated and evaluated over 70 TPP1 variants. The most effective mutation, R465G, increased the melting temperature of TPP1 from 55.6°C to 64.4°C and increased its enzymatic half-life at 60°C from 5.4 min to 21.9 min. However, the intracellular half-life of R465G and all other variants tested in cultured LINCL patient-derived lymphoblasts was similar to that of WT TPP1. These results provide structure/function insights into TPP1 and indicate that improving in vitro thermal stability alone is insufficient to generate TPP1 variants with improved physiological stability. This conclusion is supported by a proteome-wide analysis that indicates that lysosomal proteins have higher melting temperatures but also higher turnover rates than proteins of other organelles. These results have implications for similar efforts where protein engineering approaches, which are frequently evaluated in vitro, may be considered for improving the physiological properties of proteins, particularly those that function in the lysosomal environment.
Collapse
Affiliation(s)
- Aaron M. Collier
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
| | - Narendra Kuber
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
| | | | - Anurag Modak
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
| | - David E. Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
- Department of Biochemistry and Molecular Biology, Rutgers
University, Piscataway, NJ 08854
| | - Vikas Nanda
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
- Department of Biochemistry and Molecular Biology, Rutgers
University, Piscataway, NJ 08854
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers
University, Piscataway, NJ 08854
- Department of Biochemistry and Molecular Biology, Rutgers
University, Piscataway, NJ 08854
| |
Collapse
|
20
|
Balobanov V, Chertkova R, Egorova A, Dolgikh D, Bychkova V, Kirpichnikov M. The Kinetics of Amyloid Fibril Formation by de Novo Protein Albebetin and Its Mutant Variants. Biomolecules 2020; 10:E241. [PMID: 32033353 PMCID: PMC7072675 DOI: 10.3390/biom10020241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 11/17/2022] Open
Abstract
Engineering of amyloid structures is one of the new perspective areas of protein engineering. Studying the process of amyloid formation can help find ways to manage it in the interests of medicine and biotechnology. One of the promising candidates for the structural basis of artificial functional amyloid fibrils is albebetin (ABB), an artificial protein engineered under the leadership of O.B. Ptitsyn. Various aspects of the amyloid formation of this protein and some methods for controlling this process are investigated in this paper. Four stages of amyloid fibrils formation by this protein from the first non-fibrillar aggregates to mature fibrils and large micron-sized complexes have been described in detail. Dependence of albebetin amyloids formation on external conditions and some mutations also have been described. The introduction of similar point mutations in the two structurally identical α-β-β motifs of ABB lead to different amiloidogenesis kinetics. The inhibitory effect of a disulfide bond and high pH on amyloid fibrils formation, that can be used to control this process, was shown. The results of this work are a good basis for the further design and use of ABB-based amyloid constructs.
Collapse
Affiliation(s)
- Vitalii Balobanov
- Institute of Protein Research, Pushchino, Moscow 142290, Russia (V.B.)
| | - Rita Chertkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya st. 16/10, Moscow 117997, Russia
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow 117312, Russia
| | - Anna Egorova
- Institute of Protein Research, Pushchino, Moscow 142290, Russia (V.B.)
| | - Dmitry Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya st. 16/10, Moscow 117997, Russia
- Biology Department, Lomonosov Moscow State University, Leninskie gory, 1/12, Moscow 119899, Russia
| | | | - Mikhail Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho–Maklaya st. 16/10, Moscow 117997, Russia
- Biology Department, Lomonosov Moscow State University, Leninskie gory, 1/12, Moscow 119899, Russia
| |
Collapse
|
21
|
Rege NK, Liu M, Dhayalan B, Chen YS, Smith NA, Rahimi L, Sun J, Guo H, Yang Y, Haataja L, Phillips NFB, Whittaker J, Smith BJ, Arvan P, Ismail-Beigi F, Weiss MA. "Register-shift" insulin analogs uncover constraints of proteotoxicity in protein evolution. J Biol Chem 2020; 295:3080-3098. [PMID: 32005662 DOI: 10.1074/jbc.ra119.011389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Globular protein sequences encode not only functional structures (the native state) but also protein foldability, i.e. a conformational search that is both efficient and robustly minimizes misfolding. Studies of mutations associated with toxic misfolding have yielded insights into molecular determinants of protein foldability. Of particular interest are residues that are conserved yet dispensable in the native state. Here, we exploited the mutant proinsulin syndrome (a major cause of permanent neonatal-onset diabetes mellitus) to investigate whether toxic misfolding poses an evolutionary constraint. Our experiments focused on an invariant aromatic motif (PheB24-PheB25-TyrB26) with complementary roles in native self-assembly and receptor binding. A novel class of mutations provided evidence that insulin can bind to the insulin receptor (IR) in two different modes, distinguished by a "register shift" in this motif, as visualized by molecular dynamics (MD) simulations. Register-shift variants are active but defective in cellular foldability and exquisitely susceptible to fibrillation in vitro Indeed, expression of the corresponding proinsulin variant induced endoplasmic reticulum stress, a general feature of the mutant proinsulin syndrome. Although not present among vertebrate insulin and insulin-like sequences, a prototypical variant ([GlyB24]insulin) was as potent as WT insulin in a rat model of diabetes. Although in MD simulations the shifted register of receptor engagement is compatible with the structure and allosteric reorganization of the IR-signaling complex, our results suggest that this binding mode is associated with toxic misfolding and so is disallowed in evolution. The implicit threat of proteotoxicity limits sequence variation among vertebrate insulins and insulin-like growth factors.
Collapse
Affiliation(s)
- Nischay K Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Ming Liu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia; Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, Heping District, 300052 China
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Nicholas A Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Leili Rahimi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jinhong Sun
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Huan Guo
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Yanwu Yang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Leena Haataja
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Nelson F B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Brian J Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Peter Arvan
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| |
Collapse
|
22
|
Zheng N, Karra P, VandenBerg MA, Kim JH, Webber MJ, Holland WL, Chou DHC. Synthesis and Characterization of an A6-A11 Methylene Thioacetal Human Insulin Analogue with Enhanced Stability. J Med Chem 2019; 62:11437-11443. [PMID: 31804076 PMCID: PMC7217704 DOI: 10.1021/acs.jmedchem.9b01589] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin has been a life-saving drug for millions of people with diabetes. However, several challenges exist which limit therapeutic benefits and reduce patient convenience. One key challenge is the fibrillation propensity, which necessitates refrigeration for storage. To address this limitation, we chemically synthesized and evaluated a methylene thioacetal human insulin analogue (SCS-Ins). The synthesized SCS-Ins showed enhanced serum stability and aggregation resistance while retaining bioactivity compared with native insulin.
Collapse
Affiliation(s)
- Nan Zheng
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, United States
| | - Prasoona Karra
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, United States
| | - Michael A. VandenBerg
- Department of Chemical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Jin Hwan Kim
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, United States
| | - Matthew J. Webber
- Department of Chemical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - William L. Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, United States
| | - Danny Hung-Chieh Chou
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, United States
| |
Collapse
|
23
|
Xiong X, Blakely A, Karra P, VandenBerg MA, Ghabash G, Whitby F, Zhang YW, Webber MJ, Holland WL, Hill CP, Chou DHC. Novel four-disulfide insulin analog with high aggregation stability and potency. Chem Sci 2019; 11:195-200. [PMID: 32110371 PMCID: PMC7012051 DOI: 10.1039/c9sc04555d] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
A novel four-disulfide insulin analog was designed with retained bioactivity and increased fibrillation stability.
Although insulin was first purified and used therapeutically almost a century ago, there is still a need to improve therapeutic efficacy and patient convenience. A key challenge is the requirement for refrigeration to avoid inactivation of insulin by aggregation/fibrillation. Here, in an effort to mitigate this problem, we introduced a 4th disulfide bond between a C-terminal extended insulin A chain and residues near the C-terminus of the B chain. Insulin activity was retained by an analog with an additional disulfide bond between residues A22 and B22, while other linkages tested resulted in much reduced potency. Furthermore, the A22-B22 analog maintains the native insulin tertiary structure as demonstrated by X-ray crystal structure determination. We further demonstrate that this four-disulfide analog has similar in vivo potency in mice compared to native insulin and demonstrates higher aggregation stability. In conclusion, we have discovered a novel four-disulfide insulin analog with high aggregation stability and potency.
Collapse
Affiliation(s)
- Xiaochun Xiong
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | - Alan Blakely
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | - Prasoona Karra
- Department of Nutrition and Integrative Physiology , University of Utah , Salt Lake City UT 84112 , USA
| | - Michael A VandenBerg
- Department of Chemical & Biomolecular Engineering , University of Notre Dame , IN 46556 , USA
| | - Gabrielle Ghabash
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | - Frank Whitby
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | - Yi Wolf Zhang
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering , University of Notre Dame , IN 46556 , USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology , University of Utah , Salt Lake City UT 84112 , USA
| | - Christopher P Hill
- Department of Biochemistry , University of Utah , Salt Lake City UT 84112 , USA . ;
| | | |
Collapse
|
24
|
Mao R, Chen Y, Chi Z, Wang Y. Insulin and its single-chain analogue. Appl Microbiol Biotechnol 2019; 103:8737-8751. [PMID: 31637493 DOI: 10.1007/s00253-019-10170-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 12/26/2022]
|
25
|
Brunel FM, Mayer JP, Gelfanov VM, Zaykov AN, Finan B, Perez-Tilve D, DiMarchi RD. A Disulfide Scan of Insulin by [3 + 1] Methodology Exhibits Site-Specific Influence on Bioactivity. ACS Chem Biol 2019; 14:1829-1835. [PMID: 31343157 DOI: 10.1021/acschembio.9b00420] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Insulin is the principal hormone involved in the regulation of metabolism and has served a seminal role in the treatment of diabetes. Building upon advances in insulin synthetic methodology, we have developed a straightforward route to novel insulins containing a fourth disulfide bond in a [3 + 1] fashion establishing the first disulfide scan of the hormone. All the targeted analogs accommodated the constraint to demonstrate an unexpected conformational flexibility of native insulin. The bioactivity was established for the constrained (4-DS) and unconstrained (3-DS) analogs by in vitro methods, and extended to in vivo study for select peptides. We also identified residue B10 as a preferred anchor to introduce a tether that would regulate insulin bioactivity. We believe that the described [3 + 1] methodology might constitute the preferred approach for performing similar disulfide scanning in peptides that contain multiple disulfides.
Collapse
Affiliation(s)
- Florence M. Brunel
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - John P. Mayer
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - Vasily M. Gelfanov
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | | | - Brian Finan
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States
| | - Richard D. DiMarchi
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
- Department, of Chemistry,Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
26
|
Dec R, Koliński M, Dzwolak W. Beyond amino acid sequence: disulfide bonds and the origins of the extreme amyloidogenic properties of insulin's H‐fragment. FEBS J 2019; 286:3194-3205. [DOI: 10.1111/febs.14849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/08/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Robert Dec
- Faculty of Chemistry, Biological and Chemical Research Centre University of Warsaw Poland
| | - Michał Koliński
- Faculty of Chemistry, Biological and Chemical Research Centre University of Warsaw Poland
- Mossakowski Medical Research Centre Polish Academy of Sciences Bioinformatics Laboratory Warsaw Poland
| | - Wojciech Dzwolak
- Faculty of Chemistry, Biological and Chemical Research Centre University of Warsaw Poland
| |
Collapse
|
27
|
Salar S, Jafari M, Kaboli SF, Mehrnejad F. The role of intermolecular interactions on the encapsulation of human insulin into the chitosan and cholesterol-grafted chitosan polymers. Carbohydr Polym 2019; 208:345-355. [DOI: 10.1016/j.carbpol.2018.12.083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/04/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022]
|
28
|
Abramson A, Caffarel-Salvador E, Khang M, Dellal D, Silverstein D, Gao Y, Frederiksen MR, Vegge A, Hubálek F, Water JJ, Friderichsen AV, Fels J, Kirk RK, Cleveland C, Collins J, Tamang S, Hayward A, Landh T, Buckley ST, Roxhed N, Rahbek U, Langer R, Traverso G. An ingestible self-orienting system for oral delivery of macromolecules. Science 2019; 363:611-615. [PMID: 30733413 PMCID: PMC6430586 DOI: 10.1126/science.aau2277] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 01/04/2019] [Indexed: 12/22/2022]
Abstract
Biomacromolecules have transformed our capacity to effectively treat diseases; however, their rapid degradation and poor absorption in the gastrointestinal (GI) tract generally limit their administration to parenteral routes. An oral biologic delivery system must aid in both localization and permeation to achieve systemic drug uptake. Inspired by the leopard tortoise's ability to passively reorient, we developed an ingestible self-orienting millimeter-scale applicator (SOMA) that autonomously positions itself to engage with GI tissue. It then deploys milliposts fabricated from active pharmaceutical ingredients directly through the gastric mucosa while avoiding perforation. We conducted in vivo studies in rats and swine that support the applicator's safety and, using insulin as a model drug, demonstrated that the SOMA delivers active pharmaceutical ingredient plasma levels comparable to those achieved with subcutaneous millipost administration.
Collapse
Affiliation(s)
- Alex Abramson
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ester Caffarel-Salvador
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Minsoo Khang
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David Dellal
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David Silverstein
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuan Gao
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Andreas Vegge
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - František Hubálek
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Jorrit J Water
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Anders V Friderichsen
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Johannes Fels
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Rikke Kaae Kirk
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Cody Cleveland
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Joy Collins
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Siddartha Tamang
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alison Hayward
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomas Landh
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Stephen T Buckley
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Niclas Roxhed
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ulrik Rahbek
- Global Research Technologies, Global Drug Discovery, and Device R&D, Novo Nordisk A/S, Copenhagen, Denmark
| | - Robert Langer
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Media Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
29
|
Siposova K, Sedlak E, Kozar T, Nemergut M, Musatov A. Dual effect of non-ionic detergent Triton X-100 on insulin amyloid formation. Colloids Surf B Biointerfaces 2018; 173:709-718. [PMID: 30384267 DOI: 10.1016/j.colsurfb.2018.10.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 10/28/2022]
Abstract
Atomic force microscopy, Thioflavin T (ThT) fluorescence assay, circular dichroism spectroscopy, differential scanning calorimetry, and molecular modeling techniques have been employed to investigate the amyloid aggregation of insulin in the presence of non-ionic detergent, Triton X-100 (TX-100). In contrast to recently described inhibition of lysozyme amyloid formation by non-ionic detergents (Siposova, 2017), the amyloid aggregation of insulin in the presence of sub-micellar TX-100 concentration exhibits two dissimilar phases. The first, inhibition phase, is observed at the protein to detergent molar ratio of 1:0.1 to 1:1. During this phase, the insulin amyloid fibril formation is inhibited by TX-100 up to ∼60%. The second, "morphological" phase, is observed at increasing detergent concentration, corresponding to protein:detergent molar ratio of ∼1:1 - 1:10. Under these conditions a significant increase of the steady-state ThT fluorescence intensities and a dramatically changed morphology of the insulin fibrils were observed. Increasing of the detergent concentration above the CMC led to complete inhibition of amyloidogenesis. Analysis of the experimental and molecular modeling results suggests an existence of up to six TX-100 binding sites within dimer of insulin with different binding energy. The physiological relevance of the results is discussed.
Collapse
Affiliation(s)
- Katarina Siposova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01, Kosice, Slovakia.
| | - Erik Sedlak
- Center for Interdisciplinary Biosciences, TIP - P.J. Safarik University, Jesenna 5, 041 54, Kosice, Slovakia
| | - Tibor Kozar
- Center for Interdisciplinary Biosciences, TIP - P.J. Safarik University, Jesenna 5, 041 54, Kosice, Slovakia; Laboratory of Information Technologies, Joint Institute for Nuclear Research, Joliot-Curie 6, 141980, Dubna, Moscow Region, Russia
| | - Michal Nemergut
- Department of Biophysics, Faculty of Science, P.J. Safarik University, Jesenna 5, 041 54, Kosice, Slovakia
| | - Andrey Musatov
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01, Kosice, Slovakia
| |
Collapse
|
30
|
Synthesis of disulfide-rich heterodimeric peptides through an auxiliary N, N-crosslink. Commun Chem 2018. [DOI: 10.1038/s42004-018-0036-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
31
|
Hong Q, Zhou S, Zhao H, Peng J, Li Y, Shang Y, Wu M, Zhang W, Lu S, Li S, Yu S, Wang W, Wang Q. Allergenicity of recombinant Humulus japonicus pollen allergen 1 after combined exposure to ozone and nitrogen dioxide. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 234:707-715. [PMID: 29241157 DOI: 10.1016/j.envpol.2017.11.078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/07/2017] [Accepted: 11/24/2017] [Indexed: 06/07/2023]
Abstract
Ozone (O3) and nitrogen dioxide (NO2) are thought to play primary roles in aggravating air pollution-induced health problems. However, the effects of joint O3/NO2 on the allergenicity of pollen allergens are unclear. Humulus japonicus pollen allergen 1 (Hum j1) is a profilin protein that causes widespread pollinosis in eastern Asia. In order to study the effects of combined O3/NO2 on the allergenicity of Hum j1, tandem six-histidine peptide tag (His6)-fused recombinant Hum j1 (rHum j1) was expressed in a prokaryotic system and purified through His6 affinity chromatography. The purified rHum j1 was used to immunize SD rats. Rat sera with high titers of IgG and IgE antibodies against rHum j1 were used for allergenicity quantification. The rHum j1 was exposed to O3/NO2, and changes in allergenicity of the exposed rHum j1 were assayed using the immunized rat antibodies. Tandem LC-MS/LC (liquid chromatography-mass spectrometer/liquid chromatography spectrometer) chromatography and UV and circular dichroism (CD) spectroscopy were used to study the structural changes in rHum j1. Our data demonstrated that a novel disulfide bond between the sulfhydryl groups of two neighboring cysteine molecules was formed after the rHum j1 exposure to joint O3/NO2, and therefore IgE-binding affinity was increased and the allergenicity was reinforced. Our results provided clues to elucidate the mechanism behind air pollution-induced increase in pollinosis prevalence.
Collapse
Affiliation(s)
- Qiang Hong
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Shumin Zhou
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Hui Zhao
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Jiaxian Peng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yang Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yu Shang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Minghong Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wei Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Senlin Lu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Shuijun Li
- Shanghai Xuhui Center Hospital, Shanghai 200031, China
| | - Shen Yu
- Shanghai Xuhui Center Hospital, Shanghai 200031, China
| | - Weiqian Wang
- School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Qingyue Wang
- School of Science and Engineering, Saitama University, Saitama 338-8570, Japan.
| |
Collapse
|
32
|
Glidden MD, Yang Y, Smith NA, Phillips NB, Carr K, Wickramasinghe NP, Ismail-Beigi F, Lawrence MC, Smith BJ, Weiss MA. Solution structure of an ultra-stable single-chain insulin analog connects protein dynamics to a novel mechanism of receptor binding. J Biol Chem 2018; 293:69-88. [PMID: 29114034 PMCID: PMC5766920 DOI: 10.1074/jbc.m117.808667] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
Domain-minimized insulin receptors (IRs) have enabled crystallographic analysis of insulin-bound "micro-receptors." In such structures, the C-terminal segment of the insulin B chain inserts between conserved IR domains, unmasking an invariant receptor-binding surface that spans both insulin A and B chains. This "open" conformation not only rationalizes the inactivity of single-chain insulin (SCI) analogs (in which the A and B chains are directly linked), but also suggests that connecting (C) domains of sufficient length will bind the IR. Here, we report the high-resolution solution structure and dynamics of such an active SCI. The hormone's closed-to-open transition is foreshadowed by segmental flexibility in the native state as probed by heteronuclear NMR spectroscopy and multiple conformer simulations of crystallographic protomers as described in the companion article. We propose a model of the SCI's IR-bound state based on molecular-dynamics simulations of a micro-receptor complex. In this model, a loop defined by the SCI's B and C domains encircles the C-terminal segment of the IR α-subunit. This binding mode predicts a conformational transition between an ultra-stable closed state (in the free hormone) and an active open state (on receptor binding). Optimization of this switch within an ultra-stable SCI promises to circumvent insulin's complex global cold chain. The analog's biphasic activity, which serendipitously resembles current premixed formulations of soluble insulin and microcrystalline suspension, may be of particular utility in the developing world.
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
| | - Yanwu Yang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nicholas A Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - 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
| | | | - 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 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
| | - Brian J Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
33
|
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: 28] [Impact Index Per Article: 4.0] [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
|
34
|
Liu F, Li P, Gelfanov V, Mayer J, DiMarchi R. Synthetic Advances in Insulin-like Peptides Enable Novel Bioactivity. Acc Chem Res 2017; 50:1855-1865. [PMID: 28771323 DOI: 10.1021/acs.accounts.7b00227] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Insulin is a miraculous hormone that has served a seminal role in the treatment of insulin-dependent diabetes for nearly a century. Insulin resides within in a superfamily of structurally related peptides that are distinguished by three invariant disulfide bonds that anchor the three-dimensional conformation of the hormone. The additional family members include the insulin-like growth factors (IGF) and the relaxin-related set of peptides that includes the so-called insulin-like peptides. Advances in peptide chemistry and rDNA-based synthesis have enabled the preparation of multiple insulin analogues. The translation of these methods from insulin to related peptides has presented unique challenges that pertain to differing biophysical properties and unique amino acid compositions. This Account presents a historical context for the advances in the chemical synthesis of insulin and the related peptides, with division into two general categories where disulfide bond formation is facilitated by native conformational folding or alternatively orthogonal chemical reactivity. The inherent differences in biophysical properties of insulin-like peptides, and in particular within synthetic intermediates, have constituted a central limitation to achieving high yield synthesis of properly folded peptides. Various synthetic approaches have been advanced in the past decade to successfully address this challenge. The use of chemical ligation and metastable amide bond surrogates are two of the more important synthetic advances in the preparation of high quality synthetic precursors to high potency peptides. The discovery and application of biomimetic connecting peptides simplifies proper disulfide formation and the subsequent traceless removal by chemical methods dramatically simplifies the total synthesis of virtually any two-chain insulin-like peptide. We report the application of these higher synthetic yield methodologies to the preparation of insulin-like peptides in support of exploratory in vivo studies requiring a large quantity of peptide. Tangentially, we demonstrate the use of these methods to study the relative importance of the IGF-1 connecting peptide to its biological activity. We report the translation of these finding in search of an insulin analog that might be comparably enhanced by a suitable connecting peptide for interaction with the insulin receptor, as occurs with IGF-1 and its receptor. The results identify a unique receptor site in the IGF-1 receptor from which this enhancement derives. The selective substitution of this specific IGF-1 receptor sequence into the homologous site in the insulin receptor generated a chimeric receptor that was equally capable of signaling with insulin or IGF-1. This novel receptor proved to enhance the potency of lower affinity insulin ligands when they were supplemented with the IGF-1 connecting peptide that similarly enhanced IGF-1 activity at its receptor. The chimeric insulin receptor demonstrated no further enhancement of potency for native insulin when it was similarly prepared as a single-chain analogue with a native IGF-1 connecting peptide. These results suggest a more highly evolved insulin receptor structure where the requirement for an additional structural element to achieve high potency interaction as demonstrated for IGF-1 is no longer required.
Collapse
Affiliation(s)
- Fa Liu
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Pengyun Li
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Vasily Gelfanov
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - John Mayer
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana 46241, United States
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
35
|
Holub JM. Small Scaffolds, Big Potential: Developing Miniature Proteins as Therapeutic Agents. Drug Dev Res 2017; 78:268-282. [PMID: 28799168 DOI: 10.1002/ddr.21408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Preclinical Research Miniature proteins are a class of oligopeptide characterized by their short sequence lengths and ability to adopt well-folded, three-dimensional structures. Because of their biomimetic nature and synthetic tractability, miniature proteins have been used to study a range of biochemical processes including fast protein folding, signal transduction, catalysis and molecular transport. Recently, miniature proteins have been gaining traction as potential therapeutic agents because their small size and ability to fold into defined tertiary structures facilitates their development as protein-based drugs. This research overview discusses emerging developments involving the use of miniature proteins as scaffolds to design novel therapeutics for the treatment and study of human disease. Specifically, this review will explore strategies to: (i) stabilize miniature protein tertiary structure; (ii) optimize biomolecular recognition by grafting functional epitopes onto miniature protein scaffolds; and (iii) enhance cytosolic delivery of miniature proteins through the use of cationic motifs that facilitate endosomal escape. These objectives are discussed not only to address challenges in developing effective miniature protein-based drugs, but also to highlight the tremendous potential miniature proteins hold for combating and understanding human disease. Drug Dev Res 78 : 268-282, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The complexity of modern insulin-based therapy for type I and type II diabetes mellitus and the risks associated with excursions in blood-glucose concentration (hyperglycemia and hypoglycemia) have motivated the development of 'smart insulin' technologies (glucose-responsive insulin, GRI). Such analogs or delivery systems are entities that provide insulin activity proportional to the glycemic state of the patient without external monitoring by the patient or healthcare provider. The present review describes the relevant historical background to modern GRI technologies and highlights three distinct approaches: coupling of continuous glucose monitoring (CGM) to deliver devices (algorithm-based 'closed-loop' systems), glucose-responsive polymer encapsulation of insulin, and molecular modification of insulin itself. RECENT FINDINGS Recent advances in GRI research utilizing each of the three approaches are illustrated; these include newly developed algorithms for CGM-based insulin delivery systems, glucose-sensitive modifications of existing clinical analogs, newly developed hypoxia-sensitive polymer matrices, and polymer-encapsulated, stem-cell-derived pancreatic β cells. SUMMARY Although GRI technologies have yet to be perfected, the recent advances across several scientific disciplines that are described in this review have provided a path towards their clinical implementation.
Collapse
Affiliation(s)
- Nischay K. Rege
- Department of Biochemistry and Medical Scientist Training Program, Case Western Reserve University
| | | | - Michael A. Weiss
- Chairman of Institute for Therapeutic Protein Design, Departments of Biomedical Engineering, Biochemistry, and Medicine
| |
Collapse
|
37
|
Enhancing thermal stability of a highly concentrated insulin formulation with Pluronic F-127 for long-term use in microfabricated implantable devices. Drug Deliv Transl Res 2017; 7:529-543. [DOI: 10.1007/s13346-017-0381-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
38
|
Wu F, Mayer JP, Gelfanov VM, Liu F, DiMarchi RD. Synthesis of Four-Disulfide Insulin Analogs via Sequential Disulfide Bond Formation. J Org Chem 2017; 82:3506-3512. [DOI: 10.1021/acs.joc.6b03078] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Fangzhou Wu
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - John P. Mayer
- Novo Nordisk
Research
Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Vasily M. Gelfanov
- Novo Nordisk
Research
Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Fa Liu
- Novo Nordisk
Research
Center Indianapolis, Indianapolis, Indiana 46241, United States
| | - Richard D. DiMarchi
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
- Novo Nordisk
Research
Center Indianapolis, Indianapolis, Indiana 46241, United States
| |
Collapse
|
39
|
Pechenov S, Bhattacharjee H, Yin D, Mittal S, Subramony JA. Improving drug-like properties of insulin and GLP-1 via molecule design and formulation and improving diabetes management with device & drug delivery. Adv Drug Deliv Rev 2017; 112:106-122. [PMID: 28153578 DOI: 10.1016/j.addr.2017.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 01/20/2017] [Accepted: 01/25/2017] [Indexed: 12/25/2022]
Abstract
There is an increased incidence of diabetes worldwide. The discovery of insulin revolutionized the management of diabetes, the revelation of glucagon-like peptide-1 (GLP-1) and introduction of GLP-1 receptor agonists to clinical practice was another breakthrough. Continued translational research resulted in better understanding of diabetes, which, in combination with cutting-edge biology, chemistry, and pharmaceutical tools, have allowed for the development of safer, more effective and convenient insulins and GLP-1. Advances in self-administration of insulin and GLP-1 receptor agonist therapies with use of drug-device combination products have further improved the outcomes of diabetes management and quality of life for diabetic patients. The synergies of insulin and GLP-1 receptor agonist actions have led to development of devices that can deliver both molecules simultaneously. New chimeric GLP-1-incretins and insulin-GLP-1-incretin molecules are also being developed. The objective of this review is to summarize molecular designs to improve the drug-like properties of insulin and GLP-1 and to highlight the continued advancement of drug-device combination products to improve diabetes management.
Collapse
Affiliation(s)
| | - Himanshu Bhattacharjee
- Merck Research Laboratories, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Daniel Yin
- Merck Research Laboratories, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Sachin Mittal
- Merck Research Laboratories, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | | |
Collapse
|
40
|
Schlein M. Insulin Formulation Characterization-the Thioflavin T Assays. AAPS JOURNAL 2016; 19:397-408. [PMID: 28000098 DOI: 10.1208/s12248-016-0028-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/08/2016] [Indexed: 11/30/2022]
Abstract
The insulin molecule was discovered in 1921. Shortly thereafter, its propensity towards amyloid fibril formation, fibrillation, was observed and described in the literature as a "precipitate." In the past decades, the increased incidence of type 2 diabetes has reached global epidemic proportions. This has emphasized the demands for both insulin production and the development of modern insulin products for unmet medical needs. Bringing such new insulin drug products to the market for the benefit of patients requires that many CMC-related processes are understood, described, and controlled. One potential undesired process is insulin fibril formation. The compound thioflavin T (ThT) is known as a fluorescent probe for amyloid fibrils. As such, ThT is utilized in a versatile research assay in microtiter plate format, the ThT assay. This review will describe an experimental set-up using not only a ThT microtiter plate assay but also two orthogonal methods. The use of the ThT assay in research and characterization of insulin analogues, as well as formulations of insulin, is described by cases drawn from the scientific literature and patents. The ThT assay is compared to other physical stability tests and in conclusion the advantages and limitations of the assay are compared.
Collapse
Affiliation(s)
- Morten Schlein
- Injectable Formulation Research, Global Research, Novo Nordisk A/S, Novo Nordisk Park H6.S.09.1, DK2760, Maaloev, Denmark.
| |
Collapse
|
41
|
Mills BJ, Laurence Chadwick JS. Effects of localized interactions and surface properties on stability of protein-based therapeutics. ACTA ACUST UNITED AC 2016; 70:609-624. [PMID: 27861887 DOI: 10.1111/jphp.12658] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/04/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Protein-based therapeutics garner significant attention because of exquisite specificity and limited side effects and are now being used to accomplish targeted delivery of small-molecule drugs. This review identifies and highlights individual chemical attributes and categorizes how site-specific changes affect protein stability based on published high-resolution molecular analyses. KEY FINDINGS Because it is challenging to determine the mechanisms by which the stability of large, complex molecules is altered and data are sparse, smaller, therapeutic proteins (insulin, erythropoietin, interferons) are examined alongside antibody data. Integrating this large pool of information with the limited available studies on antibodies reveals common mechanisms by which specific alterations affect protein structure and stability. SUMMARY Physical and chemical stability of therapeutic proteins and antibody drug conjugates (ADCs) is of critical importance because insufficient stability prevents molecules from making it to market. Individual moieties on/near the surface of proteins have substantial influence on structure and stability. Seemingly small, superficial modification may have far-reaching consequences on structure, conformational dynamics, and solubility of the protein, and hence physical stability of the molecule. Chemical modifications, whether spontaneous (e.g. oxidation, deamidation) or intentional, as with ADCs, may adversely impact stability by disrupting local surface properties or higher order protein structure.
Collapse
Affiliation(s)
- Brittney J Mills
- Department of Chemistry, The University of Kansas, Lawrence, KS, USA
| | - Jennifer S Laurence Chadwick
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA.,BioAnalytix Inc., Cambridge, MA, USA
| |
Collapse
|
42
|
Karas JA, Patil NA, Tailhades J, Sani MA, Scanlon DB, Forbes BE, Gardiner J, Separovic F, Wade JD, Hossain MA. Total Chemical Synthesis of an Intra-A-Chain Cystathionine Human Insulin Analogue with Enhanced Thermal Stability. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201607101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- John A. Karas
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne VIC 3010 Australia
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
- CSIRO; Materials Science and Engineering; Clayton VIC 3010 Australia
| | - Nitin A. Patil
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne VIC 3010 Australia
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
| | - Julien Tailhades
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne VIC 3010 Australia
| | - Marc-Antoine Sani
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
| | - Denis B. Scanlon
- Department of Chemistry; University of Adelaide; Adelaide SA 5005 Australia
| | - Briony E. Forbes
- School of Medicine; Flinders University; Bedford Park SA 5042 Australia
| | - James Gardiner
- CSIRO; Materials Science and Engineering; Clayton VIC 3010 Australia
| | - Frances Separovic
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
| | - John D. Wade
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne VIC 3010 Australia
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
| | - Mohammed Akhter Hossain
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne VIC 3010 Australia
- School of Chemistry; Bio21 Institute; University of Melbourne; Melbourne VIC 3010 Australia
| |
Collapse
|
43
|
Karas JA, Patil NA, Tailhades J, Sani MA, Scanlon DB, Forbes BE, Gardiner J, Separovic F, Wade JD, Hossain MA. Total Chemical Synthesis of an Intra-A-Chain Cystathionine Human Insulin Analogue with Enhanced Thermal Stability. Angew Chem Int Ed Engl 2016; 55:14743-14747. [PMID: 27761974 DOI: 10.1002/anie.201607101] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Indexed: 12/17/2022]
Abstract
Despite recent advances in the treatment of diabetes mellitus, storage of insulin formulations at 4 °C is still necessary to minimize chemical degradation. This is problematic in tropical regions where reliable refrigeration is not ubiquitous. Some degradation byproducts are caused by disulfide shuffling of cystine that leads to covalently bonded oligomers. Consequently we examined the utility of the non-reducible cystine isostere, cystathionine, within the A-chain. Reported herein is an efficient method for forming this mimic using simple monomeric building blocks. The intra-A-chain cystathionine insulin analogue was obtained in good overall yield, chemically characterized and demonstrated to possess native binding affinity for the insulin receptor isoform B. It was also shown to possess significantly enhanced thermal stability indicating potential application to next-generation insulin analogues.
Collapse
Affiliation(s)
- John A Karas
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia.,CSIRO, Materials Science and Engineering, Clayton, VIC, 3010, Australia
| | - Nitin A Patil
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Julien Tailhades
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Marc-Antoine Sani
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Denis B Scanlon
- Department of Chemistry, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Briony E Forbes
- School of Medicine, Flinders University, Bedford Park, SA, 5042, Australia
| | - James Gardiner
- CSIRO, Materials Science and Engineering, Clayton, VIC, 3010, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - John D Wade
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Mohammed Akhter Hossain
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
44
|
Switzar L, Nicolardi S, Rutten JW, Oberstein SAJL, Aartsma-Rus A, van der Burgt YEM. In-Depth Characterization of Protein Disulfide Bonds by Online Liquid Chromatography-Electrochemistry-Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:50-8. [PMID: 26369777 PMCID: PMC4686567 DOI: 10.1007/s13361-015-1258-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 05/04/2023]
Abstract
Disulfide bonds are an important class of protein post-translational modifications, yet this structurally crucial modification type is commonly overlooked in mass spectrometry (MS)-based proteomics approaches. Recently, the benefits of online electrochemistry-assisted reduction of protein S-S bonds prior to MS analysis were exemplified by successful characterization of disulfide bonds in peptides and small proteins. In the current study, we have combined liquid chromatography (LC) with electrochemistry (EC) and mass analysis by Fourier transform ion cyclotron resonance (FTICR) MS in an online LC-EC-MS platform to characterize protein disulfide bonds in a bottom-up proteomics workflow. A key advantage of a LC-based strategy is the use of the retention time in identifying both intra- and interpeptide disulfide bonds. This is demonstrated by performing two sequential analyses of a certain protein digest, once without and once with electrochemical reduction. In this way, the "parent" disulfide-linked peptide detected in the first run has a retention time-based correlation with the EC-reduced peptides detected in the second run, thus simplifying disulfide bond mapping. Using this platform, both inter- and intra-disulfide-linked peptides were characterized in two different proteins, ß-lactoglobulin and ribonuclease B. In order to prevent disulfide reshuffling during the digestion process, proteins were digested at a relatively low pH, using (a combination of) the high specificity proteases trypsin and Glu-C. With this approach, disulfide bonds in ß-lactoglobulin and ribonuclease B were comprehensively identified and localized, showing that online LC-EC-MS is a useful tool for the characterization of protein disulfide bonds.
Collapse
Affiliation(s)
- Linda Switzar
- Department of Human Genetics, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
- , Albinusdreef 2, Postzone S3, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Julie W Rutten
- Department of Human Genetics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | | | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Yuri E M van der Burgt
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
45
|
Vinther TN, Kjeldsen TB, Jensen KJ, Hubálek F. The road to the first, fully active and more stable human insulin variant with an additional disulfide bond. J Pept Sci 2015; 21:797-806. [DOI: 10.1002/psc.2822] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/14/2015] [Accepted: 08/19/2015] [Indexed: 12/21/2022]
Affiliation(s)
| | | | - Knud J. Jensen
- Faculty of Science, Department of Chemistry; University of Copenhagen; DK-1871 Frederiksberg Denmark
| | | |
Collapse
|
46
|
Vinther TN, Pettersson I, Huus K, Schlein M, Steensgaard DB, Sørensen A, Jensen KJ, Kjeldsen T, Hubalek F. Additional disulfide bonds in insulin: Prediction, recombinant expression, receptor binding affinity, and stability. Protein Sci 2015; 24:779-88. [PMID: 25627966 PMCID: PMC4420526 DOI: 10.1002/pro.2649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/26/2015] [Indexed: 11/07/2022]
Abstract
The structure of insulin, a glucose homeostasis-controlling hormone, is highly conserved in all vertebrates and stabilized by three disulfide bonds. Recently, we designed a novel insulin analogue containing a fourth disulfide bond located between positions A10-B4. The N-terminus of insulin's B-chain is flexible and can adapt multiple conformations. We examined how well disulfide bond predictions algorithms could identify disulfide bonds in this region of insulin. In order to identify stable insulin analogues with additional disulfide bonds, which could be expressed, the Cβ cut-off distance had to be increased in many instances and single X-ray structures as well as structures from MD simulations had to be used. The analogues that were identified by the algorithm without extensive adjustments of the prediction parameters were more thermally stable as assessed by DSC and CD and expressed in higher yields in comparison to analogues with additional disulfide bonds that were more difficult to predict. In contrast, addition of the fourth disulfide bond rendered all analogues resistant to fibrillation under stress conditions and all stable analogues bound to the insulin receptor with picomolar affinities. Thus activity and fibrillation propensity did not correlate with the results from the prediction algorithm. Statement: A fourth disulfide bond has recently been introduced into insulin, a small two-chain protein containing three native disulfide bonds. Here we show that a prediction algorithm predicts four additional four disulfide insulin analogues which could be expressed. Although the location of the additional disulfide bonds is only slightly shifted, this shift impacts both stability and activity of the resulting insulin analogues.
Collapse
Affiliation(s)
- Tine N Vinther
- Diabetes Research UnitNovo Nordisk A/S, DK-2760, Måløv, Denmark
| | | | - Kasper Huus
- Diabetes Research UnitNovo Nordisk A/S, DK-2760, Måløv, Denmark
| | - Morten Schlein
- Diabetes Research UnitNovo Nordisk A/S, DK-2760, Måløv, Denmark
| | | | - Anders Sørensen
- Diabetes Research UnitNovo Nordisk A/S, DK-2760, Måløv, Denmark
| | - Knud J Jensen
- Department of Chemistry, Faculty of Science, University of CopenhagenDK-1871, Frederiksberg, Denmark
| | - Thomas Kjeldsen
- Diabetes Research UnitNovo Nordisk A/S, DK-2760, Måløv, Denmark
| | | |
Collapse
|
47
|
Yang H, Liu L, Li J, Chen J, Du G. Rational Design to Improve Protein Thermostability: Recent Advances and Prospects. CHEMBIOENG REVIEWS 2015. [DOI: 10.1002/cben.201400032] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
48
|
Pitman DJ, Banerjee S, Macari SJ, Castaldi CA, Crone DE, Bystroff C. Exploring the folding pathway of green fluorescent protein through disulfide engineering. Protein Sci 2015; 24:341-53. [PMID: 25516354 PMCID: PMC4353360 DOI: 10.1002/pro.2621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/09/2014] [Indexed: 11/12/2022]
Abstract
We have introduced two disulfide crosslinks into the loop regions on opposite ends of the beta barrel in superfolder green fluorescent protein (GFP) in order to better understand the nature of its folding pathway. When the disulfide on the side opposite the N/C-termini is formed, folding is 2× faster, unfolding is 2000× slower, and the protein is stabilized by 16 kJ/mol. But when the disulfide bond on the side of the termini is formed we see little change in the kinetics and stability. The stabilization upon combining the two crosslinks is approximately additive. When the kinetic effects are broken down into multiple phases, we observe Hammond behavior in the upward shift of the kinetic m-value of unfolding. We use these results in conjunction with structural analysis to assign folding intermediates to two parallel folding pathways. The data are consistent with a view that the two fastest transition states of folding are "barrel closing" steps. The slower of the two phases passes through an intermediate with the barrel opening occurring between strands 7 and 8, while the faster phase opens between 9 and 4. We conclude that disulfide crosslink-induced perturbations in kinetics are useful for mapping the protein folding pathway.
Collapse
Affiliation(s)
- Derek J Pitman
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
| | - Shounak Banerjee
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
| | - Stephen J Macari
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
| | - Christopher A Castaldi
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
| | - Donna E Crone
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
| | - Christopher Bystroff
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies
- Department of Computer Science, Rensselaer Polytechnic InstituteTroy, New York
| |
Collapse
|
49
|
Bouvier ES, Koza SM. Advances in size-exclusion separations of proteins and polymers by UHPLC. Trends Analyt Chem 2014. [DOI: 10.1016/j.trac.2014.08.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
50
|
Thompson JM, Di Gregorio A. Insulin-like genes in ascidians: findings in Ciona and hypotheses on the evolutionary origins of the pancreas. Genesis 2014; 53:82-104. [PMID: 25378051 DOI: 10.1002/dvg.22832] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 10/13/2014] [Accepted: 10/16/2014] [Indexed: 12/22/2022]
Abstract
Insulin plays an extensively characterized role in the control of sugar metabolism, growth and homeostasis in a wide range of organisms. In vertebrate chordates, insulin is mainly produced by the beta cells of the endocrine pancreas, while in non-chordate animals insulin-producing cells are mainly found in the nervous system and/or scattered along the digestive tract. However, recent studies have indicated the notochord, the defining feature of the chordate phylum, as an additional site of expression of insulin-like peptides. Here we show that two of the three insulin-like genes identified in Ciona intestinalis, an invertebrate chordate with a dual life cycle, are first expressed in the developing notochord during embryogenesis and transition to distinct areas of the adult digestive tract after metamorphosis. In addition, we present data suggesting that the transcription factor Ciona Brachyury is involved in the control of notochord expression of at least one of these genes, Ciona insulin-like 2. Finally, we review the information currently available on insulin-producing cells in ascidians and on pancreas-related transcription factors that might control their expression.
Collapse
Affiliation(s)
- Jordan M Thompson
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, New York
| | | |
Collapse
|