1
|
Pahal S, Huang F, Singh P, Sharma N, Pham HP, Tran TBT, Sakhrie A, Akbaba H, Duc Nguyen T. Enhancing vaccine stability in transdermal microneedle platforms. Drug Deliv Transl Res 2025:10.1007/s13346-025-01854-4. [PMID: 40240731 DOI: 10.1007/s13346-025-01854-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Micron-scale needles, so-called microneedles (MNs) offer a minimally invasive, nearly painless, and user-friendly method for effective intradermal immunization. Maintaining the stability of antigens and therapeutics is the primary challenge in producing vaccine or drug-loaded MNs. The manufacturing of MNs patches involves processes at ambient or higher temperatures and various physio-mechanical stresses that can impact the therapeutic efficacy of sensitive biologics or vaccines. Therefore, it is crucial to develop techniques that safeguard vaccines and other biological payloads within MNs. Despite growing research interest in deploying MNs as an efficient tool for delivering vaccines, there is no comprehensive review that integrates the strategies and efforts to preserve the thermostability of vaccine payloads to ensure compatibility with MNs fabrication. The discussion delves into various physical and chemical approaches for stabilizing antigens in vaccine formulations, which are subsequently integrated into the MNs matrix. The primary focus is to comprehensively examine the challenges associated with the translation of thermostable vaccine MNs for clinical applications while considering a safe, cost-effective approach with a regulatory roadmap. The recent cutting-edge advances facilitating flexible and scalable manufacturing of stabilized MNs patches have been emphasized. In conclusion, the ability to stabilize vaccines and therapeutics for MNs applications could bolster the effectiveness, safety and user-compliance for various drugs and vaccines, potentially offering a substantial impact on global public health.
Collapse
Affiliation(s)
- Suman Pahal
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Feifei Huang
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
| | - Parbeen Singh
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Nidhi Sharma
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Hoang-Phuc Pham
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
| | - Thi Bao Tram Tran
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Aseno Sakhrie
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Hasan Akbaba
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir, 35100, Turkey
| | - Thanh Duc Nguyen
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
2
|
Wu X, Zhang Y, Song S, Liu S, Ma F, Ma R, Shi L. Functional nanochaperones for PEGylated insulin delivery in long-term glycemic control. Biomater Sci 2024; 12:5742-5752. [PMID: 39382287 DOI: 10.1039/d4bm01163e] [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: 10/10/2024]
Abstract
PEGylation is a promising strategy for modulating the physicochemical properties and improving the therapeutic efficacy of protein drugs. However, the application of multi-PEGylation frequently results in diminished protein activity. A single low molecular weight PEG (5 kDa) modified at the amino terminus of the B chain preserves the biological activity of insulin and moderately improves its pharmacokinetics. Nonetheless, this modification offers limited protein stabilization. Furthermore, overdoses still carry the risk of hypoglycemia, posing challenges for the clinical application of PEGylated insulin. Here, we constructed multifunctional nanochaperones featuring phenylboronic acid (PBA) modified hydrophobic microdomains and nitrilotriacetic acid (NTA)-based coordination domains (PN-nChaps) for PEGylated insulin delivery. This delivery strategy effectively overcomes the limitations associated with PEGylation by enhancing the stability and reducing the immunogenicity of PEGylated insulin, while enabling glucose-responsive controlled release. PEGylated insulin with nanochaperone carrier demonstrates a prolonged half-life (t1/2 = 18.66 h), facilitates on-demand release, and minimizes the risk of hypoglycemia. This approach provides a safe and effective strategy for long-term glycemic management in diabetic patients.
Collapse
Affiliation(s)
- Xiaohui Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| | - Yanli Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| | - Shuoshuo Song
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| | - Sainan Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| | - Feihe Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Rujiang Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, PR China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| |
Collapse
|
3
|
Grishin SY, Surin AK, Galzitskaya OV. Investigation of new non-toxic inhibitors of fibril formation and preservatives for insulin preparations its analogues. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 145:113-143. [PMID: 40324845 DOI: 10.1016/bs.apcsb.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Insulin and its analogue formulations are the main components in the therapy of insulin-dependent forms of diabetes. Insulin and its analogues can form amyloid-like aggregates during long-term storage and local concentration increases, leading to reduced therapeutic efficacy and potential side effects such as insulin amyloidosis. The aim of this study was to identify and propose new non-toxic inhibitors and preservatives to replace phenol in insulin formulations. The peptide FVNQH and phenol red were studied as promising inhibitors of fibril formation of insulin, lispro, and glargine in vitro using the specific amyloid dye thioflavin T. The peptide FVNQH and phenol red (0.5-1 mg/mL) showed a bacteriostatic effect on the E. coli K-12 strain after 24 h. The fibril-inhibiting and antimicrobial effects of these substances were similar to the effect of phenol at a concentration of 0.5 mg/mL. Thus, the identified inhibitors can potentially replace phenolic components in slowing amyloid aggregation and increase the stability of insulin and its analogues.
Collapse
Affiliation(s)
- Sergei Y Grishin
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia
| | - Alexey K Surin
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia; Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia; State Research Center for Applied Microbiology and Biotechnology, Obolensk, Russia
| | - Oxana V Galzitskaya
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia; Gamaleya Research Centre of Epidemiology and Microbiology, Moscow, Russia.
| |
Collapse
|
4
|
Liu Q, Yu Y, Wei G. Oncogenic R248W mutation induced conformational perturbation of the p53 core domain and the structural protection by proteomimetic amyloid inhibitor ADH-6. Phys Chem Chem Phys 2024; 26:20068-20086. [PMID: 39007865 DOI: 10.1039/d4cp02046d] [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: 07/16/2024]
Abstract
The involvement of p53 aggregation in cancer pathogenesis emphasizes the importance of unraveling the mechanisms underlying mutation-induced p53 destabilization. And understanding how small molecule inhibitors prevent the conversion of p53 into aggregation-primed conformations is pivotal for the development of therapeutics targeting p53-aggregation-associated cancers. A recent experimental study highlights the efficacy of the proteomimetic amyloid inhibitor ADH-6 in stabilizing R248W p53 and inhibiting its aggregation in cancer cells by interacting with the p53 core domain (p53C). However, it remains mostly unclear how R248W mutation induces destabilization of p53C and how ADH-6 stabilizes this p53C mutant and inhibits its aggregation. Herein, we conducted all-atom molecular dynamics simulations of R248W p53C in the absence and presence of ADH-6, as well as that of wild-type (WT) p53C. Our simulations reveal that the R248W mutation results in a shift of helix H2 and β-hairpin S2-S2' towards the mutation site, leading to the destruction of their neighboring β-sheet structure. This further facilitates the formation of a cavity in the hydrophobic core, and reduces the stability of the β-sandwich. Importantly, two crucial aggregation-prone regions (APRs) S9 and S10 are disturbed and more exposed to solvent in R248W p53C, which is conducive to p53C aggregation. Intriguingly, ADH-6 dynamically binds to the mutation site and multiple destabilized regions in R248W p53C, partially inhibiting the shift of helix H2 and β-hairpin S2-S2', thus preventing the disruption of the β-sheets and the formation of the cavity. ADH-6 also reduces the solvent exposure of APRs S9 and S10, which disfavors the aggregation of R248W p53C. Moreover, ADH-6 can preserve the WT-like dynamical network of R248W p53C. Our study elucidates the mechanisms underlying the oncogenic R248W mutation induced p53C destabilization and the structural protection of p53C by ADH-6.
Collapse
Affiliation(s)
- Qian Liu
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People's Republic of China.
| | - Yawei Yu
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People's Republic of China.
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People's Republic of China.
| |
Collapse
|
5
|
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
|
6
|
Das A, Sah P, Saraogi I. Dual Role of a Fluorescent Small Molecule as a Sensor and Inhibitor of Protein Fibrillation. Chem Asian J 2023; 18:e202201309. [PMID: 36594929 DOI: 10.1002/asia.202201309] [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: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/04/2023]
Abstract
Ordered fibrillar aggregates of proteins, called amyloids, are prevalent in several diseases like Alzheimer's, Parkinson's, and Type II diabetes. The key challenge in the treatment of such diseases is the early detection of protein fibrillation and its effective inhibition using extrinsic agents. Thus, molecules that can both detect and inhibit protein fibril formation have great diagnostic and therapeutic utility. Using insulin as a model protein, we report the dual action of an isoquinoline based molecule, named MK14 which detects and prevents insulin fibrillation. Dose dependent inhibition of insulin fibrillation by MK14 gave an IC50 value of 9 μM, and mechanistic investigations suggested that MK14 prevented the elongation of fibrils by interacting with pre-fibrillar intermediates. The fluorescence of MK14 enhanced upon binding to fibrils of insulin as well as those of α-synuclein, the protein involved in Parkinson's disease. MK14 is an environmentally sensitive fluorophore, which could also detect amorphous aggregates of insulin. The dual nature of MK14 as an inhibitor and detector of protein fibrillation makes it an attractive lead compound for monitoring and disrupting protein amyloidogenesis.
Collapse
Affiliation(s)
- Anirban Das
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| | - Pooja Sah
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India.,Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| |
Collapse
|
7
|
Maity D. Inhibition of Amyloid Protein Aggregation Using Selected Peptidomimetics. ChemMedChem 2023; 18:e202200499. [PMID: 36317359 DOI: 10.1002/cmdc.202200499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Indexed: 11/24/2022]
Abstract
Aberrant protein aggregation leads to the formation of amyloid fibrils. This phenomenon is linked to the development of more than 40 irremediable diseases such as Alzheimer's disease, Parkinson's disease, type 2 diabetes, and cancer. Plenty of research efforts have been given to understanding the underlying mechanism of protein aggregation, associated toxicity, and the development of amyloid inhibitors. Recently, the peptidomimetic approach has emerged as a potential tool to modulate several protein-protein interactions (PPIs). In this review, we discussed selected peptidomimetic-based approaches for the modulation of important amyloid proteins (Islet Amyloid Polypeptide, Amyloid Beta, α-synuclein, mutant p53, and insulin) aggregation. This approach holds a powerful platform for creating an essential stepping stone for the vital development of anti-amyloid therapeutic agents.
Collapse
Affiliation(s)
- Debabrata Maity
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
8
|
Gelb M, Messina KMM, Vinciguerra D, Ko JH, Collins J, Tamboline M, Xu S, Ibarrondo FJ, Maynard HD. Poly(trehalose methacrylate) as an Excipient for Insulin Stabilization: Mechanism and Safety. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37410-37423. [PMID: 35968684 PMCID: PMC9412841 DOI: 10.1021/acsami.2c09301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/21/2022] [Indexed: 05/07/2023]
Abstract
Insulin, the oldest U.S. Food and Drug Administration (FDA)-approved recombinant protein and a World Health Organization (WHO) essential medicine for treating diabetes globally, faces challenges due to its storage instability. One approach to stabilize insulin is the addition of poly(trehalose methacrylate) (pTrMA) as an excipient. The polymer increases the stability of the peptide to heat and mechanical agitation and has a low viscosity suitable for injection and pumps. However, the safety and stabilizing mechanism of pTrMA is not yet known and is required to understand the potential suitability of pTrMA as an insulin excipient. Herein is reported the immune response, biodistribution, and insulin plasma lifetime in mice, as well as investigation into insulin stabilization. pTrMA alone or formulated with ovalbumin did not elicit an antibody response over 3 weeks in mice, and there was no observable cytokine production in response to pTrMA. Micropositron emission tomography/microcomputer tomography of 64Cu-labeled pTrMA showed excretion of 78-79% ID/cc within 24 h and minimal liver accumulation at 6-8% ID/cc when studied out to 120 h. Further, the plasma lifetime of insulin in mice was not altered by added pTrMA. Formulating insulin with 2 mol equiv of pTrMA improved the stability of insulin to standard storage conditions: 46 weeks at 4 °C yielded 87.0% intact insulin with pTrMA present as compared to 7.8% intact insulin without the polymer. The mechanism by which pTrMA-stabilized insulin was revealed to be a combination of inhibiting deamidation of amino acid residues and preventing fibrillation, followed by aggregation of inactive and immunogenic amyloids all without complexing insulin into its hexameric state, which could delay the onset of insulin activity. Based on the data reported here, we suggest that pTrMA stabilizes insulin as an excipient without adverse effects in vivo and is promising to investigate further for the safe formulation of insulin.
Collapse
Affiliation(s)
- Madeline
B. Gelb
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Kathryn M. M. Messina
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Daniele Vinciguerra
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeong Hoon Ko
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeffrey Collins
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - Mikayla Tamboline
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - Shili Xu
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine,
University of California, Los Angeles, California 90095-1735, United States
| | - F. Javier Ibarrondo
- Division
of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1569, United States
| | - Heather D. Maynard
- Department
of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| |
Collapse
|
9
|
Das A, Gangarde YM, Pariary R, Bhunia A, Saraogi I. An amphiphilic small molecule drives insulin aggregation inhibition and amyloid disintegration. Int J Biol Macromol 2022; 218:981-991. [PMID: 35907468 DOI: 10.1016/j.ijbiomac.2022.07.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
The aggregation of proteins into ordered fibrillar structures called amyloids, and their disintegration represent major unsolved problems that limit the therapeutic applications of several proteins. For example, insulin, commonly used for the treatment of diabetes, is susceptible to amyloid formation upon exposure to non-physiological conditions, resulting in a loss of its biological activity. Here, we report a novel amphiphilic molecule called PAD-S, which acts as a chemical chaperone and completely inhibits fibrillation of insulin and its biosimilars. Mechanistic investigations and molecular docking lead to the conclusion that PAD-S binds to key hydrophobic regions of native insulin, thereby preventing its self-assembly. PAD-S treated insulin was biologically active as indicated by its ability to phosphorylate Akt, a protein in the insulin signalling pathway. PAD-S is non-toxic and protects cells from insulin amyloid induced cytotoxicity. The high aqueous solubility and easy synthetic accessibility of PAD-S facilitates its potential use in commercial insulin formulations. Notably, PAD-S successfully disintegrated preformed insulin fibrils to non-toxic smaller fragments. Since the structural and mechanistic features of amyloids are common to several human pathologies, the understanding of the amyloid disaggregation activity of PAD-S will inform the development of small molecule disaggregators for other amyloids.
Collapse
Affiliation(s)
- Anirban Das
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, MP, India
| | - Yogesh M Gangarde
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, MP, India
| | - Ranit Pariary
- Department of Biophysics, Bose Institute, Sector V, EN 80, Bidhan Nagar, Kolkata 700 091, India
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, Sector V, EN 80, Bidhan Nagar, Kolkata 700 091, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, MP, India; Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, MP, India.
| |
Collapse
|
10
|
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: 37] [Impact Index Per Article: 12.3] [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
|
11
|
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: 0.7] [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
|
12
|
Bucur P, Fülöp I, Sipos E. Insulin Complexation with Cyclodextrins-A Molecular Modeling Approach. Molecules 2022; 27:465. [PMID: 35056780 PMCID: PMC8778189 DOI: 10.3390/molecules27020465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 11/21/2022] Open
Abstract
Around 5% of the population of the world is affected with the disease called diabetes mellitus. The main medication of the diabetes is the insulin; the active form is the insulin monomer, which is an instable molecule, because the long storage time, or the high temperature, can cause the monomer insulin to adapt an alternative fold, rich in β-sheets, which is pharmaceutically inactive. The aim of this study is to form different insulin complexes with all the cyclodextrin used for pharmaceutical excipients (native cyclodextrin, methyl, hydroxyethyl, hydroxypropyl and sulfobutylether substituted β-cyclodextrin), in silico condition, with the AutoDock molecular modeling program, to determine the best type of cyclodextrin or cyclodextrin derivate to form a complex with an insulin monomer, to predict the molar ratio, the conformation of the complex, and the intermolecular hydrogen bonds formed between the cyclodextrin and the insulin. From the results calculated by the AutoDock program it can be predicted that insulin can make a stable complex with 5-7 molecules of hydroxypropyl-β-cyclodextrin or sulfobutylether-β-cyclodextrin, and by forming a complex potentially can prevent or delay the amyloid fibrillation of the insulin and increase the stability of the molecule.
Collapse
Affiliation(s)
- Pálma Bucur
- Department of Drugs Industry and Pharmaceutical Management, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Targu Mures, Romania; (P.B.); (E.S.)
| | - Ibolya Fülöp
- Department of Toxicology and Biopharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Targu Mures, Romania
| | - Emese Sipos
- Department of Drugs Industry and Pharmaceutical Management, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Targu Mures, Romania; (P.B.); (E.S.)
| |
Collapse
|
13
|
Morgan GJ. Transient disorder along pathways to amyloid. Biophys Chem 2021; 281:106711. [PMID: 34839162 DOI: 10.1016/j.bpc.2021.106711] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/15/2023]
Abstract
High-resolution structures of amyloid fibrils formed from normally-folded proteins have revealed non-native conformations of the polypeptide chains. Attaining these conformations apparently requires transition from the native state via a highly disordered conformation, in contrast to earlier models that posited a role for assembly of partially folded proteins. Modifications or interactions that extend the lifetime or constrain the conformations of these disordered states could act to enhance or suppress amyloid formation. Understanding how the properties of both the folded and transiently disordered structural ensembles influence the process of amyloid formation is a substantial challenge, but research into the properties of intrinsically disordered proteins will deliver important insights.
Collapse
Affiliation(s)
- Gareth J Morgan
- The Amyloidosis Center and Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
14
|
Pereira LMB, Cali MP, Marchi RC, Pazin WM, Carlos RM. Luminescent imaging of insulin amyloid aggregation using a sensitive ruthenium-based probe in the red region. J Inorg Biochem 2021; 224:111585. [PMID: 34450412 DOI: 10.1016/j.jinorgbio.2021.111585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 10/20/2022]
Abstract
A sensitive and selective strategy to identify insulin fibrils remains a challenge for researchers in amyloid protein research. Thus, it is critical to detect, in vitro, the species generated during amyloid aggregation, particularly the fibrillar species. Here we demonstrate that the luminescent complex cis-[Ru(phen)2(3,4Apy)2]2+ (RuApy; phen = 1,10-phenanthroline; 3,4Apy = 3,4-diaminopyridine) is a rapid, low-cost alternative to in vitro detection of fibrillar insulin, using conventional optical techniques. The RuApy complex displays emission intensity enhancement at 655 nm when associated with insulin, which enables imaging of the conformational changes of the protein's self-aggregation. The complex shows high sensitivity to fibrillar insulin with a limit of detection of 0.85 μM and binding affinity of 12.40 ± 1.84 μM which is comparable to those of Thioflavin T and Congo red, with the advantage of minimizing background fluorescence, absorption of light by biomolecules, and light scattering from physiologic salts in the medium.
Collapse
Affiliation(s)
- Lorena M B Pereira
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Mariana P Cali
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Rafael C Marchi
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Wallance M Pazin
- Department of Physics, Universidade Estadual Paulista, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Rose M Carlos
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil.
| |
Collapse
|
15
|
Morgan GJ. Barriers to Small Molecule Drug Discovery for Systemic Amyloidosis. Molecules 2021; 26:3571. [PMID: 34208058 PMCID: PMC8230685 DOI: 10.3390/molecules26123571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Inhibition of amyloid fibril formation could benefit patients with systemic amyloidosis. In this group of diseases, deposition of amyloid fibrils derived from normally soluble proteins leads to progressive tissue damage and organ failure. Amyloid formation is a complex process, where several individual steps could be targeted. Several small molecules have been proposed as inhibitors of amyloid formation. However, the exact mechanism of action for a molecule is often not known, which impedes medicinal chemistry efforts to develop more potent molecules. Furthermore, commonly used assays are prone to artifacts that must be controlled for. Here, potential mechanisms by which small molecules could inhibit aggregation of immunoglobulin light-chain dimers, the precursor proteins for amyloid light-chain (AL) amyloidosis, are studied in assays that recapitulate different aspects of amyloidogenesis in vitro. One molecule reduced unfolding-coupled proteolysis of light chains, but no molecules inhibited aggregation of light chains or disrupted pre-formed amyloid fibrils. This work demonstrates the challenges associated with drug development for amyloidosis, but also highlights the potential to combine therapies that target different aspects of amyloidosis.
Collapse
Affiliation(s)
- Gareth J Morgan
- Section of Hematology and Medical Oncology, Amyloidosis Center, Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
16
|
Gangarde YM, Das A, Ajit J, Saraogi I. Synthesis and Evaluation of Arylamides with Hydrophobic Side Chains for Insulin Aggregation Inhibition. Chempluschem 2021; 86:750-757. [PMID: 33949802 DOI: 10.1002/cplu.202100036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/17/2021] [Indexed: 11/10/2022]
Abstract
Insulin, a peptide hormone, forms fibrils under aberrant physiological conditions leading to a reduction in its biological activity. To ameliorate insulin aggregation, we have synthesized a small library of oligopyridylamide foldamers decorated with different combination of hydrophobic side chains. Screening of these compounds for insulin aggregation inhibition using a Thioflavin-T assay resulted in the identification of a few hit molecules. The best hit molecule, BPAD2 inhibited insulin aggregation with an IC50 value of 0.9 μM. Mechanistic analyses suggested that BPAD2 inhibited secondary nucleation and elongation processes during aggregation. The hit molecules worked in a mechanistically distinct manner, thereby underlining the importance of structure-activity relationship studies in obtaining a molecular understanding of protein aggregation.
Collapse
Affiliation(s)
- Yogesh M Gangarde
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| | - Anirban Das
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| | - Jainu Ajit
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India.,Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, 462066, MP, India
| |
Collapse
|
17
|
Alam S, Nasreen S, Ahmad A, Darokar MP, Khan F. Detection of Natural Inhibitors against Human Liver Cancer Cell Lines through QSAR, Molecular Docking and ADMET Studies. Curr Top Med Chem 2021; 21:686-695. [PMID: 33280598 DOI: 10.2174/1568026620666201204155830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/22/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Liver cancer is ranked as the fifth most prevalent and third most lethal cancer worldwide. The incidence rates of this cancer are on the rise, and only limited treatment options are available. METHODS To identify and optimize the inhibitors of liver cancer cell-lines, a QSAR model was developed by using multiple linear regression methods. The robustness of the model was validated through statistical methods and wet-lab experiments. RESULTS The developed QSAR models yielded high activity descriptor relationship accuracy of 91%, referred to by regression coefficient (r2= 0.91), and a high activity prediction accuracy of 89%. The external predicted (pred_r2) ability of the model was found to be 90%. CONCLUSION The QSAR study indicates that chemical descriptors such as to measure of electronegative atom count (Epsilon3), atom type count descriptors (MMFF_10), number of a carbon atom connected with four single bonds (SssssCE- index), molecular weight and, number of oxygen atom connected with two aromatic bonds (SaaOE-index) are significantly correlated with anticancer activity. The model, which was validated statistically and through wet-lab experiments, was further used in the virtual screening of potential inhibitors against the liver cancer cell line WRL68. ADMET risk screening, synthetic accessibility, and Lipinski's rule of five are used to filter false positive hits. AfterwardS, to achieve a set of aligned ligand poses and rank the predicted active compounds, docking studies were carried out. The studied compounds and their metabolites were also analyzed for different pharmacokinetics parameters. Finally, a series of compounds was proposed as anticancer agents.
Collapse
Affiliation(s)
- Sarfaraz Alam
- Metabolic & Structural Biology Department, CSIR-Central Institute of Medicinal & Aromatic Plants, Lucknow 226015 (Uttar Pradesh), India
| | - Sadaf Nasreen
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal & Aromatic Plants, Lucknow 226015 (Uttar Pradesh), India
| | - Ateeque Ahmad
- Process Chemistry and Technology Department, CSIR-Central Institute of Medicinal & Aromatic Plants, Lucknow 226015 (Uttar Pradesh), India
| | - Mahendra Pandurang Darokar
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal & Aromatic Plants, Lucknow 226015 (Uttar Pradesh), India
| | - Feroz Khan
- Metabolic & Structural Biology Department, CSIR-Central Institute of Medicinal & Aromatic Plants, Lucknow 226015 (Uttar Pradesh), India
| |
Collapse
|
18
|
Iqbal H, Verma AK, Yadav P, Alam S, Shafiq M, Mishra D, Khan F, Hanif K, Negi AS, Chanda D. Antihypertensive Effect of a Novel Angiotensin II Receptor Blocker Fluorophenyl Benzimidazole: Contribution of cGMP, Voltage-dependent Calcium Channels, and BK Ca Channels to Vasorelaxant Mechanisms. Front Pharmacol 2021; 12:611109. [PMID: 33859561 PMCID: PMC8042648 DOI: 10.3389/fphar.2021.611109] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/09/2021] [Indexed: 12/03/2022] Open
Abstract
Background: The current study presents the novel angiotensin II receptor blocker fluorophenyl benzimidazole (FPD) as an antihypertensive agent in the SHR model of hypertension. We investigated the role of cGMP, voltage-dependent L-type calcium channels, and BKCa channels in the vasorelaxant mechanisms of FPD in the rat superior mesenteric artery. Methods: The antihypertensive effect of FPD was examined using an invasive technique measuring blood pressure in SHR animals. Using a myograph, tension measurement was completed in the superior mesenteric artery to elucidate the mechanisms of vasorelaxation involving AT1 receptors, the NO/cGMP pathway, L-type calcium channels, and BKCa channels. Ion flux (Ca2+, K+) studies were conducted in aortic smooth muscle cells. Putative targets proteins were determined by in silico docking studies. A safety evaluation of FPD was carried out using Swiss albino mice. Results: FPD significantly decreased blood pressure in SHR. It relaxed superior mesenteric arteries in a concentration-dependent manner and significantly inhibited angiotensin II-induced contraction. The relaxation response was also mediated by an increase in tissue cGMP levels, inhibition of L-type calcium channels, and the opening of BKCa channels. FPD further enhanced efflux of K+ and inhibited Bay K8644-stimulated Ca2+ influx in aortic smooth muscle cells and docked well in an in silico study with the targets. It was well tolerated in the toxicity study. Conclusion: The present study reports the antihypertensive activity of novel AT-1 receptor blocker FPD at 50 and 100 mg kg−1 with cGMP, L-type calcium channels, and BKCa channels as putative targets of vasorelaxation, and was found safe in oral toxicity.
Collapse
Affiliation(s)
- Hina Iqbal
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Amit Kumar Verma
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Pankaj Yadav
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Sarfaraz Alam
- Computational Biology Lab, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Mohammad Shafiq
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Divya Mishra
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Feroz Khan
- Computational Biology Lab, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Arvind Singh Negi
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Debabrata Chanda
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| |
Collapse
|