1
|
Nguyen V, Côté-Cyr M, Finatto AN, Babych M, Nguyen PT, Sebastiao M, Bourgault S. Probing the relationships between self-assembly and the antimicrobial activity of amyloidogenic peptides: The islet amyloid polypeptide as a case study. Biochim Biophys Acta Gen Subj 2025; 1869:130812. [PMID: 40268062 DOI: 10.1016/j.bbagen.2025.130812] [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: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
Antimicrobial peptides (AMPs) are key components of the innate immune system across diverse organisms. Interestingly, some AMPs can adopt β-sheet secondary structure and self-assemble into amyloid-like fibrils. Recent works have also revealed that amyloidogenic peptides exhibit antimicrobial properties and share a common mechanism of plasma membrane perturbation with AMPs. In this study, we explored the relationships between the antimicrobial activity of amyloidogenic peptides and their self-assembly by using the islet amyloid polypeptide (IAPP) as a model. IAPP is an aggregation-prone 37-residue hormone whose pancreatic deposition and accumulation are associated with type II diabetes. Antimicrobial assays revealed that IAPP monomers and prefibrillar aggregates, including soluble oligomers, inhibit the growth of Escherichia coli and Staphylococcus epidermidis. Additionally, monomeric and prefibrillar proteospecies perturbed anionic lipid vesicles that mimic bacterial plasma membrane and decrease the metabolic activity. In contrast, pre-assembled amyloid fibrils exhibited weak antimicrobial activities and lipid membrane perturbation, although they agglutinated bacteria avidly. By taking advantage of residue-specific substitutions that modulate the aggregation propensity, we observed that derivatives with hindered amyloidogenicity retained antimicrobial activities, while those with accelerated kinetics of amyloid self-assembly had weaker antimicrobial effect. Moreover, by modulating the propensity of IAPP to fold into an α-helix, we observed that amyloid formation is not a prerequisite for the antimicrobial activity, while the destabilization of helical folding reduced IAPP antimicrobial activity. This study provides fundamental mechanistic insights of the modest antimicrobial activity of IAPP and highlights that precaution should be taken before generalizing the antimicrobial potential of self-assembling amyloid polypeptides.
Collapse
Affiliation(s)
- Vy Nguyen
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada
| | - Arthur Nery Finatto
- Department of Large Animal Clinical Sciences, University of Saskatchewan, Saskatoon S7N 5B4, Canada
| | - Margaryta Babych
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada
| | - Phuong Trang Nguyen
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada
| | - Mathew Sebastiao
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada; Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Canada.
| |
Collapse
|
2
|
Tang L, Sun M, Chen J, Dai Q, Xue S, Liu C, Zhang M. Peptide-functionalized nanocapsules for targeted inhibition of β2-microglobulin amyloid aggregation. J Mater Chem B 2025; 13:3319-3324. [PMID: 39928035 DOI: 10.1039/d4tb01347f] [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: 02/11/2025]
Abstract
Dialysis-related amyloidosis (DRA) is a severe complication in patients undergoing long-term dialysis, primarily driven by the deposition of β2-microglobulin (β2m) amyloid fibrils. The effective sequestration and removal of β2m from the bloodstream represent key therapeutic strategies for managing DRA. In this study, we developed a β2m-binding peptide (KDWSFYILAHTEF, denoted as CF)-functionalized nanocomposite (NC-CF), consisting of a protein nanocapsule surface modified with CF peptides to enable specific β2m binding. NC-CF effectively modulates β2m aggregation, transforming slender fibrils into larger clumps while providing steric hindrance to prevent further aggregation. With a high adsorption capacity, 1 μg of NC-CF can adsorb approximately 1 μg of β2m during dialysis, highlighting its potential as an efficient adsorbent for in vitro β2m removal. Furthermore, NC-CF exhibits excellent biocompatibility and significantly mitigates β2m aggregate-induced cytotoxicity, achieving a cell protection rate exceeding 70%. These findings suggest that NC-CF holds great promise as a cytoprotective agent and a nanoinhibitor of β2m aggregation in vivo. Overall, NC-CF offers a novel and effective approach for alleviating DRA by simultaneously removing β2m and safeguarding cells against amyloid-induced toxicity.
Collapse
Affiliation(s)
- Lin Tang
- Department of Medical Imaging, Qilu Medical University, 255100, P. R. China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Miao Sun
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Junnan Chen
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Song Xue
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing 102206, P. R. China.
| |
Collapse
|
3
|
Yang L, Chen J, Yao Z, Cai J, Zhang H, Wang Z, Guo H, Zha Y. Associations of multiple plasma metals with the risk of type 2 diabetes in Chinese adults: A cross-sectional study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117941. [PMID: 40009940 DOI: 10.1016/j.ecoenv.2025.117941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Evidence regarding the associations between co-exposure to multiple metals and diabetes risk was scarce. This study aimed to evaluate the associations of multiple metals with diabetes risk using multiple statistical methods. This cross-sectional study included 192 diabetic patients and 189 healthy subjects. We employed inductively coupled plasma mass spectrometry (ICP-MS) to determine the plasma concentrations of 18 metals. Least absolute shrinkage and selection operator (LASSO) regression, logistic regression, and Bayesian kernel machine regression (BKMR) were applied to evaluate associations of multiple metals with diabetes risk comprehensively. These models consistently suggested that aluminium and selenium were positively associated with diabetes risk, while manganese, rubidium, and lead were negatively associated with diabetes risk. Age-specific differences of selenium and sex-specific differences of manganese in diabetes risk were also observed based on stratified analyses. According to RCS analyses, we obtained dose-response relationships between metals and diabetes risk:(1) there were inverted U-shaped associations of plasma aluminium and selenium with diabetes risk, with the threshold close to 20.5µg/L and 75.9µg/L, respectively (both P for overall < 0.05; both P for non-linearity < 0.05). (2) There were L-shaped associations of rubidium and lead with diabetes risk, with the turning point close to 144.5µg/L and 2.5µg/L, respectively (both P for overall < 0.05; both P for non-linearity < 0.05). (3) Manganese was linearly and negatively correlated with diabetes risk when concentrations of manganese were less than approximately 4.2 μg/L (P for overall < 0.05; P for non-linearity = 0.268). The BKMR model also revealed a negative combined effect of metal mixtures on diabetes risk and potential interactions between six pairs of metals (aluminium-manganese, aluminium-selenium, aluminium-rubidium, aluminium-lead, manganese-selenium, and manganese-rubidium). In summary, we need to pay attention to the role of low plasma levels of aluminium, selenium, manganese, rubidium, and lead in diabetes, especially regarding their safety windows.
Collapse
Affiliation(s)
- Liting Yang
- Biomedical Research Institute of Hubei University of Medicine, Shiyan 442000, China
| | - Jin Chen
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Zijun Yao
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China; Public Health Service Center of Wuhan East Lake Scenic District, Wuhan 430077, China
| | - Junwei Cai
- Department of Endocrinology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Han Zhang
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Zhen Wang
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Huailan Guo
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China.
| | - Yongjiu Zha
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
4
|
Wang Z, Wei J, Zhang X, Ji H, Fu S, Gao Z, Li H. Nitration of Tyr37 alters the aggregation pathway of hIAPP and enhances its cytotoxicity. Int J Biol Macromol 2025; 286:138367. [PMID: 39643176 DOI: 10.1016/j.ijbiomac.2024.138367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
The amyloid aggregation of hIAPP and the increased level of oxidative stress are closely related to the occurrence and development of type 2 diabetes (T2D). Protein tyrosine nitration is a common post-translational modification under oxidative stress conditions. We previously found that tyrosine nitrated hIAPP (3-NT-hIAPP) has higher cytotoxicity than wild type hIAPP. In order to further elucidate the mechanism by which tyrosine nitration enhances the toxicity of hIAPP, we systematically studied the effect of tyrosine nitration on hIAPP aggregation and its impact on INS-1 cells. Collective experimental data from ThT, RLS, DLS, zeta potentials, Bis-ANS, 1H NMR, TEM, dye leakage and hemolysis confirmed that tyrosine nitration accelerates hIAPP aggregation, consistent with tyrosine nitration reducing hIAPP zeta potential, but 3-NT-hIAPP mainly undergoes an off-pathway aggregation to form amorphous aggregates, even in the presence of POPC/POPG LUVs. Further, our results confirmed that the most toxic species are the small amorphous aggregates formed by 3-NT-hIAPP, which is more stable and toxic than hIAPP oligomers. Collectively, these data suggest that tyrosine nitration can increase cytotoxicity of hIAPP by modulating its amyloidogenicity. This study provides new support for the fact that oxidative stress promotes the development of T2D from the view of nitrative stress.
Collapse
Affiliation(s)
- Zhilong Wang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Jingjing Wei
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Xuan Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Haoran Ji
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Shitao Fu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China.
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China.
| |
Collapse
|
5
|
Abioye RO, Adetula OH, Hum JD, Udenigwe CC. Influence of anti-fibrillation TNGQ peptide and rutin combination on β-cell cytoprotective effects against IAPP-induced cell death and oxidative stress. Biochem Biophys Res Commun 2024; 739:150976. [PMID: 39556938 DOI: 10.1016/j.bbrc.2024.150976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/21/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024]
Abstract
Type 2 diabetes development has been associated with islet amyloid polypeptide (IAPP) fibrillation. IAPP fibrils have various deleterious effects, such as oxidative stress and disruption of cellular membrane integrity, resulting in pancreatic β-cell toxicity. Rutin, a plant polyphenol, possesses promising cytoprotective effects as a fibrillation inhibitor. Similarly, bioactive peptides have been identified as potential inhibitors to IAPP fibrillation. In this study, the effect of peptide/polyphenol mixtures consisting of rutin and each peptide, TNGQ, MANT, and YMSV, on anti-fibrillation activity and cellular response was elucidated. Results indicated a 54.7-75.1 % decrease in thioflavin T fluorescence, confirming anti-fibrillation activity. The combination decreased the average particle diameters of IAPP more than the single inhibitors, suggesting a combined effect of peptide/rutin mixtures in enhancing anti-fibrillation activity. IAPP fibrillation-induced rat insulinoma RIN-m cell death was minimized in the presence of the peptide/rutin mixture, but the activity was lower relative to rutin alone, suggesting a non-additive effect of the mixtures. Transmission electron microscopy showed a near-complete inhibition of IAPP fibrillation by TNGQ/rutin mixtures, which translated to a decreased production of membrane-bound IAPP oligomers in RIN-m cells based on immunofluorescence staining. Additionally, TNGQ/rutin mixtures significantly decreased reactive oxygen species production by 30 %, higher than the effects of single inhibitors, but no effect was observed on glucose-stimulated insulin secretion. The results demonstrate the potential of multifunctional compounds as dual inhibitor systems in controlling IAPP fibrillation and provide insight into the implications of peptide/polyphenol mixtures towards the rational development of novel anti-diabetic nutraceutical combinations.
Collapse
Affiliation(s)
- Raliat O Abioye
- School of Nutrition Sciences, Health Sciences, University of Ottawa, Ottawa, K1H 8M5, Canada; Department of Chemistry and Biomolecular Sciences, Science, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada
| | - Oluwasemilogo H Adetula
- School of Nutrition Sciences, Health Sciences, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Julia Diem Hum
- School of Nutrition Sciences, Health Sciences, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Chibuike C Udenigwe
- School of Nutrition Sciences, Health Sciences, University of Ottawa, Ottawa, K1H 8M5, Canada; Department of Chemistry and Biomolecular Sciences, Science, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada; University Food Properties and Nutrient Bioavailability, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.
| |
Collapse
|
6
|
Tyczyńska M, Hunek G, Kawecka W, Brachet A, Gędek M, Kulczycka K, Czarnek K, Flieger J, Baj J. Association Between Serum Concentrations of (Certain) Metals and Type 2 Diabetes Mellitus. J Clin Med 2024; 13:7443. [PMID: 39685901 DOI: 10.3390/jcm13237443] [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: 10/28/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
The findings regarding trace element concentrations in patients diagnosed with type 2 diabetes and healthy controls are inconsistent, and therefore, we determined to gather them in the form of a review to further indicate the need for more advanced knowledge development. In our study, we reviewed articles and studies that involved the topics of micronutrient and metal associations with the occurrence and development of type 2 diabetes. We mainly included works regarding human-based studies, but with limited research results, animal-based research was also taken into account. With some newer studies, we reached for initial assumptions of previous statements. The results indicated that higher serum levels of lead, cadmium, arsenic, bromine, barium, strontium, nickel, aluminum, calcium, copper, and ferritin are positively associated with diabetic prevalence. Both too-low and too-high levels of zinc, selenium, and magnesium may be connected to the development of diabetes. Chromium has the capability of insulin response modulation, with enhanced insulin-cell binding, and thus, lower serum levels of chromium can be found in diabetic patients. There are contradictory discoveries regarding manganese. Its supplementation can possibly cease the development of insulin resistance and type 2 diabetes. On the contrary, other studies reported that there is no such connection. Our work indicates that, as micronutrients play a significant role in the pathogenesis of metabolic disorders, more research regarding their bodily homeostasis and type 2 diabetes should be conducted.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Gabriela Hunek
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Weronika Kawecka
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Adam Brachet
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Marta Gędek
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Kinga Kulczycka
- Institute of Medical Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1, 20-708 Lublin, Poland
| | - Katarzyna Czarnek
- Institute of Medical Sciences, The John Paul II Catholic University of Lublin, Konstantynów 1, 20-708 Lublin, Poland
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| |
Collapse
|
7
|
Zhang J, Mesias VSD, Chesney AD, Anand VK, Feng X, Hsing IM, Hansmann UHE, Huang J. Differential effects of SARS-CoV-2 amyloidogenic segments on the aggregation and toxicity of human islet amyloid polypeptide within membrane environments. Int J Biol Macromol 2024; 283:137930. [PMID: 39579816 DOI: 10.1016/j.ijbiomac.2024.137930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Human islet amyloid polypeptide (hIAPP), an intrinsically disordered protein (IDP), plays a significant role in the pathogenesis of type 2 diabetes through its aggregation. Recent studies have suggested that certain viral protein segments exhibit amyloidogenic potential and may influence its amyloid aggregations associated with pathogenesis. However, the potential link between recurrent SARS-CoV-2 infections and the exacerbation of type 2 diabetes remains poorly understood. In this study, we explore how the amyloidogenic segments of SARS-CoV-2, specifically SK9 and FI10, influence the aggregation of hIAPP and the toxicity of the resulting conformers in a membrane environment. To investigate this, we utilized a range of biophysical techniques, including circular dichroism, nuclear magnetic resonance, atomic force microscopy, dynamic light scattering, fluorescence assays, and cell cytotoxicity assays, complemented by molecular dynamics simulations. Our results indicate that SK9 and FI10 promote hIAPP aggregation in a membrane-mimicking environment, forming distinct aggregate structures. Specifically, SK9 accelerates rapid fibril formation due to inter-chain interactions, while FI10 stabilizes oligomeric aggregates primarily through intra-chain contacts. These results reveal the differential effects of viral protein segments on amyloid formation pathways and aggregate characteristics, providing new insights into the mechanisms of amyloid aggregation for developing better therapeutic strategies against amyloid-associated diseases, particularly diabetes.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Vince St Dollente Mesias
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Andrew D Chesney
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States
| | - Vignesh K Anand
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States
| | - Xianzhen Feng
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - I-Ming Hsing
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ulrich H E Hansmann
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States.
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
8
|
Song J. In the Beginning: Let Hydration Be Coded in Proteins for Manifestation and Modulation by Salts and Adenosine Triphosphate. Int J Mol Sci 2024; 25:12817. [PMID: 39684527 DOI: 10.3390/ijms252312817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Water exists in the beginning and hydrates all matter. Life emerged in water, requiring three essential components in compartmentalized spaces: (1) universal energy sources driving biochemical reactions and processes, (2) molecules that store, encode, and transmit information, and (3) functional players carrying out biological activities and structural organization. Phosphorus has been selected to create adenosine triphosphate (ATP) as the universal energy currency, nucleic acids for genetic information storage and transmission, and phospholipids for cellular compartmentalization. Meanwhile, proteins composed of 20 α-amino acids have evolved into extremely diverse three-dimensional forms, including folded domains, intrinsically disordered regions (IDRs), and membrane-bound forms, to fulfill functional and structural roles. This review examines several unique findings: (1) insoluble proteins, including membrane proteins, can become solubilized in unsalted water, while folded cytosolic proteins can acquire membrane-inserting capacity; (2) Hofmeister salts affect protein stability by targeting hydration; (3) ATP biphasically modulates liquid-liquid phase separation (LLPS) of IDRs; (4) ATP antagonizes crowding-induced protein destabilization; and (5) ATP and triphosphates have the highest efficiency in inducing protein folding. These findings imply the following: (1) hydration might be encoded in protein sequences, central to manifestation and modulation of protein structures, dynamics, and functionalities; (2) phosphate anions have a unique capacity in enhancing μs-ms protein dynamics, likely through ionic state exchanges in the hydration shell, underpinning ATP, polyphosphate, and nucleic acids as molecular chaperones for protein folding; and (3) ATP, by linking triphosphate with adenosine, has acquired the capacity to spacetime-specifically release energy and modulate protein hydration, thus possessing myriad energy-dependent and -independent functions. In light of the success of AlphaFolds in accurately predicting protein structures by neural networks that store information as distributed patterns across nodes, a fundamental question arises: Could cellular networks also handle information similarly but with more intricate coding, diverse topological architectures, and spacetime-specific ATP energy supply in membrane-compartmentalized aqueous environments?
Collapse
Affiliation(s)
- Jianxing Song
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260, Singapore
| |
Collapse
|
9
|
Naik AR, Save SN, Sahoo SS, Yadav SS, Kumar A, Chugh J, Sharma S. Metabolic perturbations associated with hIAPP-induced insulin resistance in skeletal muscles: Implications to the development of type 2 diabetes. Int J Biochem Cell Biol 2024; 176:106665. [PMID: 39322038 DOI: 10.1016/j.biocel.2024.106665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
The human islet amyloid polypeptide (hIAPP) tends to misfold and self-assemble to form amyloid fibrils, which has been associated with the loss of function and viability of pancreatic β-cells in type 2 diabetes mellitus (T2DM). The role of hIAPP in the development of insulin resistance (a hallmark of T2DM) in skeletal muscles - the major sites for glucose utilization - needs further investigation. Even though, insulin-resistant conditions have been known to stimulate hIAPP aggregation, the events that lead to the development of insulin resistance due to hIAPP aggregation in skeletal muscles remain unidentified. Here, we have attempted to identify metabolic perturbations in L6 myotubes that were exposed to increasing concentrations of recombinant hIAPP for different time durations. It was observed that hIAPP exposure was associated with increased mitochondrial and cellular ROS levels, loss in mitochondrial membrane potential and viability of the myotubes. Metabolomic investigations of hIAPP-treated myotubes revealed significant perturbations in o-phosphocholine, sn-glycero-3-phosphocholine and dimethylamine levels (p < 0.05). Therefore, we anticipate that defects in glycerophospholipid metabolism and the associated oxidative stress and membrane damage may play key roles in the development of insulin resistance due to protein misfolding in skeletal muscles. In summary, the perturbed metabolites and their pathways have not only the potential to be used as early biomarkers to predict the onset of insulin resistance and T2DM but also as therapeutic targets for the effective management of the same.
Collapse
Affiliation(s)
- Arya R Naik
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Shreyada N Save
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Soumya S Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Saurabh S Yadav
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, Indian institute of technology Bombay, Powai, Mumbai, Maharashtra 400076, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
10
|
Babych M, Garelja ML, Nguyen PT, Hay DL, Bourgault S. Converting the Amyloidogenic Islet Amyloid Polypeptide into a Potent Nonaggregating Peptide Ligand by Side Chain-to-Side Chain Macrocyclization. J Am Chem Soc 2024; 146:25513-25526. [PMID: 39225636 DOI: 10.1021/jacs.4c05297] [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: 09/04/2024]
Abstract
The islet amyloid polypeptide (IAPP), also known as amylin, is a hormone playing key physiological roles. However, its aggregation and deposition in the pancreatic islets are associated with type 2 diabetes. While this peptide adopts mainly a random coil structure in solution, its secondary conformational conversion into α-helix represents a critical step for receptor activation and contributes to amyloid formation and associated cytotoxicity. Considering the large conformational landscape and high amyloidogenicity of the peptide, as well as the complexity of the self-assembly process, it is challenging to delineate the delicate interplay between helical folding, peptide aggregation, and receptor activation. In the present study, we probed the roles of helical folding on the function-toxicity duality of IAPP by restricting its conformational ensemble through side chain-to-side chain stapling via azide-alkyne cycloaddition. Intramolecular macrocyclization (i; i + 4) constrained IAPP into α-helix and inhibited its aggregation into amyloid fibrils. These helical derivatives slowed down the self-assembly of unmodified IAPP. Site-specific macrocyclization modulated the capacity of IAPP to perturb lipid bilayers and cell plasma membrane and reduced, or even fully inhibited, the cytotoxicity associated with aggregation. Furthermore, the α-helical IAPP analogs showed moderate to high potency toward cognate G protein-coupled receptors. Overall, these results indicate that macrocyclization represents a promising strategy to protect an amyloidogenic peptide hormone from aggregation and associated toxicity, while maintaining high receptor activity.
Collapse
Affiliation(s)
- Margaryta Babych
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
| | - Michael L Garelja
- Department of Pharmacology and Toxicology, University of Otago, 18 Frederick Street, Dunedin 9016, New Zealand
| | - Phuong Trang Nguyen
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, 18 Frederick Street, Dunedin 9016, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 3A Symonds Street, Auckland 92019, New Zealand
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, C.P. 8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
| |
Collapse
|
11
|
Nabi F, Ahmad O, Khan A, Hassan MN, Hisamuddin M, Malik S, Chaari A, Khan RH. Natural compound plumbagin based inhibition of hIAPP revealed by Markov state models based on MD data along with experimental validations. Proteins 2024; 92:1070-1084. [PMID: 38497314 DOI: 10.1002/prot.26682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
Human islet amyloid polypeptide (amylin or hIAPP) is a 37 residue hormone co-secreted with insulin from β cells of the pancreas. In patients suffering from type-2 diabetes, amylin self-assembles into amyloid fibrils, ultimately leading to the death of the pancreatic cells. However, a research gap exists in preventing and treating such amyloidosis. Plumbagin, a natural compound, has previously been demonstrated to have inhibitory potential against insulin amyloidosis. Our investigation unveils collapsible regions within hIAPP that, upon collapse, facilitates hydrophobic and pi-pi interactions, ultimately leading to aggregation. Intriguingly plumbagin exhibits the ability to bind these specific collapsible regions, thereby impeding the aforementioned interactions that would otherwise drive hIAPP aggregation. We have used atomistic molecular dynamics approach to determine secondary structural changes. MSM shows metastable states forming native like hIAPP structure in presence of PGN. Our in silico results concur with in vitro results. The ThT assay revealed a striking 50% decrease in fluorescence intensity at a 1:1 ratio of hIAPP to Plumbagin. This finding suggests a significant inhibition of amyloid fibril formation by plumbagin, as ThT fluorescence directly correlates with the presence of these fibrils. Further TEM images revealed disappearance of hIAPP fibrils in plumbagin pre-treated hIAPP samples. Also, we have shown that plumbagin disrupts the intermolecular hydrogen bonding in hIAPP fibrils leading to an increase in the average beta strand spacing, thereby causing disaggregation of pre-formed fibrils demonstrating overall disruption of the aggregation machinery of hIAPP. Our work is the first to report a detailed atomistic simulation of 22 μs for hIAPP. Overall, our studies put plumbagin as a potential candidate for both preventive and therapeutic candidate for hIAPP amyloidosis.
Collapse
Affiliation(s)
- Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Owais Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Adeeba Khan
- Zakir Hussain College of Engineering and Technology, Aligarh Muslim University, Aligarh, India
| | - Md Nadir Hassan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Malik Hisamuddin
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Sadia Malik
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Ali Chaari
- Premedical Division, Weill Cornell Medicine Qatar, Qatar Foundation, Doha, Qatar
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
12
|
Jordan J, Gibb CL, Tran T, Yao W, Rose A, Mague JT, Easson MW, Gibb BC. Anion Binding to Ammonium and Guanidinium Hosts: Implications for the Reverse Hofmeister Effects Induced by Lysine and Arginine Residues. J Org Chem 2024; 89:6877-6891. [PMID: 38662908 PMCID: PMC11110012 DOI: 10.1021/acs.joc.4c00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024]
Abstract
Anions have a profound effect on the properties of soluble proteins. Such Hofmeister effects have implications in biologics stability, protein aggregation, amyloidogenesis, and crystallization. However, the interplay between the important noncovalent interactions (NCIs) responsible for Hofmeister effects is poorly understood. To contribute to improving this state of affairs, we report on the NCIs between anions and ammonium and guanidinium hosts 1 and 2, and the consequences of these. Specifically, we investigate the properties of cavitands designed to mimic two prime residues for anion-protein NCIs─lysines and arginines─and the solubility consequences of complex formation. Thus, we report NMR and ITC affinity studies, X-ray analysis, MD simulations, and anion-induced critical precipitation concentrations. Our findings emphasize the multitude of NCIs that guanidiniums can form and how this repertoire qualitatively surpasses that of ammoniums. Additionally, our studies demonstrate the ease by which anions can dispense with a fraction of their hydration-shell waters, rearrange those that remain, and form direct NCIs with the hosts. This raises many questions concerning how solvent shell plasticity varies as a function of anion, how the energetics of this impact the different NCIs between anions and ammoniums/guanidiniums, and how this affects the aggregation of solutes at high anion concentrations.
Collapse
Affiliation(s)
- Jacobs
H. Jordan
- The
Southern Regional Research Center, Agricultural Research Service, US Department of Agriculture, 1100 Allen Toussaint Blvd., New Orleans, Louisiana 70124, United States
| | - Corinne L.D. Gibb
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Thien Tran
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Wei Yao
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Austin Rose
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Joel T. Mague
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Michael W. Easson
- The
Southern Regional Research Center, Agricultural Research Service, US Department of Agriculture, 1100 Allen Toussaint Blvd., New Orleans, Louisiana 70124, United States
| | - Bruce C. Gibb
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| |
Collapse
|
13
|
Laxio Arenas J, Lesma J, Ha-Duong T, Ranjan Sahoo B, Ramamoorthy A, Tonali N, Soulier JL, Halgand F, Giraud F, Crousse B, Kaffy J, Ongeri S. Composition and Conformation of Hetero- versus Homo-Fluorinated Triazolamers Influence their Activity on Islet Amyloid Polypeptide Aggregation. Chemistry 2024; 30:e202303887. [PMID: 38478740 DOI: 10.1002/chem.202303887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Indexed: 04/11/2024]
Abstract
Novel fluorinated foldamers based on aminomethyl-1,4-triazolyl-difluoroacetic acid (1,4-Tz-CF2) units were synthesized and their conformational behaviour was studied by NMR and molecular dynamics. Their activity on the aggregation of the human islet amyloid polypeptide (hIAPP) amyloid protein was evaluated by fluorescence spectroscopy and mass spectrometry. The fluorine labelling of these foldamers allowed the analysis of their interaction with the target protein. We demonstrated that the preferred extended conformation of homotriazolamers of 1,4-Tz-CF2 unit increases the aggregation of hIAPP, while the hairpin-like conformation of more flexible heterotriazolamers containing two 1,4-Tz-CF2 units mixed with natural amino acids from the hIAPP sequence reduces it, and more efficiently than the parent natural peptide. The longer heterotriazolamers having three 1,4-Tz-CF2 units adopting more folded hairpin-like and ladder-like structures similar to short multi-stranded β-sheets have no effect. This work demonstrates that a good balance between the structuring and flexibility of these foldamers is necessary to allow efficient interaction with the target protein.
Collapse
Affiliation(s)
- José Laxio Arenas
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Jacopo Lesma
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Tap Ha-Duong
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Bikash Ranjan Sahoo
- Biophysics, Department of Chemistry, Biomedical Engineering, Michigan Neuroscience Institute, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Ayyalusamy Ramamoorthy
- Biophysics, Department of Chemistry, Biomedical Engineering, Michigan Neuroscience Institute, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Nicolo Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Jean-Louis Soulier
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Frédéric Halgand
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405, Orsay, France
| | - François Giraud
- Equipe Biologie et Chimie Structurales, Dept Chimie et Biologie Structurales et Analytiques, ICSN, CNRS, Université Paris Saclay, 1 avenue de la terrasse, 91190, Gif sur Yvette, France
| | - Benoît Crousse
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Julia Kaffy
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| | - Sandrine Ongeri
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400, Orsay, France
| |
Collapse
|
14
|
Liang L, Zhang Y, Zhu Y, Bai J, Ni Y, Wan J, Yue H, Zhao Q, Li H. Structures and Dynamics of β-Rich Oligomers of ATTR (105-115) Assembly. ACS Chem Neurosci 2024; 15:1356-1365. [PMID: 38483181 DOI: 10.1021/acschemneuro.3c00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Transthyretin (TTR) is a tetrameric homologous protein that can dissociate into monomers. Misfolding and aggregation of TTR can lead to amyloid transthyretin amyloidosis (ATTR), which can cause many diseases (e.g., senile systemic amyloidosis, familial amyloid cardiomyopathy, and familial amyloid polyneuropathy). Despite growing evidence indicating that small oligomers play a critical role in regulating cytotoxicity, the structures of these oligomeric intermediates and their conformational transformations are still unclear, impeding our understanding of neurodegenerative mechanisms and the development of therapeutics targeting early aggregation species. The TTR monomer protein consists of various fragments prone to self-aggregation, including the residue 105-115 sequence. Therefore, our study investigated the assembly progress of ATTR (105-115) peptides using all-atom molecular dynamics simulations. The findings indicate that the probability of β-sheet content increases with increasing numbers of peptides. Additionally, interactions between hydrophobic residues L110 and L111 are crucial for the formation of a β-rich oligomer formation. These β-rich oligomers may adopt β-barrel conformations, potentially toxic oligomer species. Free-energy analysis reveals that β-barrel conformations serve as intermediates for these β-rich oligomers. Our insights into the structural ensemble dynamics of ATTR (105-115) contribute to understanding the physical mechanisms underlying the β-barrel oligomers of ATTR. These findings may shed light on the pathological role of ATTR in neurodegenerative diseases and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Liqun Liang
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Yuqi Zhang
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Yanyan Zhu
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Juxia Bai
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Yangyang Ni
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Junfeng Wan
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| | - Haiyan Yue
- Naval Medical Center of PLA, Department of Gastroenterology, Naval Medical University, Shanghai 200433, China
| | - Qingjie Zhao
- Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Huiyu Li
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai 200090, China
| |
Collapse
|
15
|
Akdag M, van Schijndel V, Sinnige T. Islet amyloid polypeptide tagged with green fluorescent protein localises to mitochondria and forms filamentous aggregates in Caenorhabditis elegans. Biophys Chem 2024; 307:107180. [PMID: 38241827 DOI: 10.1016/j.bpc.2024.107180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Type 2 diabetes (T2D) is the most common form of diabetes and represents a growing health concern. A characteristic feature of T2D is the aggregation of islet amyloid polypeptide (IAPP), which is thought to be associated with the death of pancreatic β-cells. Inhibiting IAPP aggregation is a promising therapeutic avenue to treat T2D, but the mechanisms of aggregation and toxicity are not yet fully understood. Caenorhabditis elegans is a well-characterised multicellular model organism that has been extensively used to study protein aggregation diseases. In this study, we aimed to develop a simple in vivo model to investigate IAPP aggregation and toxicity based on expression in the C. elegans body wall muscle cells. We show that IAPP tagged with green fluorescent protein (GFP) localises to mitochondria not only in muscle cells but also when expressed in the intestine, in line with previous observations in mouse and human pancreatic β-cells. The IAPP-GFP fusion protein forms solid aggregates, which have a filamentous appearance as seen by electron microscopy. However, the animals expressing IAPP-GFP in the body wall muscle cells do not display a strong motility phenotype, suggesting that the IAPP-GFP aggregates are not considerably toxic. Nevertheless, the mitochondrial localisation and aggregate formation may be useful read-outs to screen for IAPP-solubilizing compounds as a therapeutic strategy for T2D.
Collapse
Affiliation(s)
- Mehmet Akdag
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Vera van Schijndel
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Tessa Sinnige
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands.
| |
Collapse
|
16
|
Abioye RO, Udenigwe CC. Structural basis and functional significance of food-derived inhibitors of islet amyloid polypeptide fibrillation toward antidiabetic effects. Curr Opin Food Sci 2024; 56:101146. [DOI: 10.1016/j.cofs.2024.101146] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Sarkar D, Saha S, Krishnamoorthy J, Bhunia A. Application of singular value decomposition analysis: Insights into the complex mechanisms of amyloidogenesis. Biophys Chem 2024; 306:107157. [PMID: 38184980 DOI: 10.1016/j.bpc.2023.107157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
Amyloidogenesis, with its multifaceted nature spanning from peptide self-assembly to membrane-mediated structural transitions, presents a significant challenge for the interdisciplinary scientific community. Here, we emphasize on how Singular Value Decomposition (SVD) can be employed to reveal hidden patterns and dominant modes of interaction that govern the complex process of amyloidogenesis. We first utilize SVD analysis on Circular Dichroism (CD) spectral datasets to identify the intermediate structural species emerging during peptide-membrane interactions and to determine binding constants more precisely than conventional methods. We investigate the monomer loss kinetics associated with peptide self-assembly using Nuclear Magnetic Resonance (NMR) dataset and determine the global kinetic parameters through SVD. Furthermore, we explore the seeded growth of amyloid fibrils by analyzing a time-dependent NMR dataset, shedding light on the kinetic intricacies of this process. Our analysis uncovers two distinct states in the aggregation of Aβ40 and pinpoints key residues responsible for this seeded growth. To strengthen our findings and enhance their robustness, we validate those using simulated data, thereby highlighting the physical interpretations derived from SVD. Overall, SVD analysis offers a model-free, global kinetic perspective, enabling the selection of optimal kinetic models. This study not only contributes valuable insights into the dynamics but also highlights the versatility of SVD in unravelling complex processes of amyloidogenesis.
Collapse
Affiliation(s)
- Dibakar Sarkar
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Salt Lake, Sector V, Kolkata 700 091, India
| | - Sudipto Saha
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, Salt Lake, Sector V, Kolkata 700 091, India
| | | | - Anirban Bhunia
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Salt Lake, Sector V, Kolkata 700 091, India.
| |
Collapse
|
18
|
Qiao Q, Wei G, Song Z. Structural diversity in the membrane-bound hIAPP dimer correlated with distinct membrane disruption mechanisms. Phys Chem Chem Phys 2024; 26:7090-7102. [PMID: 38345763 DOI: 10.1039/d3cp05887e] [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: 02/23/2024]
Abstract
Amyloid deposits of the human islet amyloid polypeptide (hIAPP) have been identified in 90% of patients with type II diabetes. Cellular membranes accelerate the hIAPP fibrillation, and the integrity of membranes is also disrupted at the same time, leading to the apoptosis of β cells in pancreas. The molecular mechanism of hIAPP-induced membrane disruption, especially during the initial membrane disruption stage, has not been well understood yet. Herein, we carried out extensive all-atom molecular dynamics simulations investigating the hIAPP dimerization process in the anionic POPG membrane, to provide the detailed molecular mechanisms during the initial hIAPP aggregation stage in the membrane environment. Compared to the hIAPP monomer on the membrane, we observed not only an increase of α-helical structures, but also a substantial increase of β-sheet structures upon spontaneous dimerization. Moreover, the random coiled and α-helical dimer structures insert deep into the membrane interior with a few inter-chain contacts at the C-terminal region, while the β-sheet-rich structures reside on the membrane surface accompanied by strong inter-chain hydrophobic interactions. The coexistence of α and β structures constitutes a diverse structural ensemble of the membrane-bound hIAPP dimer. From α-helical to β-sheet structures, the degree of membrane disruption decreases gradually, and thus the membrane damage induced by random coiled and α-helical structures precedes that induced by β-sheet structures. We speculate that insertion of random coiled and α-helical structures contributes to the initial stage of membrane damage, while β-sheet structures on the membrane surface are more involved in the later stage of fibril-induced membrane disruption.
Collapse
Affiliation(s)
- Qin Qiao
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
- Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai 200032, China
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Science (Ministry of Education), Fudan University, Shanghai 200438, China
| | - Zhijian Song
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
- Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai 200032, China
| |
Collapse
|
19
|
Santos N, Segura L, Lewis A, Pham T, Cheng KH. Multiscale Modeling of Macromolecular Interactions between Tau-Amylin Oligomers and Asymmetric Lipid Nanodomains That Link Alzheimer's and Diabetic Diseases. Molecules 2024; 29:740. [PMID: 38338484 PMCID: PMC10856442 DOI: 10.3390/molecules29030740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/17/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024] Open
Abstract
The molecular events of protein misfolding and self-aggregation of tau and amylin are associated with the progression of Alzheimer's and diabetes, respectively. Recent studies suggest that tau and amylin can form hetero-tau-amylin oligomers. Those hetero-oligomers are more neurotoxic than homo-tau oligomers. So far, the detailed interactions between the hetero-oligomers and the neuronal membrane are unknown. Using multiscale MD simulations, the lipid binding and protein folding behaviors of hetero-oligomers on asymmetric lipid nanodomains or raft membranes were examined. Our raft membranes contain phase-separated phosphatidylcholine (PC), cholesterol, and anionic phosphatidylserine (PS) or ganglioside (GM1) in one leaflet of the lipid bilayer. The hetero-oligomers bound more strongly to the PS and GM1 than other lipids via the hydrophobic and hydrophilic interactions, respectively, in the raft membranes. The hetero-tetramer disrupted the acyl chain orders of both PC and PS in the PS-containing raft membrane, but only the GM1 in the GM1-containing raft membrane as effectively as the homo-tau-tetramer. We discovered that the alpha-helical content in the heterodimer was greater than the sum of alpha-helical contents from isolated tau and amylin monomers on both raft membranes, indicative of a synergetic effect of tau-amylin interactions in surface-induced protein folding. Our results provide new molecular insights into understanding the cross-talk between Alzheimer's and diabetes.
Collapse
Affiliation(s)
- Natalia Santos
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA; (N.S.); (L.S.); (A.L.)
| | - Luthary Segura
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA; (N.S.); (L.S.); (A.L.)
| | - Amber Lewis
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA; (N.S.); (L.S.); (A.L.)
| | - Thuong Pham
- Physics Department, Trinity University, San Antonio, TX 78212, USA;
| | - Kwan H. Cheng
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA; (N.S.); (L.S.); (A.L.)
- Physics Department, Trinity University, San Antonio, TX 78212, USA;
| |
Collapse
|
20
|
Yang B, Lu T, Wang S, Li C, Li C, Li F. Interfacial effect on the ability of peptide-modified gold nanoclusters to inhibit hIAPP fibrillation and cytotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184202. [PMID: 37541643 DOI: 10.1016/j.bbamem.2023.184202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023]
Abstract
Deposit of amyloid peptides in the cells is related to various amyloidosis diseases. A variety of nanomaterials have been developed to resist amyloid deposit. Most of the research on the inhibition of nanomaterials against amyloid aggregation are undertaken in solution, while the membranes that may mediate fibrillar aggregation and affect interaction of inhibitors with amyloid peptides in biotic environment are little taken into account. In this study, we synthesized three kinds of gold nanoclusters modified with cysteine (C@AuNCs), glutathione (GSH@AuNCs) and a peptide derived from the core region of hIAPP fibrillation (C-HL-8P@AuNCs), and investigated their inhibitory activities against hIAPP fibrillation in the absence and presence of lipid vesicles (POPC/POPG 4:1 LUVs) by the experiments of ThT fluorescence kinetics, AFM and CD. We also explored the inhibitions of hIAPP-induced membrane damage and cytotoxicity by peptide@AuNCs using fluorescent dye leakage and cell viability assays. Our study revealed that the inhibitory efficiency of these peptide@AuNCs against hIAPP fibrillation follows C-HL-8P@AuNCs≅GSH@AuNCs>C@AuNCs in lipid-free solution and C-HL-8P@AuNCs≫GSH@AuNCs>C@AuNCs in lipid membrane environment. Compared with the results obtained in lipid-free solution, the inhibitions of hIAPP fibrillation observed in lipid membrane environment were more associated with the inhibitions of hIAPP-induced damages of lipid vesicles and INS-1 cells (C-HL-8P@AuNCs≫GSH@AuNCs>C@AuNCs). An additional hydrophobic interaction with the homologous core region of hIAPP, which is only provided by C-HL-8P@AuNCs and largely suppressed in lipid-free solution, enhanced in the membrane environment and therefore made C-HL-8P@AuNCs much more powerful than GSH@AuNCs and C@AuNCs in the inhibitions of hIAPP fibrillation and cytotoxicity.
Collapse
Affiliation(s)
- Boqi Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Tong Lu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Shuyu Wang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Chengyao Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Chen Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China.
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China.
| |
Collapse
|
21
|
Regmi D, Shen F, Stanic A, Islam M, Du D. Effect of phospholipid liposomes on prion fragment (106-128) amyloid formation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184199. [PMID: 37454869 DOI: 10.1016/j.bbamem.2023.184199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Misfolding and aggregation of cellular prion protein (PrPc) is a major molecular process involved in the pathogenesis of prion diseases. Here, we studied the aggregation properties of a prion fragment peptide PrP(106-128). The results show that the peptide aggregates in a concentration-dependent manner in an aqueous solution and that the aggregation is sensitive to pH and the preformed amyloid seeds. Furthermore, we show that the zwitterionic POPC liposomes moderately inhibit the aggregation of PrP(106-128), whereas POPC/cholesterol (8:2) vesicles facilitate peptide aggregation likely due to the increase of the lipid packing order and membrane rigidity in the presence of cholesterol. In addition, anionic lipid vesicles of POPG and POPG/cholesterol above a certain concentration accelerate the aggregation of the peptide remarkably. The strong electrostatic interactions between the N-terminal region of the peptide and POPG may constrain the conformational plasticity of the peptide, preventing insertion of the peptide into the inner side of the membrane and thus promoting fibrillation on the membrane surface. The results suggest that the charge properties of the membrane, the composition of the liposomes, and the rigidity of lipid packing are critical in determining peptide adsorption on the membrane surface and the efficiency of the membrane in catalyzing peptide oligomeric nucleation and amyloid formation. The peptide could be used as an improved model molecule to investigate the mechanistic role of the crucial regions of PrP in aggregation in a membrane-rich environment and to screen effective inhibitors to block key interactions between these regions and membranes for preventing PrP aggregation.
Collapse
Affiliation(s)
- Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Fengyun Shen
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Aleksander Stanic
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Majedul Islam
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
22
|
Muñoz-Gutiérrez C, Adasme-Carreño F, Alzate-Morales J, Ireta J. Effect of strand register in the stability and reactivity of crystals from peptides forming amyloid fibrils. Phys Chem Chem Phys 2023; 25:23885-23893. [PMID: 37642522 DOI: 10.1039/d3cp01762a] [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: 08/31/2023]
Abstract
Amyloids are cytotoxic protein aggregates that deposit in human tissues, leading to several health disorders. Their aggregates can also exhibit catalytic properties, and they have been used as candidates for the development of functional biomaterials. Despite being polymorphic, amyloids often assemble as cross-β fibrils formed by in-register β sheet layers. Recent studies of some amyloidogenic protein segments revealed that they crystallize as antiparallel out-of-register β sheets. Such arrangement has been proposed to be responsible for the cytotoxicity in amyloid diseases, however, there is still no consensus on the molecular mechanism. Interestingly, two amyloidogenic peptide segments, NFGAILS and FGAILSS, arrange into out-of-register and in-register β sheets, respectively, even though they solely differ by one aminoacid residue at both termini. In this work, we used density functional theory (DFT) to address how the strand register contributes into the packing and molecular properties of the NFGAILS and FGAILSS crystals. Our results show that the out-of-register structure is substantially more stable, at 0 K, than the in-register one due to stronger inter-strand contacts. Based on an analysis of the electrostatic potential of the crystal slabs, it is suggested that the out-of-register may potentially interact with negatively charged groups, like those found in cell membranes. Moreover, calculated reactivity descriptors indicate a similar outcome, where only the out-of-register peptide exhibits intrinsic reactive surface sites at the exposed amine and carboxylic groups. It is therefore suggested that the out-of-register arrangement may indeed be crucial for amyloid cytotoxicity. The findings presented here could help to further our understanding of amyloid aggregation, function, and toxicity.
Collapse
Affiliation(s)
- Camila Muñoz-Gutiérrez
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Campus Talca, 1 Poniente No. 1141, Box 721, Talca, Chile
| | - Francisco Adasme-Carreño
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca 3480112, Chile
- Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480112, Chile
| | - Jans Alzate-Morales
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Campus Talca, 1 Poniente No. 1141, Box 721, Talca, Chile
| | - Joel Ireta
- Departamento de Química, División de Ciencias Básicas e Ingeniería, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-534, Ciudad de México 09340, Mexico.
| |
Collapse
|
23
|
Kaur A, Goyal B. In silico design and identification of new peptides for mitigating hIAPP aggregation in type 2 diabetes. J Biomol Struct Dyn 2023; 42:10006-10021. [PMID: 37691445 DOI: 10.1080/07391102.2023.2254411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/27/2023] [Indexed: 09/12/2023]
Abstract
The aberrant misfolding and self-aggregation of human islet amyloid polypeptide (hIAPP or amylin) into cytotoxic aggregates are implicated in the pathogenesis of type 2 diabetes (T2D). Among various inhibitors, short peptides derived from the amyloidogenic regions of hIAPP have been employed as hIAPP aggregation inhibitors due to their low immunogenicity, biocompatibility, and high chemical diversity. Recently, hIAPP fragment HSSNN18-22 was identified as an amyloidogenic sequence and displayed higher antiproliferative activity to RIN-5F cells. Various hIAPP aggregation inhibitors have been designed by chemical modifications of the highly amyloidogenic sequence (NFGAIL) of hIAPP. In this work, a library of pentapeptides based on fragment HSSNN18-22 was designed and assessed for their efficacy in blocking hIAPP aggregation using an integrated computational screening approach. The binding free energy calculations by molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method identified HSSQN and HSSNQ that bind to hIAPP monomer with a binding affinity of -21.25 ± 4.90 and -19.73 ± 3.10 kcal/mol, respectively, which is notably higher as compared to HSSNN (-11.90 ± 4.12 kcal/mol). The sampling of the non aggregation-prone helical conformation was notably increased from 23.5 ± 3.0 in the hIAPP monomer to 38.1 ± 3.6, and 33.8 ± 3.0% on the incorporation of HSSQN, and HSSNQ, respectively, which indicate reduced aggregation propensity of hIAPP monomer. The pentapeptides, HSSQN and HSSNQ, identified as hIAPP aggregation inhibitors in this work can be further conjugated with various metal chelating peptides to yield more efficacious and clinically relevant multifunctional modulators for targeting various pathological hallmarks of T2D.
Collapse
Affiliation(s)
- Apneet Kaur
- School of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, India
| | - Bhupesh Goyal
- School of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, India
| |
Collapse
|
24
|
Li MH, Zhang X, London E, Raleigh DP. Impact of Ca 2+ on membrane catalyzed IAPP amyloid formation and IAPP induced vesicle leakage. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184161. [PMID: 37121365 PMCID: PMC10735052 DOI: 10.1016/j.bbamem.2023.184161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
Human islet amyloid polypeptide (hIAPP, also known as amylin) is a 37 amino acid pancreatic polypeptide hormone that plays a role in regulating glucose levels, but forms pancreatic amyloid in type-2 diabetes. The process of amyloid formation by hIAPP contributes to β-cell death in the disease. Multiple mechanisms of hIAPP induced toxicity of β-cells have been proposed including disruption of cellular membranes. However, the nature of hIAPP membrane interactions and the effect of ions and other molecules on hIAPP membrane interactions are not fully understood. Many studies have used model membranes with a high content of anionic lipids, often POPS, however the concentration of anionic lipids in the β-cell plasma membrane is low. Here we study the concentration dependent effect of Ca2+ (0 to 50 mM) on hIAPP membrane interactions using large unilamellar vesicles (LUVs) with anionic lipid content ranging from 0 to 50 mol%. We find that Ca2+ does not effectively inhibit hIAPP amyloid formation and hIAPP induced membrane leakage from binary LUVs with a low percentage of POPS, but has a greater effect on LUVs with a high percentage of POPS. Mg2+ had very similar effects, and the effects of Ca2+ and Mg2+ can be largely rationalized by the neutralization of POPS charge. The implications for hIAPP-membrane interactions are discussed.
Collapse
Affiliation(s)
- Ming-Hao Li
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Xiaoxue Zhang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States
| | - Erwin London
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States; Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States; Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, United States.
| | - Daniel P Raleigh
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States; Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States.
| |
Collapse
|
25
|
Sanders HM, Chalyavi F, Fields CR, Kostelic MM, Li MH, Raleigh DP, Zanni MT, Marty MT. Interspecies Variation Affects Islet Amyloid Polypeptide Membrane Binding. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:986-990. [PMID: 37126782 PMCID: PMC10330443 DOI: 10.1021/jasms.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The aggregation of islet amyloid polypeptide (IAPP) is associated with β-cell dysfunction in type 2 diabetes (T2D) in humans. One possible mechanism of toxicity is the interaction of IAPP oligomers with lipid membranes to disrupt the bilayer integrity and/or homeostasis of the cell. Amino acid sequence variations of IAPPs between species can greatly decrease their propensity for aggregation. For example, human IAPP is toxic to β-cells, but rat and pig IAPP are not. However, it is not clear how these differences affect membrane association. Using native mass spectrometry with lipid nanodiscs, we explored the differences in the association of human, rat, and pig IAPP with lipid bilayers. We discovered that human and rat IAPP bound nanodiscs with anionic dipalmitoyl-phosphatidylglycerol (DPPG) lipids, but pig IAPP did not. Furthermore, human and rat IAPP interacted differently with the membrane. Human IAPP show potential tetramer complexes, but rat IAPP associated with the membrane sequentially. Thus, overall IAPP-bilayer interactions are not necessarily related to disease, but small differences in oligomeric behavior at the membrane may instead play a role.
Collapse
Affiliation(s)
- Henry M. Sanders
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Farzaneh Chalyavi
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Caitlyn R. Fields
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Marius M. Kostelic
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Ming-Hao Li
- Department of Chemistry and Laufer Center for Quantitative Biology, Stony Brook University, 100 Nicolls Rd., Stony Brook, New York 11794, USA
| | - Daniel P. Raleigh
- Department of Chemistry and Laufer Center for Quantitative Biology, Stony Brook University, 100 Nicolls Rd., Stony Brook, New York 11794, USA
| | - Martin T. Zanni
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael T. Marty
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
26
|
Nguyen N, Lewis A, Pham T, Sikazwe D, Cheng KH. Exploring the Role of Anionic Lipid Nanodomains in the Membrane Disruption and Protein Folding of Human Islet Amyloid Polypeptide Oligomers on Lipid Membrane Surfaces Using Multiscale Molecular Dynamics Simulations. Molecules 2023; 28:4191. [PMID: 37241931 PMCID: PMC10223233 DOI: 10.3390/molecules28104191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The aggregation of human Islet Amyloid Polypeptide (hIAPP) on cell membranes is linked to amyloid diseases. However, the physio-chemical mechanisms of how these hIAPP aggregates trigger membrane damage are unclear. Using coarse-grained and all-atom molecular dynamics simulations, we investigated the role of lipid nanodomains in the presence or absence of anionic lipids, phosphatidylserine (PS), and a ganglioside (GM1), in the membrane disruption and protein folding behaviors of hIAPP aggregates on phase-separated raft membranes. Our raft membranes contain liquid-ordered (Lo), liquid-disordered (Ld), mixed Lo/Ld (Lod), PS-cluster, and GM1-cluster nanosized domains. We observed that hIAPP aggregates bound to the Lod domain in the absence of anionic lipids, but also to the GM1-cluster- and PS-cluster-containing domains, with stronger affinity in the presence of anionic lipids. We discovered that L16 and I26 are the lipid anchoring residues of hIAPP binding to the Lod and PS-cluster domains. Finally, significant lipid acyl chain order disruption in the annular lipid shells surrounding the membrane-bound hIAPP aggregates and protein folding, particularly beta-sheet formation, in larger protein aggregates were evident. We propose that the interactions of hIAPP and both non-anionic and anionic lipid nanodomains represent key molecular events of membrane damage associated with the pathogenesis of amyloid diseases.
Collapse
Affiliation(s)
- Ngoc Nguyen
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
| | - Amber Lewis
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA;
| | - Thuong Pham
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
| | - Donald Sikazwe
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA;
| | - Kwan H. Cheng
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA;
| |
Collapse
|
27
|
Lewis A, Pham T, Nguyen N, Graf A, Cheng KH. Lipid domain boundary triggers membrane damage and protein folding of human islet amyloid polypeptide in the early pathogenesis of amyloid diseases. Biophys Chem 2023; 296:106993. [PMID: 36898349 DOI: 10.1016/j.bpc.2023.106993] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
The misfolding and self-aggregation of human Islet Amyloid Polypeptide (hIAPP) are linked to the onset of type 2 diabetes (T2D). However, the mechanism of how the disordered hIAPP aggregates trigger membrane damage leading to the loss of Islet cells in T2D is unknown. Using coarse-grained (CG) and all-atom (AA) molecular dynamics simulations, we have investigated the membrane-disruption behaviors of hIAPP oligomers on the phase-separated lipid nanodomains that mimic the highly heterogeneous lipid raft structures of cell membranes. Our results revealed that hIAPP oligomers preferentially bind to the liquid-ordered and liquid-disordered domain boundary around two hydrophobic residues at L16 and I26, and lipid acyl chain order disruption and beta-sheet formation occur upon hIAPP binding to the membrane surface. We propose that the lipid order disruption and surface-induced beta-sheet formation on the lipid domain boundary represent the early molecular events of membrane damage associated with the early pathogenesis of T2D.
Collapse
Affiliation(s)
- Amber Lewis
- Neuroscience Dept., Trinity University, San Antonio, TX, USA
| | - Thuong Pham
- Physics Dept., Trinity University, San Antonio, TX, USA
| | - Ngoc Nguyen
- Physics Dept., Trinity University, San Antonio, TX, USA
| | - Angela Graf
- Physics Dept., Trinity University, San Antonio, TX, USA
| | - Kwan H Cheng
- Neuroscience Dept., Trinity University, San Antonio, TX, USA; Physics Dept., Trinity University, San Antonio, TX, USA.
| |
Collapse
|
28
|
Naito A, Kawamura I. Dynamic membrane interaction and amyloid fibril formation of glucagon, melittin and human calcitonin. Biophys Chem 2023; 298:107025. [PMID: 37127008 DOI: 10.1016/j.bpc.2023.107025] [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: 03/01/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Glucagon is a 29-amino acid peptide hormone secreted by pancreatic α-cells and interacts with specific receptors located in various organs. Glucagon tends to form gel-like fibril aggregates that are cytotoxic. It is important to reveal the glucagon-membrane interaction to understand activity and cytotoxicity of glucagon and glucagon oligomers. In this review, first glucagon-membrane interactions are described as morphological changes in dimyristoylphosphatidylcholine (DMPC) bilayers containing glucagon in acidic and neutral conditions as compared to the case of melittin. Second, fibril formation by glucagon in acidic solution is discussed in light of morphological and structural changes. Third, kinetic analysis of glucagon fibril formation was performed using a two-step autocatalytic reaction mechanism, as investigated in the case of human calcitonin. The first step is a nuclear formation, and the second step is an autocatalytic fibril elongation. Forth, fibril formation of glucagon inside glucagon-DMPC bilayers in neutral solution under near physiological condition is described.
Collapse
Affiliation(s)
- Akira Naito
- Graduate School of Engineering, Yokohama National University, Yokohama 240-8501, Japan.
| | - Izuru Kawamura
- Graduate School of Engineering, Yokohama National University, Yokohama 240-8501, Japan
| |
Collapse
|
29
|
Zinc and iron dynamics in human islet amyloid polypeptide-induced diabetes mouse model. Sci Rep 2023; 13:3484. [PMID: 36922503 PMCID: PMC10017767 DOI: 10.1038/s41598-023-30498-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Metal homeostasis is tightly regulated in cells and organisms, and its disturbance is frequently observed in some diseases such as neurodegenerative diseases and metabolic disorders. Previous studies suggest that zinc and iron are necessary for the normal functions of pancreatic β cells. However, the distribution of elements in normal conditions and the pathophysiological significance of dysregulated elements in the islet in diabetic conditions have remained unclear. In this study, to investigate the dynamics of elements in the pancreatic islets of a diabetic mouse model expressing human islet amyloid polypeptide (hIAPP): hIAPP transgenic (hIAPP-Tg) mice, we performed imaging analysis of elements using synchrotron scanning X-ray fluorescence microscopy and quantitative analysis of elements using inductively coupled plasma mass spectrometry. We found that in the islets, zinc significantly decreased in the early stage of diabetes, while iron gradually decreased concurrently with the increase in blood glucose levels of hIAPP-Tg mice. Notably, when zinc and/or iron were decreased in the islets of hIAPP-Tg mice, dysregulation of glucose-stimulated mitochondrial respiration was observed. Our findings may contribute to clarifying the roles of zinc and iron in islet functions under pathophysiological diabetic conditions.
Collapse
|
30
|
Liu XY, Du SC, Li SL, Jiang FL, Jiang P, Liu Y. Inhibition mechanism of human insulin fibrillation by Bodipy carbon polymer dots and photothermal defibrillation effect of Bodipy carbon polymer dots modified by ThT. Biophys Chem 2023; 297:107009. [PMID: 37037121 DOI: 10.1016/j.bpc.2023.107009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023]
Abstract
Fibrillation process of human insulin (HI) is closely related to type 2 diabetes (T2D). In the present work, Carbon Polymer Dots (CPDs) was synthesized by Bodipy to control the process of insulin fibrillation. The inhibition process of insulin fibrillation with the existence of CPDs was completed investigated. The hydrophobic interaction of CPDs and insulin was used to inhibit the change of insulin's secondary structure in the lag phase and growth period. ThT fluorescence analysis and transmission electron microscopy (TEM) characterization of the CPDs were used to explore the kinetics of insulin fibrillation and regulation process by CPDs. Isothermal titration calorimetry (ITC) was applied to explore the regulatory mechanism by CPDs at all stages of the insulin fibrillation process. ThT was used to complete the chemical modification of CPDs by Friedel-Crafts alkylation, which made the CPDs maintain the characteristics of photothermal effect and also obtain the ability to bind specifically to the fibers. Finally, the process of defibrillation of human insulin fibers under the Near-infrared light's irradiation was realized. In this work, we clarified the mechanism of the regulation process by Bodipy CPDs and made CPDs able to defibrillate the insulin fibers by chemical modification.
Collapse
|
31
|
Ma L, Li X, Petersen RB, Peng A, Huang K. Probing the interactions between amyloidogenic proteins and bio-membranes. Biophys Chem 2023; 296:106984. [PMID: 36889133 DOI: 10.1016/j.bpc.2023.106984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023]
Abstract
Protein misfolding diseases (PMDs) in humans are characterized by the deposition of protein aggregates in tissues, including Alzheimer's disease, Parkinson's disease, type 2 diabetes, and amyotrophic lateral sclerosis. Misfolding and aggregation of amyloidogenic proteins play a central role in the onset and progression of PMDs, and these processes are regulated by multiple factors, especially the interaction between proteins and bio-membranes. Bio-membranes induce conformational changes in amyloidogenic proteins and affect their aggregation; on the other hand, the aggregates of amyloidogenic proteins may cause membrane damage or dysfunction leading to cytotoxicity. In this review, we summarize the factors that affect the binding of amyloidogenic proteins and membranes, the effects of bio-membranes on the aggregation of amyloidogenic proteins, mechanisms of membrane disruption by amyloidogenic aggregates, technical approaches for detecting these interactions, and finally therapeutic strategies targeting membrane damage caused by amyloidogenic proteins.
Collapse
Affiliation(s)
- Liang Ma
- Department of Pharmacy, Wuhan Mental Health Center, Wuhan, China; Department of Pharmacy, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Xi Li
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan, China.
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
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
|
33
|
Understanding the mechanism of amylin aggregation: From identifying crucial segments to tracing dominant sequential events to modeling potential aggregation suppressors. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2023; 1871:140866. [PMID: 36272537 DOI: 10.1016/j.bbapap.2022.140866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022]
Abstract
One of the most abundant, prevailing, and life-threatening human diseases that are currently baffling the scientific community is type 2 diabetes (T2D). The self-association of human amylin has been implicated in the pathogenesis of T2D, though with an inconclusive understanding of the mechanism. Hence, we focused on the characterization of the conformational ensembles of all the species that are believed to define the structural polymorphism of the aggregation process - the functional monomeric, the initially self-associated oligomeric, and the structured protofibril - by employing near-equilibrium, non-equilibrium, and equilibrium atomistic simulations on the sporadic, two familial variants (S20G and G33R), and their proline-substituted forms (S20P and G33P). The dynamic near-equilibrium assays hint toward - the abundance of helical conformation in the monomeric state, the retainment of the helicity in the initial self-associated oligomeric phase pointing toward the existence of the helix-helix association mechanism, the difference in preference of specific segments to have definite secondary structural features, the phase-dependent variability in the dominance of specific segments and mutation sites, and the simultaneous presence of generic and unique features among various sequences. Furthermore, the non-equilibrium pulling assays exemplify a generic sequential unzipping mechanism of the protofibrils, however, the sequence-dependent uniqueness comes from the difference in location and magnitude of the control of a specific terminus. Importantly, the equilibrium thermodynamic assays efficiently rank order the potential of aggregability among sequences and consequently suggests the probability of designing effective aggregation suppressors against sporadic and familial amylin variants incorporating proline as the mutation.
Collapse
|
34
|
Bortoletto AS, Graham WV, Trout G, Bonito‐Oliva A, Kazmi MA, Gong J, Weyburne E, Houser BL, Sakmar TP, Parchem RJ. Human Islet Amyloid Polypeptide (hIAPP) Protofibril-Specific Antibodies for Detection and Treatment of Type 2 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202342. [PMID: 36257905 PMCID: PMC9731688 DOI: 10.1002/advs.202202342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/26/2022] [Indexed: 05/27/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a major public health concern and is characterized by sustained hyperglycemia due to insulin resistance and destruction of insulin-producing β cells. One pathological hallmark of T2D is the toxic accumulation of human islet amyloid polypeptide (hIAPP) aggregates. Monomeric hIAPP is a hormone normally co-secreted with insulin. However, increased levels of hIAPP in prediabetic and diabetic patients can lead to the formation of hIAPP protofibrils, which are toxic to β cells. Current therapies fail to address hIAPP aggregation and current screening modalities do not detect it. Using a stabilizing capping protein, monoclonal antibodies (mAbs) can be developed against a previously nonisolatable form of hIAPP protofibrils, which are protofibril specific and do not engage monomeric hIAPP. Shown here are two candidate mAbs that can detect hIAPP protofibrils in serum and hIAPP deposits in pancreatic islets in a mouse model of rapidly progressing T2D. Treatment of diabetic mice with the mAbs delays disease progression and dramatically increases overall survival. These results demonstrate the potential for using novel hIAPP protofibril-specific mAbs as a diagnostic screening tool for early detection of T2D, as well as therapeutically to preserve β cell function and target one of the underlying pathological mechanisms of T2D.
Collapse
Affiliation(s)
- Angelina S. Bortoletto
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| | - W. Vallen Graham
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Gabriella Trout
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| | - Alessandra Bonito‐Oliva
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Manija A. Kazmi
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Jing Gong
- Celdara Medical16 Cavendish CourtLebanonNH03766USA
| | | | | | - Thomas P. Sakmar
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
- Department of Neurobiology, Care Sciences, and SocietyCenter for Alzheimer ResearchDivision of NeurogeriatricsKarolinska InstitutetSolna17164Sweden
| | - Ronald J. Parchem
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| |
Collapse
|
35
|
Fortier M, Côté-Cyr M, Nguyen V, Babych M, Nguyen PT, Gaudreault R, Bourgault S. Contribution of the 12–17 hydrophobic region of islet amyloid polypeptide in self-assembly and cytotoxicity. Front Mol Biosci 2022; 9:1017336. [PMID: 36262476 PMCID: PMC9573943 DOI: 10.3389/fmolb.2022.1017336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The islet amyloid polypeptide (IAPP) is a 37-residue aggregation-prone peptide hormone whose deposition as insoluble fibrils in the islets of Langerhans is associated with type II diabetes. Therapeutic interventions targeting IAPP amyloidogenesis, which contributes to pancreatic β-cell degeneration, remain elusive owing to the lack of understanding of the self-assembly mechanisms and of the quaternary proteospecies mediating toxicity. While countless studies have investigated the contributions of the 20–29 amyloidogenic core in self-assembly, IAPP central region, i.e. positions 11 to 19, has been less studied, notwithstanding its potential key role in oligomerization. In this context, the present study aimed at investigating the physicochemical and conformational properties driving IAPP self-assembly and associated cytotoxicity. Computational tools and all-atom molecular dynamics simulation suggested that the hydrophobic 12–17 segment promotes IAPP self-recognition and aggregation. Alanine scanning revealed that the hydrophobic side chains of Leu12, Phe15 and Val17 are critical for amyloid fibril formation. Destabilization of the α-helical folding by Pro substitution enhanced self-assembly when the pyrrolidine ring was successively introduced at positions Ala13, Asn14 and Phe15, in comparison to respective Ala-substituted counterparts. Modulating the peptide backbone flexibility at position Leu16 through successive incorporation of Pro, Gly and α-methylalanine, inhibited amyloid formation and reduced cytotoxicity, while the isobutyl side chain of Leu16 was not critical for self-assembly and IAPP-mediated toxicity. These results highlight the importance of the 12–17 hydrophobic region of IAPP for self-recognition, ultimately supporting the development of therapeutic approaches to prevent oligomerization and/or fibrillization.
Collapse
Affiliation(s)
- Mathilde Fortier
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
| | - Vy Nguyen
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
| | - Margaryta Babych
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
| | - Phuong Trang Nguyen
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
| | - Roger Gaudreault
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Department of Physics, Université de Montréal, Succursale Centre-ville, Montreal, QC, Canada
- *Correspondence: Roger Gaudreault, ; Steve Bourgault,
| | - Steve Bourgault
- Department of Chemistry, Succursale Centre-Ville, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Montreal, QC, Canada
- *Correspondence: Roger Gaudreault, ; Steve Bourgault,
| |
Collapse
|
36
|
Shardlow E, Brown L, Exley C. The influence of aluminium and copper upon the early aggregatory behaviour and size of Islet amyloid polypeptide under simulated physiological conditions. J Trace Elem Med Biol 2022; 73:127027. [PMID: 35868166 DOI: 10.1016/j.jtemb.2022.127027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIM Islet amyloid polypeptide/amylin deposition in the form of amyloid plaques is a common pathological feature observed in the pancreatic tissue of those with Type II Diabetes Mellitus. Its propensity to form amyloid fibrils and the resultant toxicity of this peptide in vivo is influenced by both the concentration and species of metal present in situ. Herein, we examine the influence of Al (III) and Cu (II), applied at equimolar and supra-stoichiometric concentrations on the initial aggregatory behaviour of amylin under near physiological conditions. METHODS Dynamic light scattering measurements, which monitored the aggregation status and size of the peptide in real time, were performed during the early lag-phase of fibrillogenesis (T ≤ 30 min) in the absence or presence of metal ions. RESULTS Islet amyloid polypeptide (10 µM) rapidly aggregated when introduced into a physiological medium favouring the formation of large, agglomerated structures (> 1000 nm) after 30 min incubation. Neither the addition of equimolar or excess metals significantly influenced the size of the peptide when intensity distributions were consulted; however, number distributions indicated that both Al (III) and Cu (II) may have had, an albeit temporary, stabilising influence upon the conformations present within solution. CONCLUSION These results infer that small oligomeric species are likely transient entities that are rapidly incorporated into large agglomerates during the very initial stages of fibrillogenesis. While both Al (III) and Cu (II) both inhibited agglomeration to some degree, their stabilising affect upon peptide aggregation was limited over the juncture of the experiments performed herein; hence, it is difficult to say whether these metal ions play a role in enhancing the toxicity of these peptides through influencing their aggregation in the short-term.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK.
| | - Lewis Brown
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| |
Collapse
|
37
|
Molecular basis of the anchoring and stabilization of human islet amyloid polypeptide in lipid hydroperoxidized bilayers. Biochim Biophys Acta Gen Subj 2022; 1866:130200. [PMID: 35820640 DOI: 10.1016/j.bbagen.2022.130200] [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: 12/23/2021] [Revised: 06/25/2022] [Accepted: 07/02/2022] [Indexed: 01/18/2023]
Abstract
The molecular structure of membrane lipids is formed by mono- or polyunsaturations on their aliphatic tails that make them susceptible to oxidation, facilitating the incorporation of hydroperoxide (R-OOH) functional groups. Such groups promote changes in both composition and complexity of the membrane significantly modifying its physicochemical properties. Human Langerhans islets amyloid polypeptide (hIAPP) is the main component of amyloid deposits found in the pancreas of patients with type-2 diabetes (T2D). hIAPP in the presence of membranes with oxidized lipid species accelerates the formation of amyloid fibrils or the formation of intermediate oligomeric structures. However, the molecular bases at the initial stage of the anchoring and stabilization of the hIAPP in a hydroperoxidized membrane are not yet well understood. To shed some light on this matter, in this contribution, three bilayer models were modeled: neutral (POPC), anionic (POPS), and oxidized (POPCOOH), and full atom Molecular Dynamics (MD) simulations were performed. Our results show that the POPCOOH bilayer increases the helicity in hIAPP when compared to POPC or POPS bilayer. The modification in the secondary structure covers the residues of the so-called amyloidogenic core of the hIAPP. Overall, the hydroperoxidation of the neutral lipids modifies both the anchoring and the stabilization of the peptide hIAPP by reducing the random conformations of the peptide and increasing of hydrogen bond population with the hydroperoxidized lipids.
Collapse
|
38
|
Dicke SS, Maj M, Fields CR, Zanni MT. Metastable intermediate during hIAPP aggregation catalyzed by membranes as detected with 2D IR spectroscopy. RSC Chem Biol 2022; 3:931-940. [PMID: 35866164 PMCID: PMC9257649 DOI: 10.1039/d2cb00028h] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
The aggregation of human islet amyloid polypeptide (hIAPP) into amyloid fibrils involves formation of oligomeric intermediates that are thought to be the cytotoxic species responsible for β-cell dysfunction in type 2 diabetes. hIAPP oligomers permeating or disrupting the cellular membrane may be one mechanism of toxicity and so measuring the structural kinetics of aggregation in the presence of membranes is of much interest. In this study, we use 2D IR spectroscopy and 13C18O isotope labeling to study the secondary structure of the oligomeric intermediates formed in solution and in the presence of phospholipid vesicles at sites L12A13, L16V17, G24A25 and V32G33. Pairs of labels monitor the couplings between associated polypeptides and the dihedral angles between adjacent residues. In solution, the L12A13 residues form an oligomeric β-sheet in addition to an α-helix whereas with the phospholipid vesicles they are α-helical throughout the aggregation process. In both solution and with DOPC vesicles, L16V17 and V32G33 have disordered structures until fibrils are formed. Similarly, under both conditions, G24A25 exhibits 3-state kinetics, created by an oligomeric intermediate with a well-defined β-sheet structure. Amyloid fibril formation is often thought to involve intermediates with exceedingly low populations that are difficult to detect experimentally. These experiments establish that amyloid fibril formation of hIAPP when catalyzed by membranes includes a metastable intermediate and that this intermediate has a similar structure at G24A25 in the FGAIL region as the corresponding intermediate in solution, thought to be the toxic species. 2D IR and 13C18O isotope labeling establish that amyloid formation of hIAPP catalyzed by membranes includes a metastable intermediate with a similar structure at G24A25 in the FGAIL region as the corresponding intermediate in solution.![]()
Collapse
Affiliation(s)
- Sidney S Dicke
- Department of Chemistry, University of Wisconsin-Madison 1101 University Avenue Madison WI 53706 USA
| | - Michał Maj
- Department of Chemistry, University of Wisconsin-Madison 1101 University Avenue Madison WI 53706 USA .,Formally at Department of Chemistry, University of Wisconsin-Madison 1101 University Avenue Madison WI 53706 USA
| | - Caitlyn R Fields
- Department of Chemistry, University of Wisconsin-Madison 1101 University Avenue Madison WI 53706 USA
| | - Martin T Zanni
- Department of Chemistry, University of Wisconsin-Madison 1101 University Avenue Madison WI 53706 USA
| |
Collapse
|
39
|
Dogan S, Paulus M, Kosfeld BR, Cewe C, Tolan M. Interaction of Human Resistin with Human Islet Amyloid Polypeptide at Charged Phospholipid Membranes. ACS OMEGA 2022; 7:22377-22382. [PMID: 35811869 PMCID: PMC9260898 DOI: 10.1021/acsomega.2c01363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
An X-ray reflectivity study on the interaction of recombinant human resistin (hRes) with fibrillation-prone human islet amyloid polypeptide (hIAPP) at anionic phospholipid Langmuir films as model membranes is presented. Aggregation and amyloid formation of hIAPP is considered the main mechanism of pancreatic β-cell loss in patients with type 2 diabetes mellitus. Resistin shows a chaperone-like ability, but also tends to form aggregates by itself. Resistin and hIAPP cross multiply metabolism pathways. In this study, we researched the potential protective effects of resistin against hIAPP-induced lipid membrane rupture. The results demonstrate that resistin can inhibit or prevent hIAPP adsorption even in the presence of aggregation-promoting negatively charged lipid interfaces. Moreover, we found strong hydrophobic interactions of resistin at the bare buffer-air interface.
Collapse
|
40
|
Abstract
Amyloids are organized suprastructural polypeptide arrangements. The prevalence of amyloid-related processes of pathophysiological relevance has been linked to aging-related degenerative diseases. Besides the role of genetic polymorphisms on the relative risk of amyloid diseases, the contributions of nongenetic ontogenic cluster of factors remain elusive. In recent decades, mounting evidences have been suggesting the role of essential micronutrients, in particular transition metals, in the regulation of amyloidogenic processes, both directly (such as binding to amyloid proteins) or indirectly (such as regulating regulatory partners, processing enzymes, and membrane transporters). The features of transition metals as regulatory cofactors of amyloid proteins and the consequences of metal dyshomeostasis in triggering amyloidogenic processes, as well as the evidences showing amelioration of symptoms by dietary supplementation, suggest an exaptative role of metals in regulating amyloid pathways. The self- and cross-talk replicative nature of these amyloid processes along with their systemic distribution support the concept of their metastatic nature. The role of amyloidosis as nutrient sensors would act as intra- and transgenerational epigenetic metabolic programming factors determining health span and life span, viability, which could participate as an evolutive selective pressure.
Collapse
Affiliation(s)
- Luís Maurício T R Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory for Macromolecules (LAMAC-DIMAV), National Institute of Metrology, Quality and Technology - INMETRO, Duque de Caxias, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Linking hIAPP misfolding and aggregation with type 2 diabetes mellitus: a structural perspective. Biosci Rep 2022; 42:231205. [PMID: 35475576 PMCID: PMC9118370 DOI: 10.1042/bsr20211297] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
There are over 40 identified human disorders that involve certain proteins folding incorrectly, accumulating in the body causing damage to cells and organs and causing disease. Type 2 Diabetes Mellitus (T2DM) is one of these protein misfolding disorders (PMDs) and involves human islet amyloid polypeptide (hIAPP) misfolding and accumulating in parts of the body, primarily in the pancreas, causing damage to islet cells and affecting glucose regulation. In this review, we have summarised our current understanding of what causes hIAPP to misfold, what conformations are found in different parts of the body with a particular focus on what is known about the structure of hIAPP and how this links to T2DM. Understanding the molecular basis behind these misfolding events is essential for understanding the role of hIAPP to develop better therapeutics since type 2 diabetes currently affects over 4.9 million people in the United Kingdom alone and is predicted to increase as our population ages.
Collapse
|
42
|
Kreiser RP, Wright AK, Sasser LR, Rinauro DJ, Gabriel JM, Hsu CM, Hurtado JA, McKenzie TL, Errico S, Albright JA, Richardson L, Jaffett VA, Riegner DE, Nguyen LT, LeForte K, Zasloff M, Hollows JE, Chiti F, Vendruscolo M, Limbocker R. A Brain-Permeable Aminosterol Regulates Cell Membranes to Mitigate the Toxicity of Diverse Pore-Forming Agents. ACS Chem Neurosci 2022; 13:1219-1231. [PMID: 35404569 PMCID: PMC9026273 DOI: 10.1021/acschemneuro.1c00840] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
![]()
The molecular composition
of the plasma membrane plays a key role
in mediating the susceptibility of cells to perturbations induced
by toxic molecules. The pharmacological regulation of the properties
of the cell membrane has therefore the potential to enhance cellular
resilience to a wide variety of chemical and biological compounds.
In this study, we investigate the ability of claramine, a blood–brain
barrier permeable small molecule in the aminosterol class, to neutralize
the toxicity of acute biological threat agents, including melittin
from honeybee venom and α-hemolysin from Staphylococcus
aureus. Our results show that claramine neutralizes
the toxicity of these pore-forming agents by preventing their interactions
with cell membranes without perturbing their structures in a detectable
manner. We thus demonstrate that the exogenous administration of an
aminosterol can tune the properties of lipid membranes and protect
cells from diverse biotoxins, including not just misfolded protein
oligomers as previously shown but also biological protein-based toxins.
Our results indicate that the investigation of regulators of the physicochemical
properties of cell membranes offers novel opportunities to develop
countermeasures against an extensive set of cytotoxic effects associated
with cell membrane disruption.
Collapse
Affiliation(s)
- Ryan P. Kreiser
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Aidan K. Wright
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Liam R. Sasser
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Dillon J. Rinauro
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Justus M. Gabriel
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Claire M. Hsu
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Jorge A. Hurtado
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Tristan L. McKenzie
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Silvia Errico
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - J. Alex Albright
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Lance Richardson
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Victor A. Jaffett
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Dawn E. Riegner
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Lam T. Nguyen
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Kathleen LeForte
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Michael Zasloff
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, District of Columbia 20010, United States
| | - Jared E. Hollows
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| |
Collapse
|
43
|
Gao G, Liu X, Gu Z, Mu Q, Zhu G, Zhang T, Zhang C, Zhou L, Shen L, Sun T. Engineering Nanointerfaces of Au 25 Clusters for Chaperone-Mediated Peptide Amyloidosis. NANO LETTERS 2022; 22:2964-2970. [PMID: 35297644 DOI: 10.1021/acs.nanolett.2c00149] [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: 06/14/2023]
Abstract
Synthetic nanomaterials possessing biomolecular-chaperone functions are good candidates for modulating physicochemical interactions in many bioapplications. Despite extensive research, no general principle to engineer nanomaterial surfaces is available to precisely manipulate biomolecular conformations and behaviors, greatly limiting attempts to develop high-performance nanochaperone materials. Here, we demonstrate that, by quantifying the length (-SCxR±, x = 3-11) and charges (R- = -COO-, R+ = -NH3+) of ligands on Au25 gold nanochaperones (AuNCs), simulating binding sites and affinities of amyloid-like peptides with AuNCs, and probing peptide folding and fibrillation in the presence of AuNCs, it is possible to precisely manipulate the peptides' conformations and, thus, their amyloidosis via customizing AuNCs nanointerfaces. We show that intermediate-length liganded AuNCs with a specific charge chaperone peptides' native conformations and thus inhibit their fibrillation, while other types of AuNCs destabilize peptides and promote their fibrillation. We offer a microscopic molecular insight into peptide identity on AuNCs and provide a guideline in customizing nanochaperones via manipulating their nanointerfaces.
Collapse
Affiliation(s)
- Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Xinglin Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhenhua Gu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Qingxue Mu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Guowei Zhu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Ting Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Cheng Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Lin Zhou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
44
|
Tempra C, Scollo F, Pannuzzo M, Lolicato F, La Rosa C. A unifying framework for amyloid-mediated membrane damage: The lipid-chaperone hypothesis. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140767. [PMID: 35144022 DOI: 10.1016/j.bbapap.2022.140767] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Over the past thirty years, researchers have highlighted the role played by a class of proteins or polypeptides that forms pathogenic amyloid aggregates in vivo, including i) the amyloid Aβ peptide, which is known to form senile plaques in Alzheimer's disease; ii) α-synuclein, responsible for Lewy body formation in Parkinson's disease and iii) IAPP, which is the protein component of type 2 diabetes-associated islet amyloids. These proteins, known as intrinsically disordered proteins (IDPs), are present as highly dynamic conformational ensembles. IDPs can partially (mis) fold into (dys) functional conformations and accumulate as amyloid aggregates upon interaction with other cytosolic partners such as proteins or lipid membranes. In addition, an increasing number of reports link the toxicity of amyloid proteins to their harmful effects on membrane integrity. Still, the molecular mechanism underlying the amyloidogenic proteins transfer from the aqueous environment to the hydrocarbon core of the membrane is poorly understood. This review starts with a historical overview of the toxicity models of amyloidogenic proteins to contextualize the more recent lipid-chaperone hypothesis. Then, we report the early molecular-level events in the aggregation and ion-channel pore formation of Aβ, IAPP, and α-synuclein interacting with model membranes, emphasizing the complexity of these processes due to their different spatial-temporal resolutions. Next, we underline the need for a combined experimental and computational approach, focusing on the strengths and weaknesses of the most commonly used techniques. Finally, the last two chapters highlight the crucial role of lipid-protein complexes as molecular switches among ion-channel-like formation, detergent-like, and fibril formation mechanisms and their implication in fighting amyloidogenic diseases.
Collapse
Affiliation(s)
- Carmelo Tempra
- Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic
| | - Federica Scollo
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Fabio Lolicato
- Heidelberg University Biochemistry Center, Heidelberg, Germany; Department of Physics, University of Helsinki, Helsinki, Finland.
| | - Carmelo La Rosa
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Catania, Italy.
| |
Collapse
|
45
|
Molecular dissection of cellular response of pancreatic islet cells to Bisphenol-A (BPA): a comprehensive review. Biochem Pharmacol 2022; 201:115068. [DOI: 10.1016/j.bcp.2022.115068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
|
46
|
Al Adem K, Shanti A, Srivastava A, Homouz D, Thomas SA, Khair M, Stefanini C, Chan V, Kim TY, Lee S. Linking Alzheimer’s Disease and Type 2 Diabetes: Characterization and Inhibition of Cytotoxic Aβ and IAPP Hetero-Aggregates. Front Mol Biosci 2022; 9:842582. [PMID: 35372522 PMCID: PMC8968156 DOI: 10.3389/fmolb.2022.842582] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic self-aggregation of β-amyloid (Aβ) peptide and islet amyloid polypeptide (IAPP) is implicated in the pathogenesis of Alzheimer’s disease (AD) and Type 2 diabetes (T2D), respectively. Increasing evidence, particularly the co-deposition of Aβ and IAPP in both brain and pancreatic tissues, suggests that Aβ and IAPP cross-interaction may be responsible for a pathological link between AD and T2D. Here, we examined the nature of IAPP-Aβ40 co-aggregation and its inhibition by small molecules. In specific, we characterized the kinetic profiles, morphologies, secondary structures and toxicities of IAPP-Aβ40 hetero-assemblies and compared them to those formed by their homo-assemblies. We demonstrated that monomeric IAPP and Aβ40 form stable hetero-dimers and hetero-assemblies that further aggregate into β-sheet-rich hetero-aggregates that are toxic (cell viability <50%) to both PC-12 cells, a neuronal cell model, and RIN-m5F cells, a pancreatic cell model for β-cells. We then selected polyphenolic candidates to inhibit IAPP or Aβ40 self-aggregation and examined the inhibitory effect of the most potent candidate on IAPP-Aβ40 co-aggregation. We demonstrated that epigallocatechin gallate (EGCG) form inter-molecular hydrogen bonds with each of IAPP and Aβ40. We also showed that EGCG reduced hetero-aggregate formation and resulted in lower β-sheets content and higher unordered structures in IAPP-Aβ40-EGCG samples. Importantly, we showed that EGCG is highly effective in reducing the toxicity of IAPP-Aβ40 hetero-aggregates on both cell models, specifically at concentrations that are equivalent to or are 2.5-fold higher than the mixed peptide concentrations. To the best of our knowledge, this is the first study to report the inhibition of IAPP-Aβ40 co-aggregation by small molecules. We conclude that EGCG is a promising candidate to prevent co-aggregation and cytotoxicity of IAPP-Aβ40, which in turn, contribute to the pathological link between AD and T2D.
Collapse
Affiliation(s)
- Kenana Al Adem
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Aya Shanti
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Amit Srivastava
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dirar Homouz
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Physics, University of Houston, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Sneha Ann Thomas
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Tae-Yeon Kim
- Department of Civil Infrastructure and Environmental Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Sungmun Lee
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Khalifa University’s Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Sungmun Lee,
| |
Collapse
|
47
|
Disaggregation of Islet Amyloid Polypeptide Fibrils as a Potential Anti-Fibrillation Mechanism of Tetrapeptide TNGQ. Int J Mol Sci 2022; 23:ijms23041972. [PMID: 35216095 PMCID: PMC8876742 DOI: 10.3390/ijms23041972] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Islet amyloid polypeptide (IAPP) fibrillation has been commonly associated with the exacerbation of type 2 diabetes prognosis. Consequently, inhibition of IAPP fibrillation to minimize β-cell cytotoxicity is an important approach towards β-cell preservation and type 2 diabetes management. In this study, we identified three tetrapeptides, TNGQ, MANT, and YMSV, that inhibited IAPP fibrillation. Using thioflavin T (ThT) fluorescence assay, circular dichroism (CD) spectroscopy, dynamic light scattering (DLS), and molecular docking, we evaluated the potential anti-fibrillation mechanism of the tetrapeptides. ThT fluorescence kinetics and microscopy as well as transmission electron microscopy showed that TNGQ was the most effective inhibitor based on the absence of normal IAPP fibrillar morphology. CD spectroscopy showed that TNGQ maintained the α-helical conformation of monomeric IAPP, while DLS confirmed the presence of varying fibrillation species. Molecular docking showed that TNGQ and MANT interact with monomeric IAPP mainly by hydrogen bonding and electrostatic interaction, with TNGQ binding at IAPP surface compared to YMSV, which had the highest docking score, but interact mainly through hydrophobic interaction in IAPP core. The highly polar TNGQ was the most active and appeared to inhibit IAPP fibrillation by disaggregation of preformed IAPP fibrils. These findings indicate the potential of TNGQ in the development of peptide-based anti-fibrillation and antidiabetic nutraceuticals.
Collapse
|
48
|
Interaction of membrane vesicles with the Pseudomonas functional amyloid protein FapC facilitates amyloid formation. BBA ADVANCES 2022; 2:100055. [PMID: 37082589 PMCID: PMC10074931 DOI: 10.1016/j.bbadva.2022.100055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Functional amyloids (FA) are proteins which are evolutionarily optimized to form highly stable fibrillar structures that strengthen the bacterial biofilm matrix. FA such as CsgA (E. coli) and FapC (Pseudomonas) are secreted to the bacterial surface where they integrate into growing fibril structures projecting from the outer membrane. FA are exposed to membrane surfaces in this process, but it remains unclear how membranes can interact with FA and potentially affect the self-assembly. Here we report the effect of different vesicles (DOPG, DMPG, DOPS, DOPC and DMPC) on the kinetics and structural endpoints of FapC fibrillation using various biophysical techniques. Particularly anionic lipids such as DMPG trigger FapC fibrillation, and the protein's second repeat sequence (R2) appears to be important for this interaction. Vesicles formed from phospholipids extracted from three different Pseudomonas strains (Δfap, ΔFapC and pfap) induce FapC fibrillation by accelerating nucleation. The general aggregation inhibitor epigallocatechin gallate (EGCG) inhibits FapC fibrillation by blocking interactions between FapC and vesicles and redirecting FapC monomers to oligomer structures. Our work indicates that biological membranes can contribute significantly to the fibrillation of functional amyloid.
Collapse
|
49
|
Mondal T, Mandal B. Proteolytic functional amyloid digests pathogenic amyloid. J Mater Chem B 2022; 10:4216-4225. [DOI: 10.1039/d2tb00640e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although amyloids are a well-known pathological structure, functional amyloids are beneficial. Functional amyloids can be engineered to cultivate desired functionality that can destroy malicious amyloids. However, not much is known...
Collapse
|
50
|
Tang H, Li Y, Kakinen A, Andrikopoulos N, Sun Y, Kwak E, Davis TP, Ding F, Ke PC. Graphene quantum dots obstruct the membrane axis of Alzheimer's amyloid beta. Phys Chem Chem Phys 2021; 24:86-97. [PMID: 34878460 PMCID: PMC8771921 DOI: 10.1039/d1cp04246g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a primary form of dementia with debilitating consequences, but no effective cure is available. While the pathophysiology of AD remains multifactorial, the aggregation of amyloid beta (Aβ) mediated by the cell membrane is known to be the cause for the neurodegeneration associated with AD. Here we examined the effects of graphene quantum dots (GQDs) on the obstruction of the membrane axis of Aβ in its three representative forms of monomers (Aβ-m), oligomers (Aβ-o), and amyloid fibrils (Aβ-f). Specifically, we determined the membrane fluidity of neuroblastoma SH-SY5Y cells perturbed by the Aβ species, especially by the most toxic Aβ-o, and demonstrated their recovery by GQDs using confocal fluorescence microscopy. Our computational data through discrete molecular dynamics simulations further revealed energetically favorable association of the Aβ species with the GQDs in overcoming peptide-peptide aggregation. Overall, this study positively implicated GQDs as an effective agent in breaking down the membrane axis of Aβ, thereby circumventing adverse downstream events and offering a potential therapeutic solution for AD.
Collapse
Affiliation(s)
- Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Yuhuan Li
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yunxiang Sun
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China
| | - Eunbi Kwak
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Thomas P. Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia,The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| |
Collapse
|