1
|
Oldam J, Tchernyshyov I, Van Eyk J, Troncoso J, Glabe CG, Agnetti G. Thioflavin T in-gel staining for ex vivo analysis of cardiac amyloid. Front Mol Biosci 2025; 12:1505250. [PMID: 40433590 PMCID: PMC12106040 DOI: 10.3389/fmolb.2025.1505250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
There are limited options to quantify and characterize amyloid species from biological samples in a simple manner. Thioflavin T (ThT) has been used for decades to stain amyloid fibrils, but to our knowledge, we were the first to use it in-gel. Thioflavin T in-gel staining is convenient as it is fast, inexpensive, accessible to most laboratories, and compatible with other fluorescent stains and downstream analyses such as mass spectrometry (MS).
Collapse
Affiliation(s)
- Joseph Oldam
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Tchernyshyov
- Advanced Clinical Biosystems Institute, Cedars-Sinai Hospital, Beverly Hills, CA, United States
| | - Jennifer Van Eyk
- Advanced Clinical Biosystems Institute, Cedars-Sinai Hospital, Beverly Hills, CA, United States
| | - Juan Troncoso
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles G. Glabe
- University of California Irvine School of Biological Sciences, Irvine, CA, United States
| | - Giulio Agnetti
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Kell DB, Pretorius E, Zhao H. A Direct Relationship Between 'Blood Stasis' and Fibrinaloid Microclots in Chronic, Inflammatory, and Vascular Diseases, and Some Traditional Natural Products Approaches to Treatment. Pharmaceuticals (Basel) 2025; 18:712. [PMID: 40430532 PMCID: PMC12114700 DOI: 10.3390/ph18050712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
'Blood stasis' (syndrome) (BSS) is a fundamental concept in Traditional Chinese Medicine (TCM), where it is known as Xue Yu (). Similar concepts exist in Traditional Korean Medicine ('Eohyul') and in Japanese Kampo medicine (Oketsu). Blood stasis is considered to underpin a large variety of inflammatory diseases, though an exact equivalent in Western systems medicine is yet to be described. Some time ago we discovered that blood can clot into an anomalous amyloid form, creating what we have referred to as fibrinaloid microclots. These microclots occur in a great many chronic, inflammatory diseases are comparatively resistant to fibrinolysis, and thus have the ability to block microcapillaries and hence lower oxygen transfer to tissues, with multiple pathological consequences. We here develop the idea that it is precisely the fibrinaloid microclots that relate to, and are largely mechanistically responsible for, the traditional concept of blood stasis (a term also used by Virchow). First, the diseases known to be associated with microclots are all associated with blood stasis. Secondly, by blocking red blood cell transport, fibrinaloid microclots provide a simple mechanistic explanation for the physical slowing down ('stasis') of blood flow. Thirdly, Chinese herbal medicine formulae proposed to treat these diseases, especially Xue Fu Zhu Yu and its derivatives, are known mechanistically to be anticoagulatory and anti-inflammatory, consistent with the idea that they are actually helping to lower the levels of fibrinaloid microclots, plausibly in part by blocking catalysis of the polymerization of fibrinogen into an amyloid form. We rehearse some of the known actions of the constituent herbs of Xue Fu Zhu Yu and specific bioactive molecules that they contain. Consequently, such herbal formulations (and some of their components), which are comparatively little known to Western science and medicine, would seem to offer the opportunity to provide novel, safe, and useful treatments for chronic inflammatory diseases that display fibrinaloid microclots, including Myalgic Encephalopathy/Chronic Fatigue Syndrome, long COVID, and even ischemic stroke.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Søltofts Plads 200, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch Private Bag X1, Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Søltofts Plads 200, 2800 Kongens Lyngby, Denmark
| | - Huihui Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100026, China;
- Institute of Ethnic Medicine and Pharmacy, Beijing University of Chinese Medicine, Beijing 100026, China
| |
Collapse
|
3
|
Chisholm TS, Hunter CA. Ligands for Protein Fibrils of Amyloid-β, α-Synuclein, and Tau. Chem Rev 2025. [PMID: 40327808 DOI: 10.1021/acs.chemrev.4c00838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Amyloid fibrils are characteristic features of many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. The use of small molecule ligands that bind to amyloid fibrils underpins both fundamental research aiming to better understand the pathology of neurodegenerative disease, and clinical research aiming to develop diagnostic tools for these diseases. To date, a large number of amyloid-binding ligands have been reported in the literature, predominantly targeting protein fibrils composed of amyloid-β (Aβ), tau, and α-synuclein (αSyn) fibrils. Fibrils formed by a particular protein can adopt a range of possible morphologies, but protein fibrils formed in vivo possess disease-specific morphologies, highlighting the need for morphology-specific amyloid-binding ligands. This review details the morphologies of Aβ, tau, and αSyn fibril polymorphs that have been reported as a result of structural work and describes a database of amyloid-binding ligands containing 4,288 binding measurements for 2,404 unique compounds targeting Aβ, tau, or αSyn fibrils.
Collapse
Affiliation(s)
- Timothy S Chisholm
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Christopher A Hunter
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| |
Collapse
|
4
|
Hima S, Remya C, Abhithaj J, Arun KG, Sabu A, Sajitha K, Rajan R, Vasudevan DM, Dileep KV. Insights into the structural and biophysical mechanisms of benzamidine-driven inhibition of human lysozyme aggregation. Int J Biol Macromol 2025; 305:141139. [PMID: 39961574 DOI: 10.1016/j.ijbiomac.2025.141139] [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: 10/15/2024] [Revised: 01/29/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Amyloid fibril formation is a hallmark of several protein misfolding diseases, including systemic hereditary amyloidosis (SHA), in which lysozyme aggregates into plaques, causing inflammation to various tissues. SHA is a rare disease with no current drug treatment options. In our efforts to identify potential therapeutics for SHA, we investigated the inhibitory effects of benzamidine (BEN) on the human lysozyme fibrillation (HLF). Multiple biophysical assays demonstrated BEN's ability to prevent the lysozyme fibrillation. Intrinsic fluorescence measurements highlighted BEN's interaction with human native lysozyme (HNL). We inferred that the binding mode of BEN to HNL through ITC, molecular docking, and molecular dynamics simulations, confirmed BEN's binding at the active site, near stretch-2 (residues 52-64). The circular dichroism (CD) analysis revealed alterations in the secondary structure of HNL and HLF upon BEN's treatment. In HLF, an increase in BEN's concentration resulted in a concentration-dependent reduction in β-sheet content. SEM and TEM analyses revealed reduced fibril formation and significant alterations in fibril morphology in the presence of BEN. Importantly, BEN exhibited no cytotoxic effects in HEK-293 cells. These results provide strong evidence of BEN's anti- amyloidogenic activity and offer a foundation for future drug development targeting lysozyme amyloidosis.
Collapse
Affiliation(s)
- Sree Hima
- Department of Health Sciences Research, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India; Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - Chandran Remya
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - J Abhithaj
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr Janaki Ammal Campus Thalassery, Kannur University, Palayad 670661, India; Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - K G Arun
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr Janaki Ammal Campus Thalassery, Kannur University, Palayad 670661, India
| | - A Sabu
- Inter University Centre for Bioscience and Department of Biotechnology & Microbiology, Dr Janaki Ammal Campus Thalassery, Kannur University, Palayad 670661, India
| | - K Sajitha
- Department of Biochemistry, Amrita School of Dentistry, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala, India
| | - Rakhi Rajan
- Department of Chemistry and Biochemistry, Price Family Foundation Institute of Structural Biology, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - D M Vasudevan
- Department of Health Sciences Research, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India; Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - K V Dileep
- Department of Health Sciences Research, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India; Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India.
| |
Collapse
|
5
|
Zhang M, Kou J, Song Z, Qiu L, Yang C, Xue Q. A Janus Amyloid-like Nanofilm Inhibits Colorectal Cancer Postoperative Recurrence and Abdominal Adhesion via Synergistic Enzyme Cascade. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:670. [PMID: 40358287 PMCID: PMC12073602 DOI: 10.3390/nano15090670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Postoperative peritoneal adhesion and high recurrence rates are critical challenges in the clinical treatment of colorectal cancer. In this study, based on amyloid-like protein self-assembly technology, a novel Janus protein film was developed. The protein film encapsulates glucose oxidase (GOx) and catalase (CAT), which is named PTL@GC. Through a one-step method involving cysteine-reduced lysozyme-induced amyloid-like self-assembly, the film was co-loaded with GOx and CAT to achieve synergistic anti-adhesion and anti-tumor recurrence effects. The Janus film features a hydrophobic side that stably adheres to the intestinal surface without exogenous chemical modification and a hydrophilic side that prevents adhesion. The loaded GOx selectively induces disulfidptosis in SLC7A11-overexpressing tumor cells, while CAT degrades H2O2 to alleviate hypoxia and inhibit oxidative stress, significantly reducing adhesion-related fibrosis. The experimental results demonstrate that PTL@GC exhibited excellent mechanical properties, high enzyme activity retention (>90%), and controllable degradability (complete metabolism within 50 days). In animal models, PTL@GC reduced postoperative adhesion area by 22.77%, decreased local tumor burden to 28.42% of the control group, and achieved an inhibition rate of 58.49%, without inducing systemic toxicity. This study presents a biologically safe and functionally synergistic approach to addressing dual complications following colorectal cancer surgery, offering potential insights for future research on multifunctional Janus materials.
Collapse
Affiliation(s)
- Man Zhang
- Department of General Surgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| | - Junhao Kou
- College of Pharmacy, Xi’an Medical University, Xi’an 710021, China
| | - Zhenyi Song
- College of Pharmacy, Xi’an Medical University, Xi’an 710021, China
| | - Ling Qiu
- Department of General Surgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| | - Chunzhao Yang
- Department of General Surgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| | - Qi Xue
- Department of General Surgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| |
Collapse
|
6
|
Zhao L, Cao Y, Xin Y, Liu C, Yang J, Li Y, Tian S, Liu Z, Jia H, Liu M, Hu M, Luo L, Meng F. Targeted Raman Visualization and Mitigation of α-Synuclein Amyloidogenesis in Living Zebrafish by a Nanobody-Decorated Polydiacetylene. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411419. [PMID: 39996265 DOI: 10.1002/smll.202411419] [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: 11/28/2024] [Revised: 02/03/2025] [Indexed: 02/26/2025]
Abstract
α-Synuclein (α-Syn) amyloidogenesis is considered a promising diagnostic marker and therapeutic target for Parkinson's disease (PD). Simultaneously visualizing and mitigating α-Syn amyloidogenesis are essential for future PD theranostics, yet they continue to pose an insurmountable challenge. This study have herein developed a nanobody-decorated polydiacetylene to approach a straightforward solution. Grafting α-Syn61-95 segment into the third complementary determining region of a parent nanobody generates an engineered nanobody X30 that can bind with α-Syn and prevent its amyloidogenesis through homotypic interaction. It next use X30 to decorate poly(deca-4,6-diynedioic acid) (PDDA), a polydiacetylene with an ultrastrong alkyne Raman signal (2120 cm-1) in the cellular silent region, to create an α-Syn targeting Raman probe PX30. The binding affinity between X30 and α-Syn can be further boosted for over 150 times attributed to the rigidity of PDDA backbone and the multivalent effect. Therefore, PX30 not only enables real-time Raman visualization of α-Syn amyloidogenesis with a high signal-to-noise ratio in living zebrafish, but also alleviates amyloidogenesis-mediated damage to zebrafish embryos by effectively inhibiting α-Syn amyloidogenesis at low stoichiometric concentrations and scavenging pathologic reactive oxygen species.
Collapse
Affiliation(s)
- Liyuan Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yujuan Cao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yanru Xin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Chenxi Liu
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Jin Yang
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Yanan Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zhenxing Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Min Hu
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry, Materia Medica School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry, Materia Medica School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
7
|
Mahato OP, Prajapati KP, Ansari M, Mittal S, Mishra N, Anand BG, Kar K. Fabricating Silver Nanoparticles with Antidiabetic d-Pinitol to Restrict Amyloid Formation of Insulin. ACS APPLIED BIO MATERIALS 2025; 8:1934-1945. [PMID: 39928478 DOI: 10.1021/acsabm.4c01224] [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: 02/12/2025]
Abstract
In this study, we synthesized silver nanoparticles functionalized with antidiabetic d-pinitol molecules and investigated their anti-amyloid effect on Insulin aggregation. Our results show that these functionalized nanoparticles effectively inhibit both spontaneous and seed-induced amyloid aggregation of Insulin in a dose-dependent manner. The d-pinitol-functionalized silver nanoparticles interact directly with the aggregation-prone regions of Insulin's partially unfolded structures, as demonstrated by quenching and computational experiments. Analysis of docking and simulation data suggests that antiamyloid activity of d-pinitol functionalized AgNPs is due to a strong Insulin-nanoparticle interaction, facilitated via hydrogen bonds. These interactions disrupt the Insulin monomer-dimer equilibrium and prevent the formation of toxic amyloid structures. The results could lead to the development of d-pinitol-based Insulin-stabilizing nanoformulations with potential antidiabetic properties.
Collapse
Affiliation(s)
- Om Prakash Mahato
- Biophysical and Biomaterials Research Laboratory, Room 310, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Kailash Prasad Prajapati
- Biophysical and Biomaterials Research Laboratory, Room 310, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Masihuzzaman Ansari
- Biophysical and Biomaterials Research Laboratory, Room 310, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shikha Mittal
- Biophysical and Biomaterials Research Laboratory, Room 310, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nishant Mishra
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Bibin Gnanadhason Anand
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Karunakar Kar
- Biophysical and Biomaterials Research Laboratory, Room 310, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
8
|
Akhtar A, Singh P, Admane N, Grover A. Salvianolic acid B prevents the amyloid transformation of A53T mutant of α-synuclein. Biophys Chem 2025; 318:107379. [PMID: 39693815 DOI: 10.1016/j.bpc.2024.107379] [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: 08/10/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder involving the progressive loss of dopaminergic neurons in the substantia nigra pars compacta triggered by the accumulation of amyloid aggregates of α-synuclein protein. This study investigates the potential of Salvianolic Acid B (SalB), a water-soluble polyphenol derived from Salvia miltiorrhiza Bunge, in modulating the aggregation of the A53T mutant of α-synuclein (A53T Syn). This mutation is associated with rapid aggregation and a higher rate of protofibril formation in early-onset familial PD. Computational and experimental approaches demonstrated Sal-B effectively prevents the amyloid fibrillation of A53T Syn by interacting with the N-terminal region and NAC domain. Sal-B particularly associates with the KTKEGV motif and NACore segment of A53T Syn by hydrophobic and hydrogen bonding interactions. Replica exchange molecular dynamics (REMD) simulations indicated that Sal-B reduces intramolecular hydrogen bonding and structural transitions into β-sheet rich conformations, thereby lowering the aggregation propensity of A53T Syn. Systematic analysis conducted using biophysical techniques and high-end microscopy has demonstrated significant inhibition in the amyloid transformation of A53T Syn corroborated by a 92 % decrease in ThT maxima at 100 μM Sal-B concentration and microscopic techniques validated the absence of mature fibrillar amyloids. DLS data revealed heterogeneous particle sizes, supporting the formation of smaller unstructured aggregates. These findings underscore Sal-B as a promising therapeutic candidate for PD and related synucleinopathies, warranting further investigation in cellular and animal models to advance potential treatments and early intervention strategies.
Collapse
Affiliation(s)
- Almas Akhtar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Payal Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nikita Admane
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
9
|
Kuila S, Misra S, Saha R, Ghosh L, Singh P, Ghosh A, Reddy KP, Pandit S, Ganguly D, Datta P, Bindu S, Nandi G, Samai S, Nanda J. Fmoc-conjugated dipeptide-based hydrogels and their pH-tuneable behaviour. SOFT MATTER 2025; 21:1676-1685. [PMID: 39902563 DOI: 10.1039/d4sm01282h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
In this work, we designed three dipeptide-based hydrogelators by attaching different hydrophilic amino acids (aspartic acid, glutamic acid, and glutamine) to Fmoc-conjugated phenylalanine. Self-assembly and gelation of the three dipeptides were studied in 50 mM phosphate buffer solutions. The gelation efficiency and kinetics of glutamine-based hydrogelators (FQ) were better than those of aspartic acid and glutamic acid-based hydrogelators FD and FE respectively at neutral pH. The lower gelation efficiency of FE and FD was due to the pH-responsive side chain (carboxylic acid) compared to FQ, where amide group was present as a side chain. Three hydrogelators exhibited better gelation efficiency at lower pHs as the anionic carboxylate group was protonated to the carboxylic group, facilitating better self-assembly and gelation processes. Thioflavin-T (ThT) binding study of hydrogels indicated the formation of β-sheet-like structure in the hydrogel state. The self-assembly process was inspected using molecular dynamic study, revealing that the newly developed FQ gelator possesses a higher aggregation tendency than FE and FD. Finally, these peptide-based injectable biomaterials were examined using fluorescence and FT-IR spectroscopy, scanning electron microscopy, and rheology.
Collapse
Affiliation(s)
- Soumen Kuila
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri, West Bengal 734013, India.
| | - Souvik Misra
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri, West Bengal 734013, India.
| | - Riya Saha
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, 700091, India
| | - Laboni Ghosh
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri, West Bengal 734013, India.
| | - Pijush Singh
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri, West Bengal 734013, India.
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Anamika Ghosh
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, 700091, India
| | - Kolimi Prashanth Reddy
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata (NIPER-K), Chunilal Bhawan, 168, Maniktala Main Road, Kolkata, West Bengal 700054, India
| | - Subhradip Pandit
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Debabani Ganguly
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, 700091, India
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata (NIPER-K), Chunilal Bhawan, 168, Maniktala Main Road, Kolkata, West Bengal 700054, India
| | - Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Gouranga Nandi
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling 734013, India
| | - Suman Samai
- Department of Chemistry, Prabhat Kumar College Contai, Contai, Purba Medinipur, West Bengal, India
| | - Jayanta Nanda
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri, West Bengal 734013, India.
| |
Collapse
|
10
|
López S, Rojo-Domínguez A, López-Simeon R, Sosa-Peinado A, Nájera H. L-tyrosine inhibits the formation of amyloid fibers of human lysozyme at physiological pH and temperature. Amino Acids 2025; 57:15. [PMID: 39955695 PMCID: PMC11830642 DOI: 10.1007/s00726-025-03445-6] [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: 10/16/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
Amyloid fibers are implicated in numerous diseases, making their study crucial for identifying effective therapeutic compounds. This research highlights the ability of L-tyrosine to inhibit the formation of amyloid fibers in human lysozyme. At a 1:1 molar ratio under physiological conditions (pH 7.4, 37 °C), L-tyrosine significantly reduces amyloid fiber formation, as evidenced by a decrease in thioflavin T fluorescence. Differential scanning calorimetry (DSC) shows a major energy requirement for temperature denaturation when the lysozyme is in the presence of L-tyrosine. Additionally, chemical denaturation experiments reveal a shift in the intrinsic fluorescence spectrum of lysozyme in the presence of L-tyrosine, indicating a direct interaction. Computational docking studies with Molecular Operating Environment (MOE) further confirm that L-tyrosine binds effectively, exhibiting similar binding energies to those of the natural substrate. This study underscores L-tyrosine's potential as a strong inhibitor of amyloid fiber formation, demonstrating its stabilizing effect on lysozyme and its promise in therapeutic applications.
Collapse
Affiliation(s)
- Santos López
- Posgrado en Ciencias Naturales E Ingeniería, Universidad Autónoma Metropolitana-Cuajimalpa, Alcaldía Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, Ciudad de México, 05348, México
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Alcaldía Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, 05348, Ciudad de Mexico, Mexico
| | - Arturo Rojo-Domínguez
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Alcaldía Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, 05348, Ciudad de Mexico, Mexico
| | - Roxana López-Simeon
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Alcaldía Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, 05348, Ciudad de Mexico, Mexico
| | - Alejandro Sosa-Peinado
- Laboratorio de Biosensores y Modelaje Molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
| | - Hugo Nájera
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Alcaldía Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, 05348, Ciudad de Mexico, Mexico.
| |
Collapse
|
11
|
Jang J, Jo Y, Park CB. NIR Light-Triggered Structural Modulation of Self-Assembled Prion Protein Aggregates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405354. [PMID: 39757410 DOI: 10.1002/smll.202405354] [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: 06/29/2024] [Revised: 12/06/2024] [Indexed: 01/07/2025]
Abstract
The self-replication of misfolded prion protein (PrP) aggregates is the major pathological event of different prion diseases, affecting mammal brains by cross-species transmission. Here, the structural modulation of PrP aggregates are reported by activated carbon materials upon near-infrared (NIR) light irradiation. Activated carbon cobalt (ACC) nanosheets are synthesized using glycerol and metal salts to utilize the charge carriers released under NIR light exposure. According to the microscopy and spectroscopy analysis results, NIR light-excited ACC nanosheets successfully dissociate the β-sheet-rich and plaque-like PrP aggregates into denatured fragments by modifying their amino acid residues. The in vitro assay results demonstrate that ACC nanosheets possess biocompatibility to neuroblastoma cells and alleviating effect against the neurotoxicity of PrP aggregates. This work suggests the first potential photodynamic platform for the future treatment of prion diseases.
Collapse
Affiliation(s)
- Jinhyeong Jang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 335 Science Road, Daejeon, 34141, Republic of Korea
- Applied Science Research Institute, Korea Advanced Institute of Science and Technology (KAIST), 335 Science Road, Daejeon, 34141, Republic of Korea
| | - Yonghan Jo
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 335 Science Road, Daejeon, 34141, Republic of Korea
| | - Chan Beum Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 335 Science Road, Daejeon, 34141, Republic of Korea
| |
Collapse
|
12
|
Clarke JR, Bacelar TS, Fernandes GG, Silva RCD, Antonio LS, Queiroz M, de Souza RV, Valadão LF, Ribeiro GS, De Lima EV, Colodeti LC, Mangeth LC, Wiecikowski A, da Silva TN, Paula-Neto HA, da Costa R, Cordeiro Y, Passos GF, Figueiredo CP. Abatacept inhibits Th17 differentiation and mitigates α-synuclein-induced dopaminergic dysfunction in mice. Mol Psychiatry 2025; 30:547-555. [PMID: 39152331 DOI: 10.1038/s41380-024-02618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is a multifaceted disease characterized by degeneration of nigrostriatal dopaminergic neurons, which results in motor and non-motor dysfunctions. Accumulation of α-synuclein (αSYN) in Lewy bodies is a key pathological feature of PD. Although the exact cause of PD remains unknown, accumulating evidence suggests that brain infiltration of T cells plays a critical role in the pathogenesis of disease, contributing to neuroinflammation and dopaminergic neurodegeneration. Here, we used a mouse model of brain-infused aggregated αSYN, which recapitulates motor and non-motor dysfunctions seen in PD patients. We found that αSYN-induced motor dysfunction in mice is accompanied by an increased number of brain-residing Th17 (IL17+ CD4+) cells, but not CD8+ T cells. To evaluate whether the modulation of T cell response could rescue αSYN-induced damage, we chronically treated animals with abatacept (8 mg/kg, sc, 3x per week), a selective T-cell co-stimulation modulator. We found that abatacept treatment decreased Th1 (IFNƔ+ CD4+) and Th17 (IL17+ CD4+) cells in the brain, rescued motor function and prevented dopaminergic neuronal loss in αSYN-infused mice. These results highlight the significance of effector CD4+ T cells, especially Th17, in the progression of PD and introduce novel possibilities for repurposing immunomodulatory drugs used for arthritis as PD-modifying therapies.
Collapse
Affiliation(s)
- Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Thiago Sa Bacelar
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Raquel Costa da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia S Antonio
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana Queiroz
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Renata V de Souza
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia F Valadão
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel S Ribeiro
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emanuelle V De Lima
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Lilian C Colodeti
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana C Mangeth
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adalgisa Wiecikowski
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Talita N da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Heitor A Paula-Neto
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Robson da Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Yraima Cordeiro
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Giselle F Passos
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
13
|
Osten V, Oepen K, Schneider D. The C-terminal α-helix is crucial for the activity of the bacterial ABC transporter BmrA. J Biol Chem 2025; 301:108098. [PMID: 39706270 PMCID: PMC11774805 DOI: 10.1016/j.jbc.2024.108098] [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: 07/19/2024] [Revised: 11/15/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024] Open
Abstract
ABC transporters are membrane integral proteins that consist of a transmembrane domain and nucleotide-binding domain (NBD). Two monomers (half-transporters) of the Bacillus subtilis ABC transporter Bacillus multidrug-resistance ATP (BmrA) dimerize to build a functional full-transporter. As all ABC exporters, BmrA uses the free energy of ATP hydrolysis to transport substrate molecules across the cell membrane. For substrate transport, a BmrA dimer undergoes major conformational changes. ATP binding drives dimerization of the NBDs followed by the hydrolysis of the nucleotides. Conserved structural elements within the NBD and transmembrane domain are crucial for dimerization and the activity of BmrA. In the BmrA structure, an α-helix is present at the C-terminus, which can be subdivided in two smaller helices. As shown here, the very C-terminal helix (fragment) is not crucial for the BmrA activity. In fact, based on Cys-scanning mutagenesis, this region is highly flexible. In contrast, a BmrA variant lacking the entire C-terminal α-helix, showed no ATPase and transport activity. Via Ala-scanning, we identified residues in the N-terminal fragment of the helix that are crucial for the BmrA activity, most likely via establishing contacts to structural elements involved in ATP recognition, binding, and/or hydrolysis.
Collapse
Affiliation(s)
- Veronika Osten
- Department of Chemistry - Biochemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Kristin Oepen
- Department of Chemistry - Biochemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Dirk Schneider
- Department of Chemistry - Biochemistry, Johannes Gutenberg-University, Mainz, Germany; Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany.
| |
Collapse
|
14
|
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
|
15
|
Paulrasu K, Caspa Gokulan R, El-Rifai W, Chen Z, Que J, Wang TC, Boutaud OG, Briegel K, Dikalov SI, Garcia-Buitrago MT, Zaika AI. Chronic Gastroesophageal Reflux Dysregulates Proteostasis in Esophageal Epithelial Cells. Cell Mol Gastroenterol Hepatol 2024; 19:101434. [PMID: 39637942 PMCID: PMC11786911 DOI: 10.1016/j.jcmgh.2024.101434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND & AIMS Gastroesophageal reflux disease (GERD) is a common digestive disorder that is characterized by esophageal tissue damage produced by exposure of the esophageal lining to the gastric refluxate. GERD can raise the risk of multiple serious complications including esophageal tumors. At the molecular levels, GERD-affected tissues are characterized by strong oxidative stress and the formation of reactive isolevuglandins (isoLGs). These products of lipid peroxidation rapidly interact with cellular proteins forming protein adducts. Here, we investigated the interrelationship between isoLG adduction and aggregation of cellular proteins. METHODS Protein misfolding and aggregation were analyzed using multiple protein misfolding and aggregation assays. Pathologic consequences of protein adduction and aggregation were studied using human and murine esophageal tissues. Surgical model of esophageal reflux injury and L2-IL1β transgenic mice were used to investigate the mechanisms of protein misfolding and aggregation. RESULTS Our studies demonstrate that gastroesophageal reflux causes protein misfolding and aggregation that is associated with severity of GERD. Dysregulation of proteostasis induces ferroptotic cell death and is mediated by modification of cellular proteins with reactive isoLGs that can be prevented by isoLG scavengers. CONCLUSIONS GERD causes dysregulation of cellular proteostasis, accumulation of isoLG protein adducts, misfolded, and aggregated proteins that promote ferroptotic cell death. Taken together, this study suggests that GERD has similarities to other known pathologic conditions that are characterized by protein misfolding and aggregation.
Collapse
Affiliation(s)
| | - Ravindran Caspa Gokulan
- Department of Surgery, University of Miami, Miami, Florida; Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, Florida
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami, Miami, Florida
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Timothy C Wang
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Olivier G Boutaud
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | | | - Sergey I Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Alexander I Zaika
- Department of Surgery, University of Miami, Miami, Florida; Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida; Department of Veterans Affairs, Miami VA Healthcare System, Miami, Florida.
| |
Collapse
|
16
|
Zargan S, Jalili H, Dabirmanesh B, Mesdaghinia S, Khajeh K. Amyloidogenesis of SARS-CoV-2 delta plus and omicron variants receptor-binding domain (RBD): impact of SUMO fusion tag. Biotechnol Lett 2024; 46:1037-1048. [PMID: 39182215 DOI: 10.1007/s10529-024-03525-9] [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: 04/13/2024] [Revised: 07/16/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
PURPOSE The RBD of SARS-CoV-2 mediates viral entry into host cells by binding to the host receptor ACE2. SARS-CoV-2 infection is linked to various health issues resembling amyloid-related problems, persuading us to investigate the amyloidogenicity of the SARS-CoV-2 spike RBD. METHODS The FoldAmyloid program was used to assess the amyloidogenic propensities in the RBD of Delta Plus and RBD of the Omicron variant, with and without the SUMO tag. After the expression of RBDs, purification, and dialysis steps were performed, subsequently the ThT assay, FTIR, and TEM were employed to check the RBD ability to form fibrils. RESULTS The ThT assay, TEM, and FTIR revealed the ability of RBD to self-assemble into β-sheet-rich aggregates (48.4% β-sheet content). Additionally, the presence of the SUMO tag reduced the formation of RBD amyloid-like fibrils. The amyloidogenic potential of Omicron RBD was higher than Delta Plus, according to both in silico and experimental analyses. CONCLUSIONS The SARS-CoV-2 RBD can assemble itself by forming aggregates containing amyloid-like fibrils and the presence of a SUMO tag can significantly decrease the formation of RBD amyloid-like fibrils. In silico analysis suggested that variation in the ThT fluorescence intensity of amyloid accumulations in the two SARS-CoV-2 strains arises from specific mutations in their RBD regions.
Collapse
Affiliation(s)
- Sadegh Zargan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hasan Jalili
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saba Mesdaghinia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
17
|
Guo Y, Xia C, Cao Y, Su J, Chi W, Chen D, Yan J. Modulation of intramolecular freedom for tuning fluorescence imaging and photooxidation of amyloid-β aggregates. MATERIALS HORIZONS 2024; 11:6040-6048. [PMID: 39295492 DOI: 10.1039/d4mh00660g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Alzheimer's disease (AD) is distinguished by amyloid-β (Aβ) deposition and plaque formation, prompting significant interest in fluorescence imaging and photooxidation of Aβ aggregates for diagnostic and intervention purposes. However, the molecular engineering required to modulate fluorescence imaging and photooxidation of Aβ presents notable challenges. Here, we present the design of four small molecules (BTD-SZ, BTD-YD, BTD-TA-SZ, and BTD-TA-YD) aimed at investigating the influence of intramolecular freedom of movement on imaging and photooxidation. Notably, BTD-SZ exhibits exceptional fluorescence properties, offering promising potential for non-invasive detection of Aβ plaques in vivo. Furthermore, by converting dimethylamine into triphenylamine to restrict intramolecular freedom of movement in the aggregate state, we synthesized a photosensitizer denoted as BTD-TA-SZ. This compound demonstrates aggregation-induced photooxidation (AIP), effectively impeding Aβ aggregation under light irradiation in vivo. Thus, the modulation of intramolecular freedom of movement emerges as a pivotal molecular engineering strategy for developing photosensitizers for the diagnosis and intervention of AD, offering insights into innovative approaches for combating this debilitating condition.
Collapse
Affiliation(s)
- Yuhui Guo
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Chunli Xia
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519040, P. R. China
| | - Yingmei Cao
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Junyi Su
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519040, P. R. China
| | - Weijie Chi
- Collaborative Innovation Center of One Health, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China
| | - Daoyuan Chen
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519040, P. R. China
| | - Jinwu Yan
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
18
|
Kobashigawa E, Muhsin SA, Abdullah A, Allen K, Sinnott EA, Zhang MZ, Russell S, Almasri M, Zhang S. Comparative study of immunoassays, a microelectromechanical systems-based biosensor, and RT-QuIC for the diagnosis of chronic wasting disease in white-tailed deer. BMC Vet Res 2024; 20:518. [PMID: 39551756 PMCID: PMC11571681 DOI: 10.1186/s12917-024-04351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Chronic wasting disease (CWD) is a fatal transmissible spongiform encephalopathy in cervids. The disease is caused by a pathogenic prion, namely PrPSc. Currently, diagnosis of CWD relies on IHC detection of PrPSc in the obex or retropharyngeal lymph nodes (RPLN) or ELISA screening of obex and RPLN followed by IHC confirmation of positive results. In this study, we assessed the performance characteristics of two immunoassays: CWD Ag-ELISA and TeSeE ELISA, RT-QuIC, and MEMS biosensor via testing 30 CWD + and 30 CWD- white-tailed deer RPLN samples. RESULTS Both CWD Ag-ELISA and TeSeE ELISA correctly identified all CWD + and CWD- samples. A greater intra-assay coefficient of variation (CV) in S/P ratios was observed for the TeSeE ELISA (16.52%), compared to CWD Ag-ELISA (9.49%). However, the high CV did not affect the qualitative results of triplicate assays when the corresponding manufacturer's cutoff was used. The MEMS biosensor not only correctly identified all CWD + and CWD- RPLN samples, but also demonstrated a 100% detection rate for all CWD + samples at dilutions from 10- 0 to 10- 3. Evaluation of RT-QuIC indicated that the rate of false negative reactions decreased from 21.98% at 10- 2 dilution to 0% at 10- 4 and 10- 5 dilutions; and the rate of false positive reactions reduced from 56.42% at 10- 2 dilution to 8.89% and 2.22% at 10- 4 and 10- 5 dilutions, respectively. Based on a stringent threshold of 2 x the first 10 fluorescent readings of each well and a final cutoff of 2/3 positive reactions for each sample, RT-QuIC correctly identified all positive and negative samples at 10- 4 and 10- 5 dilutions. Both MEMS biosensor and RT-QuIC achieved 100% sensitivity and 100% specificity under the experimental conditions described in this study. CONCLUSIONS The two immunoassays (CWD Ag-ELISA and TeSeE ELISA) performed comparably on white-tailed deer RPLN samples. MEMS biosensor is a reliable portable tool for CWD diagnosis and RT-QuIC can be used for routine testing of CWD if appropriate testing parameters and interpretive criteria are applied.
Collapse
Affiliation(s)
- Estela Kobashigawa
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA
| | - Sura A Muhsin
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Amjed Abdullah
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Keara Allen
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA
| | - Emily A Sinnott
- Missouri Department of Conservation, 2901 W Truman Blvd, Jefferson City, MO, USA
| | - Michael Z Zhang
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA
| | - Sherri Russell
- Missouri Department of Conservation, 2901 W Truman Blvd, Jefferson City, MO, USA
| | - Mahmoud Almasri
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Shuping Zhang
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA.
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, 901 E. Campus Loop, Columbia, MO, USA.
| |
Collapse
|
19
|
Williams FN, Travis KL, Haver HN, Umano AD, Guerra-Hernandez Y, Scaglione KM. Acute stress and multicellular development alter the solubility of the Dictyostelium Sup35 ortholog ERF3. Microbiol Spectr 2024; 12:e0160724. [PMID: 39345220 PMCID: PMC11537047 DOI: 10.1128/spectrum.01607-24] [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: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Among sequenced organisms, the genome of Dictyostelium discoideum is unique in that it encodes for a massive amount of repeat-rich sequences in the coding region of genes. This results in the Dictyostelium proteome encoding for thousands of repeat-rich proteins, with nearly 24% of the Dictyostelium proteome encoding Q/N-rich regions that are predicted to be prion like in nature. To begin investigating the role of prion-like proteins in Dictyostelium, we decided to investigate ERF3, the Dictyostelium ortholog of the well-characterized yeast prion protein Sup35. ERF3 lacks the Q/N-rich region required for prion formation in yeast, raising the question of whether this protein aggregates and has prion-like properties in Dictyostelium. Here, we found that ERF3 formed aggregates in response to acute cellular stress. However, unlike bona fide prions, we were unable to detect transmission of aggregates to progeny. We further found that aggregation of this protein is driven by the ordered C-terminal domain independently of the disordered N-terminal domain. Finally, we also observed aggregation of ERF3 under conditions that induce multicellular development, suggesting that this phenomenon may play a role in Dictyostelium development. Together, these findings suggest a role for regulated protein aggregation in Dictyostelium cells under stress and during development.IMPORTANCEPrion-like proteins have both beneficial and deleterious effects on cellular health, and many organisms have evolved distinct mechanisms to regulate the behaviors of these proteins. The social amoeba Dictyostelium discoideum contains the highest proportion of proteins predicted to be prion like and has mechanisms to suppress their aggregation. However, the potential roles and regulation of these proteins remain largely unknown. Here, we demonstrate that aggregation of the Dictyostelium translation termination factor ERF3 is induced by both acute cellular stress and by multicellular development. These findings imply that protein aggregation may have a regulated and functional role in the Dictyostelium stress response and during multicellular development.
Collapse
Affiliation(s)
- Felicia N. Williams
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Kanesha L. Travis
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Holly N. Haver
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Anna D. Umano
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Yaneli Guerra-Hernandez
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - K. Matthew Scaglione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
- Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, North Carolina, USA
| |
Collapse
|
20
|
P E, Jeevanandam J, Kumar B, N AM, N T S. 2-Methoxy-4-formylphenol suppresses methylglyoxal glycation mediated structural alterations and esterase activity of hemoglobin - A multi spectroscopic, biophysical and in-silico study. Int J Biol Macromol 2024; 282:137128. [PMID: 39486728 DOI: 10.1016/j.ijbiomac.2024.137128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Glycation is the non-enzymatic reaction of glucose or its metabolites to proteins, causing irreversible changes. Methylglyoxal, a dicarbonyl, affects the structure and function of physiologically important proteins. Being a major circulatory protein, hemoglobin is highly prone to glycation. Current research focuses on identifying potent glycation inhibitors to prevent glycation and their impact on protein structure and function. The present study investigates the Advanced Glycation Endproducts (AGEs) inhibitory effects of 2-methoxy-4-formylphenol (Vanillin) against methylglyoxal mediated glycation of hemoglobin. The hemoglobin-vanillin glycation model exhibited inhibition of AGE formation, amyloid fibrils, aggregates and reduction in esterase activity. The fluorescence spectroscopic technique revealed efficient binding of vanillin and hemoglobin, with Stern Volmer plot indicating the presence of static quenching. The conformational stability of the vanillin and hemoglobin interaction was also evident from the molecular docking and dynamics studies. The proximal orientation of residues (H2 and K82 associated in esterase activity) of hemoglobin β1 chain and vanillin, supports the noted effect of reduced esterase activity in the presence of vanillin in glycated hemoglobin and the inhibition of the overall formation of AGE of hemoglobin in the presence of vanillin.
Collapse
Affiliation(s)
- Esackimuthu P
- Molecular Biophysics Lab, School of Chemical and Biotechnology, SASTRA Deemed To Be University, Thanjavur 613401, TamilNadu, India
| | - Jayanth Jeevanandam
- Molecular Biophysics Lab, School of Chemical and Biotechnology, SASTRA Deemed To Be University, Thanjavur 613401, TamilNadu, India
| | - Bhuvana Kumar
- Molecular Biophysics Lab, School of Chemical and Biotechnology, SASTRA Deemed To Be University, Thanjavur 613401, TamilNadu, India
| | - Arul Murugan N
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi 110020, India
| | - Saraswathi N T
- Molecular Biophysics Lab, School of Chemical and Biotechnology, SASTRA Deemed To Be University, Thanjavur 613401, TamilNadu, India.
| |
Collapse
|
21
|
Malta SM, Rodrigues TS, Silva MH, Marquez AS, Ferreira RB, do Prado Mascarenhas FNA, Zanon RG, Bernardes LMM, Batista LL, da Silva MNT, de Oliveira Santos D, Santos ACC, Mendes-Silva AP, Spindola FS, Ueira-Vieira C. Brazilian kefir fraction mitigates the Alzheimer-like phenotype in Drosophila melanogaster with β-amyloid overexpression model. Sci Rep 2024; 14:25474. [PMID: 39461991 PMCID: PMC11513133 DOI: 10.1038/s41598-024-76601-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition and the primary form of dementia among elderly people. The amyloidogenic hypothesis is the main theory that explains this phenomenon and describes the extracellular accumulation of amyloid beta (Aβ) peptides. Model organisms such as Drosophila melanogaster have been utilized to improve the understanding of this disease and its treatment. This study evaluated the effects of peptide and metabolic fractions of Brazilian kefir on a strain of D. melanogaster that expresses human Aβ peptide 1-42 in the eye. The parameters assessed included ommatidial organization, vacuole area, retinal thickness, and Aβ peptide quantification. The present study revealed that the fractions, particularly the peptidic fraction, significantly reduced the vacuole area and increased the retina thickness in treated flies, indicating an improvement in neurodegeneration phenotype. The peptidic fraction was also found to alter Aβ aggregation dynamics, inhibiting Aβ fibril formation, as revealed by dynamic light scattering. This study demonstrated that kefir fractions, particularly the peptidic fraction < 10 kDa, have the potential to regulate Aβ aggregation and alleviate neurodegeneration in a Drosophila melanogaster AD-like model. These findings suggest that kefir fractions could be viable for the bioprospection of novel drug prototypes for AD treatment, providing valuable insights into strategies targeting Aβ aggregation and neurodegeneration in AD.
Collapse
Affiliation(s)
- Serena Mares Malta
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil.
| | - Tamiris Sabrina Rodrigues
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | - Matheus Henrique Silva
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | - Alexandre Souza Marquez
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | - Rafael Bernardes Ferreira
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | | | - Renata Graciele Zanon
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Lucas Matos Martins Bernardes
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | - Letícia Leandro Batista
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | - Ana Carolina Costa Santos
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | | | - Foued Salmen Spindola
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil
| | - Carlos Ueira-Vieira
- Laboratory of Genetics, Institute of Biotechnology, Federal University of Uberlândia, Acre Street, 2E building, room 230, Uberlândia, MG, 38405-319, Brazil.
| |
Collapse
|
22
|
Wang MD, Yi L, Li Y, Xu R, Hu J, Hou DY, Liu C, Wang H. Homologous Peptide Foldamer Promotes FUS Aggregation and Triggers Cancer Cell Death. J Am Chem Soc 2024; 146:28669-28676. [PMID: 39403745 DOI: 10.1021/jacs.4c03420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Fused in sarcoma (FUS), a multifunctional deoxyribonucleic acid (DNA)/ribonucleic acid (RNA)-binding protein, has been implicated in various cancer types, including sarcoma and leukemia. Despite its association with these diseases, there has been limited exploration of FUS as a cancer therapy target, primarily because its dynamic nature makes it difficult to target specifically. In this study, we explored a kind of β-sheet peptide foldamer, named β4-TAT, to influence FUS aggregation by targeting its RNA recognition motifs (RRM). This approach leverages the noncovalent interaction characteristics of peptide self-assembly processes. The β4 sequence, derived from the FUS RRM β-sheet, in combination with TAT, a peptide known for its nuclear targeting capability, enables β4-TAT to bind specifically to the analogous β4 sequence within FUS. Notably, β4-TAT effectively induces FUS aggregation within cells, leading to the death of cancer cells. Our work developed a novel peptide foldamer-based strategy for inducing protein aggregation, paving the way for innovative therapeutic approaches in targeting FUS-associated cancers.
Collapse
Affiliation(s)
- Man-Di Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| | - Li Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| | - Yanying Li
- Department of Medical Cell Biology Science for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Ruiwen Xu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Jiaojiao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| |
Collapse
|
23
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
24
|
Visser BS, Lipiński WP, Spruijt E. The role of biomolecular condensates in protein aggregation. Nat Rev Chem 2024; 8:686-700. [PMID: 39134696 DOI: 10.1038/s41570-024-00635-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 09/11/2024]
Abstract
There is an increasing amount of evidence that biomolecular condensates are linked to neurodegenerative diseases associated with protein aggregation, such as Alzheimer's disease and amyotrophic lateral sclerosis, although the mechanisms underlying this link remain elusive. In this Review, we summarize the possible connections between condensates and protein aggregation. We consider both liquid-to-solid transitions of phase-separated proteins and the partitioning of proteins into host condensates. We distinguish five key factors by which the physical and chemical environment of a condensate can influence protein aggregation, and we discuss their relevance in studies of protein aggregation in the presence of biomolecular condensates: increasing the local concentration of proteins, providing a distinct chemical microenvironment, introducing an interface wherein proteins can localize, changing the energy landscape of aggregation pathways, and the presence of chaperones in condensates. Analysing the role of biomolecular condensates in protein aggregation may be essential for a full understanding of amyloid formation and offers a new perspective that can help in developing new therapeutic strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Brent S Visser
- Institute of Molecules and Materials (IMM), Radboud University, Nijmegen, The Netherlands
| | - Wojciech P Lipiński
- Institute of Molecules and Materials (IMM), Radboud University, Nijmegen, The Netherlands
| | - Evan Spruijt
- Institute of Molecules and Materials (IMM), Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
25
|
Silva-Reis SC, Costa VM, da Silva DC, Pereira DM, Correia XC, García-Mera X, Rodríguez-Borges JE, Sampaio-Dias IE. Discovery of hybrid Glypromate conjugates with neuroprotective activity against paraquat-induced toxicity. RSC Med Chem 2024:d4md00584h. [PMID: 39290380 PMCID: PMC11403921 DOI: 10.1039/d4md00584h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
Neurodegenerative disorders comprise a series of heterogeneous conditions that affect millions of people worldwide, representing a significant health burden in both developed and developing countries. Without disease-modifying treatments currently available, the development of effective neurotherapeutics is a health priority. In this work, a new series of peptide-conjugates of the Glypromate neuropeptide is reported to determine the interplay of annular constriction and neuroprotective activity. To this end, (1R,3S,4S)-2-azanorbornane-3-carboxylic acid was used as an l-proline and l-pipecolic acid surrogate in addition to functionalization of the glutamate residue with relevant active pharmaceutical ingredients (APIs), namely amantadine, memantine, and (R)-1-aminoindane. Using non-differentiated SH-SY5Y cells, conjugates 14a and 15a, functionalized with amantadine, significantly reduced protein aggregation, with 15a outperforming both Glypromate (2-fold enhancement, p < 0.05) and an equimolar mixture of Glypromate and amantadine (p < 0.0001). On the other hand, in SH-SY5Y differentiated cells, conjugate 18c functionalized with (R)-1-aminoindane counteracted the toxicity elicited by paraquat (p < 0.0001), while Glypromate was found to exacerbate the neurotoxicity. Altogether, this work adds new insights into Glypromate research by demonstrating that chemical conjugation and annular constriction are effective strategies to tune neuroprotective responses against different neurotoxic stimuli, paving the way for the development of new neurotherapeutics.
Collapse
Affiliation(s)
- Sara C Silva-Reis
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Vera M Costa
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Daniela Correia da Silva
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - David M Pereira
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Xavier Cruz Correia
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| | - Xerardo García-Mera
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela E-15782 Santiago de Compostela Spain
| | - José E Rodríguez-Borges
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| | - Ivo E Sampaio-Dias
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| |
Collapse
|
26
|
Hazra R, Bera N, Layek S, Sarkar N. Efficiency of Encapsulation of Thioflavin T (ThT) into Polar and Nonpolar Environments of Different Bile Salt Aggregates: A Femtosecond Fluorescence Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:16272-16282. [PMID: 39042864 DOI: 10.1021/acs.langmuir.4c01460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The binding of Thioflavin T (ThT) with various bile salts, a potential host molecule, has been analyzed by steady-state and time-resolved fluorescence spectroscopy. A comparative study has been executed to investigate the influence of confinement of different bile salts, namely, sodium cholate (NaCh), sodium taurocholate (NaTC), and sodium deoxycholate (NaDC) on binding and excited state torsional motion of ThT molecules. The changes in absorption and emission properties of probe molecules were found to be sensitive to increasing bile salt concentration in aqueous 0.2 (M) NaCl solutions. The photophysics of ThT mainly depends on hydrophobicity, morphology, and size of bile salt aggregates in solution. In the presence of bile salts, the emission intensity and emission lifetime of ThT increase significantly, indicating encapsulation of dye. Moreover, we have also investigated the effect of the ionic strength of the medium by sodium chloride (NaCl) on the spectroscopic properties of ThT in the restricted surroundings of aqueous bile salts. It is observed that the fluorescence lifetime of ThT in bile salts increases significantly in the presence of NaCl. The encapsulation efficiency of ThT in bile salt aggregates has been assessed by iodide (I-) as an external ionic quencher. We found that NaDC aggregates are more efficient in the modulation of photophysical properties of ThT and also provide better protection efficiency to decrease the nonradiative deactivation processes.
Collapse
Affiliation(s)
- Ritwik Hazra
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, West Bengal, India
| | - Nanigopal Bera
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, West Bengal, India
| | - Souvik Layek
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, West Bengal, India
| | - Nilmoni Sarkar
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, West Bengal, India
| |
Collapse
|
27
|
Sammani PKT, Yospanya W, Niwa T, Kohata A, Taguchi H, Kinbara K. Monitoring insulin fibrillation kinetics using chromatographic analysis. Int J Biol Macromol 2024; 275:133660. [PMID: 38969030 DOI: 10.1016/j.ijbiomac.2024.133660] [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: 05/02/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Insulin is a small protein widely used to treat patients with diabetes and is a commonly used model for protein fibrillation studies. Under specific conditions, such as low pH and high temperature, insulin monomers aggregate to form fibrils. This aggregation is problematic for manufacturing and storage of insulin. The thioflavin T (ThT) assay is commonly used to study amyloid fibrillation but suffers from several limitations, such as the effect of protein concentration, the size of the amyloid fibrillar bundles, competitive binding, and fibril aggregation, all of which hinder precise quantitative analysis. Here, we present a method for studying the kinetics of insulin fibrillation utilizing ultra-performance liquid chromatography (UPLC). This method enables the quantitative detection of soluble insulin components, including chemically modified components. The formation of a deamidated species could be monitored at the early stage of fibrillation, and this species was likely included in the fibrils. In addition, in the presence of inhibitors known to compete with ThT for binding to fibrils, UPLC analysis showed the disappearance of soluble components even though the ThT assay did not indicate the presence of fibrils. These results suggest that the UPLC-based analysis presented here can complement the ThT assay for investigating the kinetics of protein fibrillation.
Collapse
Affiliation(s)
| | - Wijak Yospanya
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| | - Tatsuya Niwa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Ai Kohata
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Hideki Taguchi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Kazushi Kinbara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan; Research Center for Autonomous Systems Materialogy (ASMat), Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| |
Collapse
|
28
|
Martínez-Camarena Á, Bellia F, Paz Clares M, Vecchio G, Nicolas J, García-España E. Polymeric Nanozyme with SOD Activity Capable of Inhibiting Self- and Metal-Induced α-Synuclein Aggregation. Chemistry 2024; 30:e202401331. [PMID: 38687026 DOI: 10.1002/chem.202401331] [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: 04/03/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Despite decades of research, Parkinson's disease is still an idiopathic pathology for which no cure has yet been found. This is partly explained by the multifactorial character of most neurodegenerative syndromes, whose generation involves multiple pathogenic factors. In Parkinson's disease, two of the most important ones are the aggregation of α-synuclein and oxidative stress. In this work, we address both issues by synthesizing a multifunctional nanozyme based on grafting a pyridinophane ligand that can strongly coordinate CuII, onto biodegradable PEGylated polyester nanoparticles. The resulting nanozyme exhibits remarkable superoxide dismutase activity together with the ability to inhibit the self-induced aggregation of α-synuclein into amyloid-type fibrils. Furthermore, the combination of the chelator and the polymer produces a cooperative effect whereby the resulting nanozyme can also halve CuII-induced α-synuclein aggregation.
Collapse
Affiliation(s)
- Álvaro Martínez-Camarena
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
- Institut Galien Paris-Saclay, CNRS, Université Paris-Saclay, Orsay, 91400, France
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, Catania, 95125, Italy
- MatMoPol Research Group, Departamento de Química Inorgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Avda. Complutense s/n, Madrid, 28040, Spain
| | - Francesco Bellia
- Istituto di Cristallografia, CNR, P. Gaifami 18, Catania, 95126, Italy
| | - M Paz Clares
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, Catania, 95125, Italy
| | - Julien Nicolas
- Institut Galien Paris-Saclay, CNRS, Université Paris-Saclay, Orsay, 91400, France
| | - Enrique García-España
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
| |
Collapse
|
29
|
Li JY, Zhou CM, Jin RL, Song JH, Yang KC, Li SL, Tan BH, Li YC. The detection methods currently available for protein aggregation in neurological diseases. J Chem Neuroanat 2024; 138:102420. [PMID: 38626816 DOI: 10.1016/j.jchemneu.2024.102420] [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: 12/31/2023] [Revised: 03/30/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
Protein aggregation is a pathological feature in various neurodegenerative diseases and is thought to play a crucial role in the onset and progression of neurological disorders. This pathological phenomenon has attracted increasing attention from researchers, but the underlying mechanism has not been fully elucidated yet. Researchers are increasingly interested in identifying chemicals or methods that can effectively detect protein aggregation or maintain protein stability to prevent aggregation formation. To date, several methods are available for detecting protein aggregates, including fluorescence correlation spectroscopy, electron microscopy, and molecular detection methods. Unfortunately, there is still a lack of methods to observe protein aggregation in situ under a microscope. This article reviews the two main aspects of protein aggregation: the mechanisms and detection methods of protein aggregation. The aim is to provide clues for the development of new methods to study this pathological phenomenon.
Collapse
Affiliation(s)
- Jing-Yi Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Cheng-Mei Zhou
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Rui-Lin Jin
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Jia-Hui Song
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, PR China
| | - Ke-Chao Yang
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Shu-Lei Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Bai-Hong Tan
- Laboratory Teaching Center of Basic Medicine, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China
| | - Yan-Chao Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Changchun city, Jilin Province 130021, PR China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
30
|
Bonnecaze L, Jumel K, Vial A, Khemtemourian L, Feuillie C, Molinari M, Lecomte S, Mathelié-Guinlet M. N-Formylation modifies membrane damage associated with PSMα3 interfacial fibrillation. NANOSCALE HORIZONS 2024; 9:1175-1189. [PMID: 38689531 DOI: 10.1039/d4nh00088a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The virulence of Staphylococcus aureus, a multi-drug resistant pathogen, notably depends on the expression of the phenol soluble modulins α3 (PSMα3) peptides, able to self-assemble into amyloid-like cross-α fibrils. Despite remarkable advances evidencing the crucial, yet insufficient, role of fibrils in PSMα3 cytotoxic activities towards host cells, the relationship between its molecular structures, assembly propensities, and modes of action remains an open intriguing problem. In this study, combining atomic force microscopy (AFM) imaging and infrared spectroscopy, we first demonstrated in vitro that the charge provided by the N-terminal capping of PSMα3 alters its interactions with model membranes of controlled lipid composition without compromising its fibrillation kinetics or morphology. N-formylation eventually dictates PSMα3-membrane binding via electrostatic interactions with the lipid head groups. Furthermore, PSMα3 insertion within the lipid bilayer is favoured by hydrophobic interactions with the lipid acyl chains only in the fluid phase of membranes and not in the gel-like ordered domains. Strikingly, our real-time AFM imaging emphasizes how intermediate protofibrillar entities, formed along PSMα3 self-assembly and promoted at the membrane interface, likely disrupt membrane integrity via peptide accumulation and subsequent membrane thinning in a peptide concentration and lipid-dependent manner. Overall, our multiscale and multimodal approach sheds new light on the key roles of N-formylation and intermediate self-assembling entities, rather than mature fibrils, in dictating deleterious interactions of PSMα3 with membrane lipids, likely underscoring its ultimate cellular toxicity in vivo, and in turn S. aureus pathogenesis.
Collapse
Affiliation(s)
- Laura Bonnecaze
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Katlyn Jumel
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Anthony Vial
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Cécile Feuillie
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Michael Molinari
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Sophie Lecomte
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | |
Collapse
|
31
|
Flint Z, Grannemann H, Baffour K, Koti N, Taylor E, Grier E, Sutton C, Johnson D, Dandawate P, Patel R, Santra S, Banerjee T. Mechanistic Insights Behind the Self-Assembly of Human Insulin under the Influence of Surface-Engineered Gold Nanoparticles. ACS Chem Neurosci 2024; 15:2359-2371. [PMID: 38728258 PMCID: PMC11157486 DOI: 10.1021/acschemneuro.4c00226] [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: 04/13/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Elucidating the underlying principles of amyloid protein self-assembly at nanobio interfaces is extremely challenging due to the diversity in physicochemical properties of nanomaterials and their physical interactions with biological systems. It is, therefore, important to develop nanoscale materials with dynamic features and heterogeneities. In this work, through engineering of hierarchical polyethylene glycol (PEG) structures on gold nanoparticle (GNP) surfaces, tailored nanomaterials with different surface properties and conformations (GNPs-PEG) are created for modulating the self-assembly of a widely studied protein, insulin, under amyloidogenic conditions. Important biophysical studies including thioflavin T (ThT) binding, circular dichroism (CD), surface plasmon resonance (SPR), and atomic force microscopy (AFM) showed that higher-molecular weight GNPs-PEG triggered the formation of amyloid fibrils by promoting adsorption of proteins at nanoparticle surfaces and favoring primary nucleation rate. Moreover, the modulation of fibrillation kinetics reduces the overall toxicity of insulin oligomers and fibrils. In addition, the interaction between the PEG polymer and amyloidogenic insulin examined using MD simulations revealed major changes in the secondary structural elements of the B chain of insulin. The experimental findings provide molecular-level descriptions of how the PEGylated nanoparticle surface modulates protein adsorption and drives the self-assembly of insulin. This facile approach provides a new avenue for systematically altering the binding affinities on nanoscale surfaces by tailoring their topologies for examining adsorption-induced fibrillogenesis phenomena of amyloid proteins. Together, this study suggests the role of nanobio interfaces during surface-induced heterogeneous nucleation as a primary target for designing therapeutic interventions for amyloid-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Zachary Flint
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Haylee Grannemann
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Kristos Baffour
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Neelima Koti
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Emma Taylor
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Ethan Grier
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Carissa Sutton
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - David Johnson
- Molecular
Graphics and Modeling Laboratory, University
of Kansas, 2034 Becker
Drive, Lawrence, Kansas 66018, United States
| | - Prasad Dandawate
- Department
of Cancer Biology, The University of Kansas
Medical Center, Kansas City, Kansas 66160, United States
| | - Rishi Patel
- Jordan
Valley Innovation Center, Missouri State
University, 542 N. Boonville
Avenue, Springfield, Missouri 65806, United States
| | - Santimukul Santra
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Tuhina Banerjee
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| |
Collapse
|
32
|
Kalitnik A, Szefczyk M, Wojciechowska AW, Wojciechowski JW, Gąsior-Głogowska M, Olesiak-Bańska J, Kotulska M. Cytotoxic Staphylococcus aureus PSMα3 inhibits the aggregation of human insulin in vitro. Phys Chem Chem Phys 2024; 26:15587-15599. [PMID: 38757742 DOI: 10.1039/d4cp00669k] [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: 05/18/2024]
Abstract
Phenol-soluble modulins (PSMs) are extracellular short amphipathic peptides secreted by the bacteria Staphylococcus aureus (S. aureus). They play an essential role in the bacterial lifecycle, biofilm formation, and stabilisation. From the PSM family, PSMα3 has been of special interest recently due to its cytotoxicity and highly stable α-helical conformation, which also remains in its amyloid fibrils. In particular, PSMα3 fibrils were shown to be composed of self-associating "sheets" of α-helices oriented perpendicular to the fibril axis, mimicking the architecture of canonical cross-β fibrils. Therefore, they were called cross-α-fibrils. PSMα3 was synthesised and verified for identity with wild-type sequences (S. aureus). Then, using several experimental techniques, we evaluated its propensity for in vitro aggregation. According to our findings, synthetic PSMα3 (which lacks the N-terminal formyl groups found in bacteria) does not form amyloid fibrils and maintains α-helical conformation in a soluble monomeric form for several days of incubation. We also evaluated the influence of PSMα3 on human insulin fibrillation in vitro, using a variety of experimental approaches in combination with computational molecular studies. First, it was shown that PSMα3 drastically inhibits the fibrillation of human insulin. The anti-fibrillation effect of PSMα3 was concentration-dependent and required a concentration ratio of PSMα3: insulin equal to or above 1 : 100. Molecular modelling revealed that PSMα3 most likely inhibits the production of insulin primary nuclei by competing for residues involved in its dimerization.
Collapse
Affiliation(s)
- Aleksandra Kalitnik
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Monika Szefczyk
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Alicja W Wojciechowska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Jakub W Wojciechowski
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Marlena Gąsior-Głogowska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Joanna Olesiak-Bańska
- Institute of Advanced Materials, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Małgorzata Kotulska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
33
|
Zhao LL, Gu YX, Dong JH, Li XT, Pan HY, Xue CY, Liu Y, Zhou YL, Zhang XX. New G-Triplex DNA Dramatically Activates the Fluorescence of Thioflavin T and Acts as a Turn-On Fluorescent Sensor for Uracil-DNA Glycosylase Activity Detection. Anal Chem 2024; 96:8458-8466. [PMID: 38710075 DOI: 10.1021/acs.analchem.4c00164] [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: 05/08/2024]
Abstract
G-triplexes are G-rich oligonucleotides composed of three G-tracts and have absorbed much attention due to their potential biological functions and attractive performance in biosensing. Through the optimization of loop compositions, DNA lengths, and 5'-flanking bases of G-rich sequences, a new stable G-triplex sequence with 14 bases (G3-F15) was discovered to dramatically activate the fluorescence of Thioflavin T (ThT), a water-soluble fluorogenic dye. The fluorescence enhancement of ThT after binding with G3-F15 reached 3200 times, which was the strongest one by far among all of the G-rich sequences. The conformations of G3-F15 and G3-F15/ThT were studied by circular dichroism. The thermal stability measurements indicated that G3-F15 was a highly stable G-triplex structure. The conformations of G3-F15 and G3-F15/ThT in the presence of different metal cations were studied thoroughly by fluorescent spectroscopy, circular dichroism, and nuclear magnetic resonance. Furthermore, using the G3-F15/ThT complex as a fluorescent probe, a robust and simple turn-on fluorescent sensor for uracil-DNA glycosylase activity was developed. This study proposes a new systematic strategy to explore new functional G-rich sequences and their ligands, which will promote their applications in diagnosis, therapy, and biosensing.
Collapse
Affiliation(s)
- Ling-Li Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Yi-Xuan Gu
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jia-Hui Dong
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xiao-Tong Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hui-Yu Pan
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chen-Yu Xue
- Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100191, China
| | - Ying Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ying-Lin Zhou
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xin-Xiang Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
34
|
Gligorijevic N, Lujic T, Mutic T, Vasovic T, de Guzman MK, Acimovic J, Stanic-Vucinic D, Cirkovic Velickovic T. Ovalbumin interaction with polystyrene and polyethylene terephthalate microplastics alters its structural properties. Int J Biol Macromol 2024; 267:131564. [PMID: 38614174 DOI: 10.1016/j.ijbiomac.2024.131564] [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: 12/29/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Contaminating microplastics can interact with food proteins in the food matrix and during digestion. This study investigated adsorption of chicken egg protein ovalbumin to polystyrene (PS, 110 and 260 μm) and polyethylene terephthalate (PET, 140 μm) MPs in acidic and neutral conditions and alterations in ovalbumin structure. Ovalbumin adsorption affinity depended on MPs size (smaller > larger), type (PS > PET) and pH (pH 3 > pH 7). In bulk solution, MPs does not change ovalbumin secondary structure significantly, but induces loosening (at pH 3) and tightening (at pH 7) of tertiary structure. Formed soft corona exclusively consists of full length non-native ovalbumin, while in hard corona also shorter ovalbumin fragments were found. At pH 7 soft corona ovalbumin has rearranged but still preserved level of ordered secondary structure, resulting in preserved thermostability and proteolytic stability, but decreased ability to form fibrils upon heating. Secondary structure changes in soft corona resemble changes in native ovalbumin induced by heat treatment (80 °C). Ovalbumin is abundantly present in corona around microplastics also in the presence of other egg white proteins. These results imply that microplastics contaminating food may bind and change structure and functional properties of the main egg white protein.
Collapse
Affiliation(s)
- Nikola Gligorijevic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia; Department of Chemistry, University of Belgrade - Institute of Chemistry, Technology and Metallurgy, National Institute of Republic of Serbia, Belgrade, Serbia
| | - Tamara Lujic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Tamara Mutic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Tamara Vasovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Maria Krishna de Guzman
- Ghent University Global Campus, Yeonsu-gu, Incheon, South Korea; Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Jelena Acimovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Dragana Stanic-Vucinic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Tanja Cirkovic Velickovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia; Serbian Academy of Sciences and Arts, Belgrade, Serbia.
| |
Collapse
|
35
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
36
|
Francisco T, Malafaia D, Melo L, Silva AMS, Albuquerque HMT. Recent Advances in Fluorescent Theranostics for Alzheimer's Disease: A Comprehensive Survey on Design, Synthesis, and Properties. ACS OMEGA 2024; 9:13556-13591. [PMID: 38559945 PMCID: PMC10975685 DOI: 10.1021/acsomega.3c10417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative dementia that is rapidly becoming a major health problem, especially in developed countries because of their increasing life expectancy. Two main problems are often associated with the disease: (i) the absence of a widely accessible "gold-standard" for early diagnosis and (ii) lack of effective therapies with disease-modifying effects. The recent success of the monoclonal antibody lecanemab played an important role not only in clarifying a possible druggable pathway but also in spelling the revival of small molecule drug discovery. Unlike bulky biologics, small molecules are structurally less complex, generally cheaper, and compatible with at-home oral consumption, making it feasible for people to start their drug regimen early and stay on it longer. In this sense, small-molecule near-infrared fluorescent theranostics have been gaining more and more attention from the scientific community, as they have the potential to simultaneously provide diagnostic outputs and deliver therapeutic action, paving the way toward personalized medicine in AD patients. They also have the potential to shift the diagnostic "status-quo" from expensive and limited-access PET radiotracers toward inexpensive and handy imaging tools widely available for primary patient screening and preclinical animal studies. Herein, we review the most recent advances in the field of fluorescent theranostics for Alzheimer's disease, detailing their design strategies, synthetic approaches and imaging and therapeutic properties in vitro and in vivo. With this Review, we intend to provide a milestone in the acquired knowledge in the field of AD theranostics, encouraging the future development of properly designed theranostic compounds with improved chances to reach clinical applications.
Collapse
Affiliation(s)
- Telmo
N. Francisco
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Daniela Malafaia
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Lúcia Melo
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Artur M. S. Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Hélio M. T. Albuquerque
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
37
|
Stepanchuk AA, Stys PK. Spectral Fluorescence Pathology of Protein Misfolding Disorders. ACS Chem Neurosci 2024; 15:898-908. [PMID: 38407017 DOI: 10.1021/acschemneuro.3c00798] [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: 02/27/2024] Open
Abstract
Protein misfolding has been extensively studied in the context of neurodegenerative disorders and systemic amyloidoses. Due to misfolding and aggregation of proteins being highly heterogeneous and generating a variety of structures, a growing body of evidence illustrates numerous ways how the aggregates contribute to progression of diseases such as Alzheimer's disease, Parkinson's disease, and prion disorders. Different misfolded species of the same protein, commonly referred to as strains, appear to play a significant role in shaping the disease clinical phenotype and clinical progression. The distinct toxicity profiles of various misfolded proteins underscore their importance. Current diagnostics struggle to differentiate among these strains early in the disease course. This review explores the potential of spectral fluorescence approaches to illuminate the complexities of protein misfolding pathology and discusses the applications of advanced spectral methods in the detection and characterization of protein misfolding disorders. By examining spectrally variable probes, current data analysis approaches, and important considerations for the use of these techniques, this review aims to provide an overview of the progress made in this field and highlights directions for future research.
Collapse
Affiliation(s)
- Anastasiia A Stepanchuk
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Peter K Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
38
|
Silva-Reis SC, Costa VM, Correia da Silva D, Pereira DM, Correia XC, Costa-Almeida HF, García-Mera X, Rodríguez-Borges JE, Sampaio-Dias IE. Exploring structural determinants of neuroprotection bias on novel glypromate conjugates with bioactive amines. Eur J Med Chem 2024; 267:116174. [PMID: 38306884 DOI: 10.1016/j.ejmech.2024.116174] [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: 10/06/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
Neurodegenerative disorders of the central nervous system (CNS) such as Alzheimer's and Parkinson's diseases, afflict millions globally, posing a significant public health challenge. Despite extensive research, a critical hurdle in effectively treating neurodegenerative diseases is the lack of neuroprotective drugs that can halt or reverse the underlying disease processes. In this work, we took advantage of the neuroprotective properties of the neuropeptide glycyl-l-prolyl-l-glutamic acid (Glypromate) for the development of new peptidomimetics using l-pipecolic acid as a proline surrogate and exploring their chemical conjugation with relevant active pharmaceutical ingredients (API) via a peptide bond. Together with prolyl-based Glypromate conjugates, a total of 36 conjugates were toxicologically and biologically evaluated. In this series, the results obtained showed that a constrained ring (l-proline) at the central position of the peptide motif accounts for enhanced toxicological profiles and biological effects using undifferentiated and differentiated human neuroblastoma SH-SY5Y cells. Additionally, it was shown that biased biological responses are API-dependent. Conjugation with (R)-1-aminoindane led to a 38-43% reduction of protein aggregation induced by Aβ25-35 (10 μM), denoting a 3.2-3.6-fold improvement in comparison with the parent neuropeptide, with no significative difference between functionalization at α and γ-carboxyl ends. On the other hand, the best-performing neuroprotective conjugate against the toxicity elicited by 6-hydroxydopamine (6-OHDA, 125 μM) was obtained by conjugation with memantine at the α-carboxyl end, resulting in a 2.3-fold improvement of the neuroprotection capacity in comparison with Glypromate neuropeptide. Altogether, the chemical strategy explored in this work shows that the neuroprotective capacity of Glypromate can be modified and fine-tuned, opening a new avenue for the development of biased neurotherapeutics for CNS-related disorders.
Collapse
Affiliation(s)
- Sara C Silva-Reis
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal; UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera M Costa
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Daniela Correia da Silva
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - David M Pereira
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Xavier Cruz Correia
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Hugo F Costa-Almeida
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Xerardo García-Mera
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, E-15782, Santiago de Compostela, Spain
| | - José E Rodríguez-Borges
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Ivo E Sampaio-Dias
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal.
| |
Collapse
|
39
|
Lage L, Rodriguez-Perez AI, Villar-Cheda B, Labandeira-Garcia JL, Dominguez-Meijide A. Angiotensin type 1 receptor activation promotes neuronal and glial alpha-synuclein aggregation and transmission. NPJ Parkinsons Dis 2024; 10:37. [PMID: 38368444 PMCID: PMC10874459 DOI: 10.1038/s41531-024-00650-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/02/2024] [Indexed: 02/19/2024] Open
Abstract
The brain renin-angiotensin system (RAS) has been related to dopaminergic degeneration, and high expression of the angiotensin II (AngII) type 1 receptor (AT1) gene is a marker of the most vulnerable neurons in humans. However, it is unknown whether AngII/AT1 overactivation affects α-synuclein aggregation and transmission. In vitro, AngII/AT1 activation increased α-synuclein aggregation in dopaminergic neurons and microglial cells, which was related to AngII-induced NADPH-oxidase activation and intracellular calcium raising. In mice, AngII/AT1 activation was involved in MPTP-induced increase in α-synuclein expression and aggregation, as they significantly decreased in mice treated with the AT1 blocker telmisartan and AT1 knockout mice. Cell co-cultures (transwells) revealed strong transmission of α-synuclein from dopaminergic neurons to astrocytes and microglia. AngII induced a higher α-synuclein uptake by microglial cells and an increase in the transfer of α-synuclein among astroglial cells. However, AngII did not increase the release of α-synuclein by neurons. The results further support brain RAS dysregulation as a major mechanism for the progression of Parkinson's disease, and AT1 inhibition and RAS modulation as therapeutic targets.
Collapse
Affiliation(s)
- Lucia Lage
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Begoña Villar-Cheda
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Antonio Dominguez-Meijide
- Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
40
|
Zimmer VC, Lauer AA, Haupenthal V, Stahlmann CP, Mett J, Grösgen S, Hundsdörfer B, Rothhaar T, Endres K, Eckhardt M, Hartmann T, Grimm HS, Grimm MOW. A bidirectional link between sulfatide and Alzheimer's disease. Cell Chem Biol 2024; 31:265-283.e7. [PMID: 37972592 DOI: 10.1016/j.chembiol.2023.10.021] [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: 12/23/2022] [Revised: 09/05/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
Reduced sulfatide level is found in Alzheimer's disease (AD) patients. Here, we demonstrate that amyloid precursor protein (APP) processing regulates sulfatide synthesis and vice versa. Different cell culture models and transgenic mice models devoid of APP processing or in particular the APP intracellular domain (AICD) reveal that AICD decreases Gal3st1/CST expression and subsequently sulfatide synthesis. In return, sulfatide supplementation decreases Aβ generation by reducing β-secretase (BACE1) and γ-secretase processing of APP. Increased BACE1 lysosomal degradation leads to reduced BACE1 protein level in endosomes. Reduced γ-secretase activity is caused by a direct effect on γ-secretase activity and reduced amounts of γ-secretase components in lipid rafts. Similar changes were observed by analyzing cells and mice brain samples deficient of arylsulfatase A responsible for sulfatide degradation or knocked down in Gal3st1/CST. In line with these findings, addition of sulfatides to brain homogenates of AD patients resulted in reduced γ-secretase activity. Human brain APP level shows a significant negative correlation with GAL3ST1/CST expression underlining the in vivo relevance of sulfatide homeostasis in AD.
Collapse
Affiliation(s)
- Valerie Christin Zimmer
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Anna Andrea Lauer
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - Viola Haupenthal
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Christoph Peter Stahlmann
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Janine Mett
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany; Biosciences Zoology/Physiology-Neurobiology, ZHMB (Center of Human and Molecular Biology), Faculty NT-Natural Science and Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Sven Grösgen
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Benjamin Hundsdörfer
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Tatjana Rothhaar
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University, 55099 Mainz, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Tobias Hartmann
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany
| | - Heike Sabine Grimm
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - Marcus Otto Walter Grimm
- Deutsches Institut für Demenzprävention (DIDP), Neurodegeneration and Neurobiology and Experimental Neurology, Saarland University, 66424 Homburg/Saar, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany.
| |
Collapse
|
41
|
Chisholm TS, Hunter CA. A closer look at amyloid ligands, and what they tell us about protein aggregates. Chem Soc Rev 2024; 53:1354-1374. [PMID: 38116736 DOI: 10.1039/d3cs00518f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
The accumulation of amyloid fibrils is characteristic of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease. Detecting these fibrils with fluorescent or radiolabelled ligands is one strategy for diagnosing and better understanding these diseases. A vast number of amyloid-binding ligands have been reported in the literature as a result. To obtain a better understanding of how amyloid ligands bind, we have compiled a database of 3457 experimental dissociation constants for 2076 unique amyloid-binding ligands. These ligands target Aβ, tau, or αSyn fibrils, as well as relevant biological samples including AD brain homogenates. From this database significant variation in the reported dissociation constants of ligands was found, possibly due to differences in the morphology of the fibrils being studied. Ligands were also found to bind to Aβ(1-40) and Aβ(1-42) fibrils with similar affinities, whereas a greater difference was found for binding to Aβ and tau or αSyn fibrils. Next, the binding of ligands to fibrils was shown to be largely limited by the hydrophobic effect. Some Aβ ligands do not fit into this hydrophobicity-limited model, suggesting that polar interactions can play an important role when binding to this target. Finally several binding site models were outlined for amyloid fibrils that describe what ligands target what binding sites. These models provide a foundation for interpreting and designing site-specific binding assays.
Collapse
Affiliation(s)
- Timothy S Chisholm
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1 EW, UK.
| | - Christopher A Hunter
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1 EW, UK.
| |
Collapse
|
42
|
Ghosh P, Shokeen K, Mondal S, Kandasamy T, Kumar S, Ghosh SS, Iyer PK. Amyloid Targeting Red Emitting AIE Dots for Diagnostic and Therapeutic Application against Alzheimer's Disease. ACS Chem Neurosci 2024; 15:268-277. [PMID: 38170988 DOI: 10.1021/acschemneuro.3c00473] [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: 01/05/2024] Open
Abstract
The emergence of neurodegenerative diseases is connected to several pathogenic factors, including metal ions, amyloidogenic proteins, and reactive oxygen species. Recent studies suggest that cytotoxicity is caused by the small, dynamic, and metastable nature of early stage oligomeric species. This work introduces a small molecule-based red-emitting probe with smart features such as increased reactivities against multiple targets, metal-free amyloid-β (Aβ), and metal-bound amyloid-β (Aβ), and most importantly, early stage oligomeric species which are associated with the most common and widespread type of dementia, Alzheimer's disease (AD). Theoretical analyses like molecular dynamics simulation and molecular docking were performed to confirm the reactivity of the molecule toward Aβ and found some excellent interactions between the molecule and the peptide. The in vitro and cellular studies demonstrated that this highly biocompatible molecule effectively reduces the structural damage to mitochondria while shielding cells from apoptosis, scavenges ROS (reactive oxygen species), and attenuates multifaceted amyloid toxicity.
Collapse
Affiliation(s)
- Priyam Ghosh
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kamal Shokeen
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Subrata Mondal
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Thirukumaran Kandasamy
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Sachin Kumar
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Siddhartha Sankar Ghosh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
- Center for Nanotechnology, Indian Institute of Technology Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Parameswar Krishnan Iyer
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
- Center for Nanotechnology, Indian Institute of Technology Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Assam 781039, India
| |
Collapse
|
43
|
Sun Z, Liu H, Dai D, Lyu H, Huang R, Wang W, Guo C. Injectable cell-laden silk acid hydrogel. Chem Commun (Camb) 2024; 60:316-319. [PMID: 38063025 DOI: 10.1039/d3cc04280d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
This study presents an injectable cell-laden hydrogel system based on silk acid, a carboxylated derivative of natural silk fibroin, which exhibits promising applications in biomedicine. The hydrogel is produced under physiological conditions (37 °C and pH 7.4) via physical crosslinking. Notably, the hydrogel demonstrates remarkable cytocompatibility, enabling efficient cell encapsulation, and exhibits good injectability. These promising results strongly indicate the potential of silk acid hydrogel for transformative applications, including 3D cell culture, targeted cell delivery, and tissue engineering.
Collapse
Affiliation(s)
- Ziyang Sun
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Haoran Liu
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Dandan Dai
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Hao Lyu
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Ruochuan Huang
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Wenzhao Wang
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, Zhejiang, 310023, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310030, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
| |
Collapse
|
44
|
Skates E, Delattre H, Schofield Z, Asally M, Soyer OS. Thioflavin T indicates mitochondrial membrane potential in mammalian cells. BIOPHYSICAL REPORTS 2023; 3:100134. [PMID: 38026684 PMCID: PMC10679866 DOI: 10.1016/j.bpr.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
The fluorescent benzothiazole dye thioflavin T (ThT) is widely used as a marker for protein aggregates, most commonly in the context of neurodegenerative disease research and diagnosis. Recently, this same dye was shown to indicate membrane potential in bacteria due to its cationic nature. This finding prompted a question whether ThT fluorescence is linked to the membrane potential in mammalian cells, which would be important for appropriate utilization of ThT in research and diagnosis. Here, we show that ThT localizes into the mitochondria of HeLa cells in a membrane-potential-dependent manner. Specifically, ThT colocalized in cells with the mitochondrial membrane potential indicator tetramethylrhodamine methyl ester (TMRM) and gave similar temporal responses as TMRM to treatment with a protonophore, carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP). Additionally, we found that presence of ThT together with exposure to blue light (λ = 405 nm), but neither factor alone, caused depolarization of mitochondrial membrane potential. This additive effect of the concentration and blue light was recapitulated by a mathematical model implementing the potential-dependent distribution of ThT and its effect on mitochondrial membrane potential through photosensitization. These results show that ThT can act as a mitochondrial membrane potential indicator in mammalian cells, when used at low concentrations and with low blue light exposure. However, it causes dissipation of the mitochondrial membrane potential depending additively on its concentrations and blue light exposure. This conclusion motivates a re-evaluation of ThT's use at micromolar range in live-cell analyses and indicates that this dye can enable future studies on the potential connections between mitochondrial membrane potential dynamics and protein aggregation.
Collapse
Affiliation(s)
- Emily Skates
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, United Kingdom
- Midlands Integrative Doctoral Training Program; University of Warwick, Coventry, United Kingdom
| | - Hadrien Delattre
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Zoe Schofield
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, United Kingdom
| | - Munehiro Asally
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, United Kingdom
| | - Orkun S. Soyer
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Warwick Integrative Synthetic Biology Centre (WISB), University of Warwick, Coventry, United Kingdom
| |
Collapse
|
45
|
Gao W, Liu W, Dong X, Sun Y. Albumin-manganese dioxide nanocomposites: a potent inhibitor and ROS scavenger against Alzheimer's β-amyloid fibrillogenesis and neuroinflammation. J Mater Chem B 2023; 11:10482-10496. [PMID: 37909060 DOI: 10.1039/d3tb01763j] [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: 11/02/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease pathologically caused by amyloid-β protein (Aβ) aggregation, oxidative stress, and neuroinflammation. The pathogenesis of AD is still uncertain and intricate, and helpful therapy has rarely been recorded. So, discovering amyloid modulators is deemed a promising avenue for preventing and treating AD. In this study, human serum albumin (HSA), a protein-based Aβ inhibitor, was utilized as a template to guide the synthesis of HSA-manganese dioxide nanocomposites (HMn NCs) through biomineralization. The in situ formed MnO2 in HSA endows this nano-platform with outstanding reactive oxygen species (ROS) scavenging capability, including superoxide dismutase-mimetic and catalase-mimetic activities, which could scavenge the plethora of superoxide anion radicals and hydrogen peroxide. More importantly, the HMn NCs show enhanced potency in suppressing Aβ fibrillization compared with HSA, which further alleviates Aβ-mediated SH-SY5Y neurotoxicity by scavenging excessive ROS. Moreover, it is demonstrated that HMn NCs reduce Aβ-related inflammation in BV-2 cells by lowering tumor necrosis factor-α and interleukin-6. Furthermore, transgenic C. elegans studies showed that HMn NCs could remove Aβ plaques, reduce ROS in CL2006 worms, and promote the lifespan extension of worms. Thus, HMn NCs provide a promising tactic to facilitate the application of multifunctional nanocomposites in AD treatment.
Collapse
Affiliation(s)
- Weiqun Gao
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| | - Wei Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| |
Collapse
|
46
|
Wang W, Qu L, Cui Z, Lu F, Li L, Liu F. Citrus Flavonoid Hesperetin Inhibits α-Synuclein Fibrillogenesis, Disrupts Mature Fibrils, and Reduces Their Cytotoxicity: In Vitro and In Vivo Studies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16174-16183. [PMID: 37870747 DOI: 10.1021/acs.jafc.3c06816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Misfolding and subsequent fibrillogenesis of α-synuclein (αSN) significantly influence the development of Parkinson's disease (PD). This study reports the inhibitory effect of citrus flavonoid hesperetin (Hst) on αSN fibrillation. Based on thioflavin T fluorometry and atomic force microscopy studies, Hst inhibited αSN fibrillation by interfering with initial nucleation and slowing the elongation rate. Furthermore, the inhibitory effect was concentration-dependent with a half-maximal inhibitory concentration of 24.4 μM. Cytotoxicity experiments showed that 100 μM Hst significantly reduced the cytotoxicity of αSN aggregates and maintained 98.4% cell activity. In addition, Hst disassembled the preprepared αSN fibrils into smaller and less-toxic aggregates. Excitingly, supplementation with 100 μM Hst inhibited the accumulation of 36.3% αSN in NL5901 and restored the amyloid-induced reduction in NL5901 lipid abundance, extending the mean lifespan of NL5901 to 23 d. These findings could support the use of Hst as a dietary supplement to regulate αSN fibrillation and prevent the development of PD.
Collapse
Affiliation(s)
- Wenqian Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Lili Qu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Zhan Cui
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Li Li
- College of Science, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| |
Collapse
|
47
|
Kumar M, Ivanova MI, Ramamoorthy A. Non-micellar ganglioside GM1 induces an instantaneous conformational change in Aβ 42 leading to the modulation of the peptide amyloid-fibril pathway. Biophys Chem 2023; 301:107091. [PMID: 37549471 DOI: 10.1016/j.bpc.2023.107091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Alzheimer's disease is a progressive degenerative condition that mainly affects cognition and memory. Recently, distinct clinical and neuropathological phenotypes have been identified in AD. Studies revealed that structural variation in Aβ fibrillar aggregates correlates with distinct disease phenotypes. Moreover, environmental surroundings, including other biomolecules such as proteins and lipids, have been shown to interact and modulate Aβ aggregation. Model membranes containing ganglioside (GM1) clusters are specifically known to promote Aβ fibrillogenesis. This study unravels the modulatory effect of non-micellar GM1, a glycosphingolipid frequently released from the damaged neuronal membranes, on Aβ42 amyloid fibril formation. Using far-UV circular dichroism experiments, we observed a change in the peptide secondary structure from random-coil to β-turn structures with subsequent generation of predominantly β-sheet-rich species upon interaction with GM1. Thioflavin-T (ThT) fluorescence assays further indicated that GM1 likely interacts with an amyloidogenic Aβ42 intermediate species leading to a possible formation of GM1-modified Aβ42 fibril. Statistically, no significant difference in toxicity to RA-differentiated SH-SY5Y cells was observed between Aβ42 fibrils and GM1-tweaked Aβ42 aggregates. Moreover, GM1-modified Aβ42 aggregates exhibited prion-like properties in catalyzing the amyloid fibril formation of both major isomers of Aβ, Aβ40, and Aβ42.
Collapse
Affiliation(s)
- Manjeet Kumar
- Biophysics, Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Magdalena I Ivanova
- Biophysics, Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-1055, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ayyalusamy Ramamoorthy
- Biophysics, Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-1055, USA.
| |
Collapse
|
48
|
Asgarshamsi MH, Dehkordi MM, Beigi HM, Fassihi A, Saghaie L. Synthesis of Some Benzothiazole Derivatives Based on 3-Hydroxypyridine-4-one and Benzaldehyde and Evaluation of Their β-Amyloid Aggregation Inhibition Using both Experimental Methods and Molecular Dynamic Simulation. Chem Biodivers 2023; 20:e202301113. [PMID: 37691012 DOI: 10.1002/cbdv.202301113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/12/2023]
Abstract
Some novel inhibitors based on the (benzo[d]thiazol-2-yl)-1-phenylmethanimine derivatives were designed to reduce the aggregation process in Alzheimer's disease. These structures seem to mimic stilbene-like scaffold, while the benzothiazole moiety "locks" the thioflavin T binding site. Other inhibitors were designed based on 2-((benzo[d]thiazol-2-ylimino)methyl)-5-(benzyloxy)-1-methylpyridin-4(H)-one derivatives. Benzo[d]thiazol-2-amine derivatives were prepared by the reaction of aniline derivatives with ammonium thiocyanate in the presence of bromine/acetic acid. Then, the reaction of amines with benzaldehyde derivatives and 5-(benzyloxy)-1-methyl-4-oxo-1,4-dihydropyridine-2-carbaldehyde gave the desired compounds. The plate reader-based fibrillation assay was done to evaluate the inhibition of Aβ aggregation. Also, molecular dynamic simulation was carried out to clarify the interaction manner of the designed compounds with Aβ formation. The biological evaluation proved 5a and 7e as the best inhibitor of the Aβ aggregation. compound 5a in the concentration of 50 μM inhibited Aβ fibril formation better than 7e. MD simulation elucidated that the Aβ aggregation inhibitors in different concentrations represented different binding conformations throughout the entire or in one area of Aβ. MD showed the ligands in lower concentrations accumulate in an area of Aβ aggregations and separate one fibril from the aggregated Aβ. On the contrary, in higher concentrations, the ligands tend to be located through the entire Aβ.
Collapse
Affiliation(s)
- Mohammad Hossein Asgarshamsi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran, 73441-81746
- Isfahan Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrdad Mohammadpour Dehkordi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran, 73441-81746
- Isfahan Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Mohammad Beigi
- Protein Biophysics, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 227, 2800, Kgs. Lyngby, Denmark
| | - Afshin Fassihi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran, 73441-81746
- Isfahan Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Lotfollah Saghaie
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran, 73441-81746
| |
Collapse
|
49
|
Peña A, Sánchez NS, Padilla-Garfias F, Ramiro-Cortés Y, Araiza-Villanueva M, Calahorra M. The Use of Thioflavin T for the Estimation and Measurement of the Plasma Membrane Electric Potential Difference in Different Yeast Strains. J Fungi (Basel) 2023; 9:948. [PMID: 37755056 PMCID: PMC10532974 DOI: 10.3390/jof9090948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
The use of the cationic, dye thioflavin T (ThT), to estimate the electric plasma membrane potential difference (PMP) via the fluorescence changes and to obtain its actual values from the accumulation of the dye, considering important correction factors by its binding to the internal components of the cell, was described previously for baker's yeast. However, it was considered important to explore whether the method developed could be applied to other yeast strains. Alternative ways to estimate the PMP by using flow cytometry and a multi-well plate reader are also presented here. The methods were tested with other strains of Saccharomyces cerevisiae (W303-1A and FY833), as well as with non-conventional yeasts: Debaryomyces hansenii, Candida albicans, Meyerozyma guilliermondii, and Rhodotorula mucilaginosa. Results of the estimation of the PMP via the fluorescence changes under different conditions were adequate with all strains. Consistent results were also obtained with several mutants of the main monovalent transporters, validating ThT as a monitor for PMP estimation.
Collapse
Affiliation(s)
- Antonio Peña
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico; (F.P.-G.); (M.A.-V.); (M.C.)
| | - Norma Silvia Sánchez
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico; (F.P.-G.); (M.A.-V.); (M.C.)
| | - Francisco Padilla-Garfias
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico; (F.P.-G.); (M.A.-V.); (M.C.)
| | - Yazmín Ramiro-Cortés
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico;
| | - Minerva Araiza-Villanueva
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico; (F.P.-G.); (M.A.-V.); (M.C.)
| | - Martha Calahorra
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, México City 04510, Mexico; (F.P.-G.); (M.A.-V.); (M.C.)
| |
Collapse
|
50
|
Zhang NY, Hou DY, Hu XJ, Liang JX, Wang MD, Song ZZ, Yi L, Wang ZJ, An HW, Xu W, Wang H. Nano Proteolysis Targeting Chimeras (PROTACs) with Anti-Hook Effect for Tumor Therapy. Angew Chem Int Ed Engl 2023; 62:e202308049. [PMID: 37486792 DOI: 10.1002/anie.202308049] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023]
Abstract
Proteolysis targeting chimera (PROTAC) is an emerging pharmacological modality with innovated post-translational protein degradation capabilities. However, off-target induced unintended tissue effects and intrinsic "hook effect" hinder PROTAC biotechnology to be maturely developed. Herein, an intracellular fabricated nano proteolysis targeting chimeras (Nano-PROTACs) modality with a center-spoke degradation network for achieving efficient dose-dependent protein degradation in tumor is reported. The PROTAC precursors are triggered by higher GSH concentrations inside tumor cells, which subsequently in situ self-assemble into Nano-PROTACs through intermolecular hydrogen bond interactions. The fibrous Nano-PROTACs can form effective polynary complexes and E3 ligases degradation network with multi-binding sites, achieving dose-dependent protein degradation with "anti-hook effect". The generality and efficacy of Nano-PROTACs are validated by degrading variable protein of interest (POI) such as epidermal growth factor receptor (EGFR) and androgen receptor (AR) in a wide-range dose-dependent manner with a 95 % degradation rate and long-lasting potency up to 72 h in vitro. Significantly, Nano-PROTACs achieve in vivo dose-dependent protein degradation up to 79 % and tumor growth inhibition in A549 and LNCap xenograft mice models, respectively. Taking advantages of in situ self-assembly strategy, the Nano-PROTACs provide a generalizable platform to promote precise clinical translational application of PROTAC.
Collapse
Affiliation(s)
- Ni-Yuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Da-Yong Hou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Department of Urology, Harbin Medical University Cancer Hospital, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Xing-Jie Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
| | - Jian-Xiao Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Man-Di Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhang-Zhi Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Li Yi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhi-Jia Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Department of Urology, Harbin Medical University Cancer Hospital, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Hong-Wei An
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wanhai Xu
- Department of Urology, Harbin Medical University Cancer Hospital, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|