1
|
Olsen WP, Larsen AKK, Christensen JL, Malle MG, Otzen DE. Investigating strategies for creating cross-linked amyloid fibril networks through branching of amyloid growth. Colloids Surf B Biointerfaces 2025; 251:114617. [PMID: 40068237 DOI: 10.1016/j.colsurfb.2025.114617] [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: 01/05/2025] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Hydrogel biomaterials have been extensively explored for applications in medicine, materials science, and the development of functionalized materials. Traditionally, hydrogels were produced using simple polymers, but advancements over recent decades have enabled the use of biological materials such as proteins, peptides, polysaccharides, and even amyloid fibrils. Among these, amyloid-based hydrogels have demonstrated unique advantages, including enhanced cell adhesion and differentiation. Furthermore, they can be engineered as living materials using bacteria capable of producing and repairing the hydrogel in situ. Here we investigate novel strategies for controlling amyloid fibrillation using the functional amyloid CsgA. We designed fusion proteins combining two CsgA moieties to explore methods for creating branched fibril networks. Our approach utilized two distinct strategies: passive and active branching. The passive strategy involved direct fusion of two CsgA moieties separated by a designed alpha-helical linker and engineered to integrate into fibrils without external intervention. The active branching approach incorporated a redox-sensitive CsgA variant containing an internal disulfide bridge that blocks fibrillation until reduced. This design allows for precise control of amyloid fibrillation in the active variants. We analyzed these constructs qualitatively approach using a combination of transmission electron microscopy (TEM), real-time atomic force microscopy (AFM), and total internal reflection fluorescence (TIRF) microscopy, supported by quantitative image analysis. While we did not observe direct evidence of fibril branching, our modifications led to significant changes in fibrillation behavior. Notably, TIRF imaging revealed a marked increase in high-density fibril regions following the activation of our engineered constructs, indicating the potential for controlled assembly of higher-order structures. These findings provide new insights into controlling amyloid fibrillation and suggest alternative strategies for manipulating fibril organization. The observed ability to alter local fibril density through chemical triggers offers promising directions for developing responsive biomaterials. We propose refinements for future design and suggest new directions to optimize amyloid-based hydrogels for next-generation biomaterial applications.
Collapse
Affiliation(s)
- William P Olsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C 8000, Denmark; Sino-Danish College (SDC), University of Chinese Academy of Sciences, Beijing, China
| | - Anne-Kathrine K Larsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C 8000, Denmark; Sino-Danish College (SDC), University of Chinese Academy of Sciences, Beijing, China
| | - Jakob L Christensen
- Department of Applied Mathematics and Computer Science, DTU Compute, DTU Technical University of Denmark, Lyngby, Denmark
| | - Mette G Malle
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C 8000, Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C 8000, Denmark.
| |
Collapse
|
2
|
Hazari MA, Kannan G, Dasgupta S, Pavan MK, Jha AK, Sultana F, Pujahari SR, Singh S, Dutta S, Pydi SP, Dutta S, Zafar H, Bhaumik P, Kumar A, Sen S. Faster Amylin Aggregation on Fibrillar Collagen I Hastens Diabetic Progression through β-Cell Death and Loss of Function. J Am Chem Soc 2025. [PMID: 40300850 DOI: 10.1021/jacs.4c15698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Amyloid deposition of the neuroendocrine peptide amylin in islet tissues is a hallmark of type 2 diabetes (T2DM), leading to β-cell toxicity through nutrient deprivation, membrane rupture, and apoptosis. Though accumulation of toxic amylin aggregates in islet matrices is well documented, the role of the islet extracellular matrix in mediating amylin aggregation and its pathological consequences remains elusive. Here, we address this question by probing amylin interaction with collagen I (Col)─whose expression in the islet tissue increases during diabetes progression. By combining multiple biophysical techniques, we show that hydrophobic, hydrophilic, and cation-π interactions regulate amylin binding to Col, with fibrillar Col driving faster amylin aggregation. Amylin-entangled Col matrices containing high amounts of amylin induce death and loss of function in INS1E β-cells. Together, our results illustrate how amylin incorporation in islet matrices through amylin-Col interactions drives T2DM progression by impacting β-cell viability and insulin secretion.
Collapse
Affiliation(s)
| | - Gautam Kannan
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Subrata Dasgupta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Musale Krushna Pavan
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
| | - Akash Kumar Jha
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Farhin Sultana
- Department of Oncogene Regulation, CNCI, Kolkata 700026, India
| | | | - Simran Singh
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | - Sarbajeet Dutta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Sai Prasad Pydi
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | | | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | - Prasenjit Bhaumik
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Ashutosh Kumar
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Shamik Sen
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
3
|
Shaw R, Patel K, Chimthanawala NMA, Sathaye S, Maji SK. Peptide-Based Functional Amyloid Hydrogel Enhances Wound Healing in Normal and Diabetic Rat Models. Adv Healthc Mater 2025; 14:e2403560. [PMID: 39935087 DOI: 10.1002/adhm.202403560] [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: 09/18/2024] [Revised: 01/23/2025] [Indexed: 02/13/2025]
Abstract
The inability to heal on time is a key component of chronic wounds, which can result in economic, psychological, and physiological burdens. Hydrogels based on amyloid can imitate the extracellular matrix and function as scaffolds for healing wounds. In this direction, a wound dressing inspired by peptide-based amyloid hydrogel is fabricated here. The results demonstrate that the amyloid hydrogel improves the three essential components of skin tissue regeneration: cell migration, proliferation, and collagen remodeling, both in vitro and in vivo. Furthermore, the amyloid hydrogel accelerates wound healing and promotes wound closure within 9 and 15 d in normal and diabetic rats, respectively. Microscopic evaluation of the wound region demonstrates the ultimate stages of regeneration and skin reformation toward normal skin compared to the untreated wound. Hematoxylin and eosin-stained hydrogel-treated wound sites reveal faster dermal bridging, angiogenesis, and epidermal repair in both acute and chronic conditions. The hydrogel creates an environment that encourages the growth of dermal fibroblasts and the release of cytokines, decreasing inflammation with concomitant enhancement of collagen production at the site of injury. Thus, these findings suggest that amyloid-based hydrogel can be a promising candidate for application in acute and chronic wound healing.
Collapse
Affiliation(s)
- Ranjit Shaw
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), IIT Bombay, Powai, Mumbai, 400076, India
| | - Niyamat M A Chimthanawala
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400019, India
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400019, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), IIT Bombay, Powai, Mumbai, 400076, India
| |
Collapse
|
4
|
Xuan Q, Cai J, Gao Y, Qiao X, Jin T, Peydayesh M, Zhou J, Sun Q, Zhan L, Liu B, Wang P, Li H, Chen C, Mezzenga R. Amyloid-Templated Ceria Nanozyme Reinforced Microneedle for Diabetic Wound Treatments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417774. [PMID: 39995378 PMCID: PMC12004906 DOI: 10.1002/adma.202417774] [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: 11/16/2024] [Revised: 02/15/2025] [Indexed: 02/26/2025]
Abstract
Amyloid fibrils have emerged as excellent templates and building blocks for the development of ordered functional materials with considerable potential in biomedical applications. Here, lysozyme amyloid fibrils (Lys-AFs) are employed as templates for the in situ synthesis of ceria nanozymes (Lys-AFs-Ceria) with ultrafine dimensions, an optimized Ce3+/Ce4+ ratio, and uniform distribution on the fibril surface, addressing the challenges of low catalytic efficiency and high susceptibility to aggregation typical of traditional methods. As a proof of concept, it is further applied Lys-AFs-Ceria to develop hydrogel/microneedle for treating bacteria-infected diabetic wounds via non-covalent interactions between polyphenols and amyloid fibrils incorporating glucose oxidase (GOX). The hydrogel/microneedle facilitates superoxide dismutase and catalase cascade catalysis by Lys-AFs-Ceria, and integrates GOX-mediated glucose consumption, synergistically achieving glucose reduction, reactive oxygen species elimination, and hypoxia alleviation in the diabetic wound infection microenvironment. In addition to antibacterial properties and tissue regeneration promotion of Lys-AFs scaffold, Lys-AFs-Ceria regulates macrophages polarization toward an anti-inflammatory M2 state. Collectively, these attributes contribute to the enhanced efficacy of diabetic wound healing, with in vivo studies demonstrating increased healing efficiency following a single application, and more in general an effective strategy toward high-catalytic and stable nanozymes.
Collapse
Affiliation(s)
- Qize Xuan
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
- State Key Laboratory of Bioreactor Engineering Center, School of BiotechnologyEast China University of Science and TechnologyShanghai200237China
| | - Jiazhe Cai
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Yuan Gao
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Xinchi Qiao
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Tonghui Jin
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
| | - Mohammad Peydayesh
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
| | - Jiangtao Zhou
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
| | - Qiyao Sun
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
| | - Lijian Zhan
- Institute for Biomedical EngineeringETH ZürichZürich8092Switzerland
| | - Bin Liu
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
| | - Ping Wang
- Department of Bioproducts and Biosystems EngineeringUniversity of MinnesotaSt PaulMN55108USA
| | - Hui Li
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Chao Chen
- Institute for Environmental Pollution and Health, School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
- State Key Laboratory of Bioreactor Engineering Center, School of BiotechnologyEast China University of Science and TechnologyShanghai200237China
| | - Raffaele Mezzenga
- Department of Health Sciences and TechnologyETH ZürichSchmelzbergstrasse 9Zürich8092Switzerland
- Department of MaterialsETH ZürichWolfgang‐Pauli‐Strasse 10Zürich8049Switzerland
| |
Collapse
|
5
|
Wang L, Zhou Y, Zhang Y, Wang Z, Li A, Chen H, Huang Y, Xiong X, Guo L. Phase-transited lysozyme coating on zirconia abutments for enhancing soft tissue seal and antibacterial activity. Int J Biol Macromol 2025; 307:142016. [PMID: 40086550 DOI: 10.1016/j.ijbiomac.2025.142016] [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: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Zirconia abutments lacking intrinsic antibacterial properties and exhibiting inadequate sealing at the zirconia abutments-soft tissue interface may cause peri-implantitis, ultimately leading to implant failure. Simultaneously improving the soft tissue seal while integrating the antibacterial properties of zirconia abutments presents significant challenges. A phase-transited lysozyme (PTL) coating was successfully constructed on zirconia abutments to enhance soft tissue attachment and antibacterial properties, aiming to safeguard peri-implant soft tissue from infection. The PTL coated zirconia abutments boosted protein synthesis and gene expression, and facilitated the proliferation and adhesion of fibroblasts and epithelial cells. The PTL coating exhibited significant antibacterial properties against the primary pathogens associated with peri-implantitis, specifically Staphylococcus aureus and Porphyromonas gingivalis. In a rabbit immediate implantation model, a firmer gingival junctional epithelium formed around PTL-coated zirconia abutments in comparison to unmodified abutments, thereby establishing a strong soft tissue barrier against pathogen invasion. The dual-function phase-transited lysozyme coating on zirconia abutments enhances soft tissue sealing and exhibits antibacterial properties, offering a practical approach to preventing peri-implantitis.
Collapse
Affiliation(s)
- Li Wang
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yang Zhou
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yunan Zhang
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Zhongke Wang
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Aodi Li
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Hongmei Chen
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Ying Huang
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Xinlei Xiong
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Ling Guo
- School of Stomatology, Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China; Department of Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
6
|
Yang Q, Miao Y, Luo J, Li C, Li X, Chen Y, Wang Y. Nanofibril-Structured Granular Hydrogels Harness Stem Cell Retention and Immunoregulation in Diabetic Microenvironment. ACS NANO 2025; 19:6795-6814. [PMID: 39932571 DOI: 10.1021/acsnano.4c11414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Granular hydrogel matrices have shown significant advantages in mesenchymal stem cell (MSC) delivery and tissue ingrowth due to their minimally invasive injection capabilities and porous structures. However, creating granular hydrogels that simultaneously mimic the nanofilamentous architecture of the natural extracellular matrix (ECM) and enhance stem cell retention and in vivo immunoregulation in a diabetic microenvironment remains challenging. In this study, we present a nanoengineered supramolecular granular hydrogel with a nanofibrillar structure designed to improve stem cell retention and regulate immune responses under diabetic conditions. The granular hydrogel matrix is assembled based on multiple hydrogen bonding and hydrophobic interactions, exhibiting a range of tunable features, including shear-thinning, injectability, self-healing, and 3D printability. Furthermore, enriched with manganese dioxide (MnO2)-amyloid fibril (AF) nanohybrids, the granular hydrogel supports MSC adhesion and stemness maintenance and can modulate the reactive oxygen species microenvironment by converting H2O2 into oxygen, thereby promoting cell viability and osteogenic differentiation of MSCs with the sustained release of Mn2+. In a two-week diabetic rat model study, the granular hydrogel demonstrates enhanced in vivo cell retention and anti-inflammatory immunomodulation properties, underscoring its potential as a promising matrix for stem cell therapy and immune regulation in diabetic conditions.
Collapse
Affiliation(s)
- Qian Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yali Miao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Jinshui Luo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Chunhui Li
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Xining Li
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
7
|
Kim H, Bae S, Kim SJ. Increased SNAI2 expression and defective collagen adhesion in cells with pediatric dementia, juvenile ceroid lipofuscinosis. Biochem Biophys Res Commun 2024; 738:150561. [PMID: 39154552 DOI: 10.1016/j.bbrc.2024.150561] [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: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Dementia-related neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), are known to be caused by accumulation of toxic proteins. However, the molecular mechanisms that cause neurodegeneration and its biophysical effects on cells remain unclear. In this study, we used juvenile neuronal ceroid lipofuscinosis (JNCL), a pediatric dementia with a clear etiology of mutations in ceroid lipofuscinosis neuronal 3 (CLN3), to explore the changes in cell adhesion, a biophysical process that regulates neuronal development and survival. We used JNCL cerebral organoid gene expression datasets to identify the biological pathways that affect neural development, and found enriched gene expression in the epithelial-mesenchymal transition (EMT) pathway and increased expression of its inducer snail family transcriptional repressor 2 (SNAI2). A cell adhesion assay using lymphoblasts from patients with JNCL revealed defective adhesion to cell culture plates, glass surfaces, collagen type I, and neuroblast-like cells. To determine whether inhibition of EMT could improve the cell adhesion of JNCL lymphoblasts, we used all-trans retinoic acid, a well-known EMT inhibitor and inducer of neural differentiation. In JNCL lymphoblasts, ATRA treatment enhanced adhesion to collagen type I and these effects were abolished by Ca2+ chelator. These results provide new insights into the role of CLN3 and cell adhesion in the pathogenesis of NDD.
Collapse
Affiliation(s)
- Hyungkuen Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Baebang, Asan, Chungnam, 31499, South Korea
| | - Sechul Bae
- Jung Cosmetic Corporation, Sinchang, Asan, Chungnam, 31537, South Korea
| | - Sung-Jo Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Baebang, Asan, Chungnam, 31499, South Korea.
| |
Collapse
|
8
|
Okyem S, Sweedler JV. Recent Advancements in the Characterization of D-Amino Acid and Isoaspartate Post-Translational Modifications. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39558451 DOI: 10.1002/mas.21916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024]
Abstract
One of the great triumphs of mass spectrometry-based peptide and protein characterization is the characterization of their modifications as most modifications have a characteristic mass shift. What happens when the modification does not change the mass of the peptide? Here, the characterization of several peptide and proteins modifications that do not involve a mass shift are highlighted. Protein and peptide synthesis on ribosomes involves L-amino acids; however, posttranslational modifications (PTMs) can convert these L-amino acids into their D-isomers. As another example, nonenzymatic PTM of aspartate leads to the formation of three different isomers, with isoaspartate being the most prevalent. Both modifications do not alter the mass of the peptide and yet can have profound impact on the physicochemical characteristics of the peptide. Several MS and ion mobility techniques are highlighted, as are other methods such as chromatography, enzymatic enrichment, and labeling. The challenges inherent to these analytical methods and prospective developments in bioinformatics and computational strategies are discussed for these zero-dalton PTMs.
Collapse
Affiliation(s)
- Samuel Okyem
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jonathan V Sweedler
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
9
|
Karanth S, Wiesenfarth M, Benthin J, Koehler M. Fava Bean Protein Nanofibrils Modulate Cell Membrane Interfaces for Biomolecular Interactions as Unveiled by Atomic Force Microscopy. Foods 2024; 13:3411. [PMID: 39517195 PMCID: PMC11545818 DOI: 10.3390/foods13213411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Functional amyloids (protein nanofibrils, PNF) synthesized from plant sources exhibit unique physicochemical and nanomechanical properties that could improve food texture. While environmental factors affecting PNFs are well-known, scientific evidence on how cells (focus on the oral cavity) respond to them under physiological conditions is lacking. Self-assembled PNFs synthesized from fava bean whole protein isolate show a strong pH- and solvent-dependent morphology and elasticity modification measured by atomic force microscopy (AFM). After incubation of PNFs with an oral mechanosensitive model cell line at pH 7.3, difference in cell-surface roughness without significant changes in the overall cell elasticity were measured. The role of cell membrane composition on supported lipid bilayers was also tested, showing an increase in membrane elasticity with increasing fibril concentration and the possible impact of annular phospholipids in binding. Genetic responses of membrane proteins involved in texture and fat perception were detected at the mRNA level by RT-qPCR assay and both mechano- and chemosensing proteins displayed responses highlighting an interface dependent interaction. The outcomes of this study provide a basis for understanding the changing physicochemical properties of PNFs and their effect on flavor perception by altering mouthfeel and fat properties. This knowledge is important in the development of plant-based texture enhancers for sensory-appealing foods that require consumer acceptance and further promote healthy diets.
Collapse
Affiliation(s)
- Sanjai Karanth
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; (S.K.); (M.W.); (J.B.)
| | - Marina Wiesenfarth
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; (S.K.); (M.W.); (J.B.)
- TUM Graduate School, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany
| | - Julia Benthin
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; (S.K.); (M.W.); (J.B.)
- TUM Graduate School, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany
| | - Melanie Koehler
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; (S.K.); (M.W.); (J.B.)
- Chair of Nutritional Systems Biology, TUM Junior Fellow, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
10
|
Zhang Y, Zhang X, Kai T, Zhang L, Li A. Lycium ruthenicum Murray derived exosome-like nanovesicles inhibit Aβ-induced apoptosis in PC12 cells via MAPK and PI3K/AKT signaling pathways. Int J Biol Macromol 2024; 277:134309. [PMID: 39089544 DOI: 10.1016/j.ijbiomac.2024.134309] [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: 05/07/2024] [Revised: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Plant-derived exosome-like nanovesicles (ELNs) are nano-sized vesicles extracted from edible plants. Lycium ruthenicum Murray (LRM) has been gaining increasing attention due to its nutritional and medicinal value, but the ELNs in LRM has not been reported. In this study, LRM-ELNs were obtained, and the proteins, lipids, microRNAs (miRNAs) and active components in LRM tissues and LRM-ELNs was analyzed by LC-MS/MS, LC-MS, high-throughput sequencing techniques, and physical and chemical analysis. LRM-ELNs can be uptaken by PC12 cells through macropinocytosis and caveolin-mediated endocytosis primarily. Transcriptomic and western blot experiments indicate that LRM-ELNs can inhibit Aβ-induced apoptosis in PC12 cells through the MAPK and PI3K/AKT signaling pathways, with miRNAs playing a crucial role. These results indicate that LRM-ELNs have the protection effect on PC12 cells and can be considered as dietary supplements for alleviating neurodegenerative diseases.
Collapse
Affiliation(s)
- Yadan Zhang
- College of Food Science and Engineering, Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha 410004, China
| | - Xiaoyu Zhang
- College of Food Science and Engineering, Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha 410004, China
| | - Tianhan Kai
- Xiang Ya School of Public Health, Central South University, Changsha 410078, China
| | - Lin Zhang
- College of Food Science and Engineering, Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha 410004, China.
| | - Anping Li
- College of Food Science and Engineering, Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha 410004, China.
| |
Collapse
|
11
|
Akbarian M, Kianpour M, Tayebi L. Fabricating Multiphasic Angiogenic Scaffolds Using Amyloid/Roxadustat-Assisted High-Temperature Protein Printing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36983-37006. [PMID: 38953207 DOI: 10.1021/acsami.4c06207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Repairing multiphasic defects is cumbersome. This study presents new soft and hard scaffold designs aimed at facilitating the regeneration of multiphasic defects by enhancing angiogenesis and improving cell attachment. Here, the nonimmunogenic, nontoxic, and cost-effective human serum albumin (HSA) fibril (HSA-F) was used to fabricate thermostable (up to 90 °C) and hard printable polymers. Additionally, using a 10.0 mg/mL HSA-F, an innovative hydrogel was synthesized in a mixture with 2.0% chitosan-conjugated arginine, which can gel in a cell-friendly and pH physiological environment (pH 7.4). The presence of HSA-F in both hard and soft scaffolds led to an increase in significant attachment of the scaffolds to the human periodontal ligament fibroblast (PDLF), human umbilical vein endothelial cell (HUVEC), and human osteoblast. Further studies showed that migration (up to 157%), proliferation (up to 400%), and metabolism (up to 210%) of these cells have also improved in the direction of tissue repair. By examining different in vitro and ex ovo experiments, we observed that the final multiphasic scaffold can increase blood vessel density in the process of per-vascularization as well as angiogenesis. By providing a coculture environment including PDLF and HUVEC, important cross-talk between these two cells prevails in the presence of roxadustat drug, a proangiogenic in this study. In vitro and ex ovo results demonstrated significant enhancements in the angiogenic response and cell attachment, indicating the effectiveness of the proposed design. This approach holds promise for the regeneration of complex tissue defects by providing a conducive environment for vascularization and cellular integration, thus promoting tissue healing.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Maryam Kianpour
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| |
Collapse
|
12
|
Peña-Díaz S, Olsen WP, Wang H, Otzen DE. Functional Amyloids: The Biomaterials of Tomorrow? ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312823. [PMID: 38308110 DOI: 10.1002/adma.202312823] [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: 11/28/2023] [Revised: 01/22/2024] [Indexed: 02/04/2024]
Abstract
Functional amyloid (FAs), particularly the bacterial proteins CsgA and FapC, have many useful properties as biomaterials: high stability, efficient, and controllable formation of a single type of amyloid, easy availability as extracellular material in bacterial biofilm and flexible engineering to introduce new properties. CsgA in particular has already demonstrated its worth in hydrogels for stable gastrointestinal colonization and regenerative tissue engineering, cell-specific drug release, water-purification filters, and different biosensors. It also holds promise as catalytic amyloid; existing weak and unspecific activity can undoubtedly be improved by targeted engineering and benefit from the repetitive display of active sites on a surface. Unfortunately, FapC remains largely unexplored and no application is described so far. Since FapC shares many common features with CsgA, this opens the window to its development as a functional scaffold. The multiple imperfect repeats in CsgA and FapC form a platform to introduce novel properties, e.g., in connecting linkers of variable lengths. While exploitation of this potential is still at an early stage, particularly for FapC, a thorough understanding of their molecular properties will pave the way for multifunctional fibrils which can contribute toward solving many different societal challenges, ranging from CO2 fixation to hydrolysis of plastic nanoparticles.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, DK - 8000, Denmark
| | - William Pallisgaard Olsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, DK - 8000, Denmark
| | - Huabing Wang
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, DK - 8000, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, Aarhus C, 8000, Denmark
| |
Collapse
|
13
|
Ivanova M, Belaya I, Kucháriková N, de Sousa Maciel I, Saveleva L, Alatalo A, Juvonen I, Thind N, Andrès C, Lampinen R, Chew S, Kanninen KM. Upregulation of Integrin beta-3 in astrocytes upon Alzheimer's disease progression in the 5xFAD mouse model. Neurobiol Dis 2024; 191:106410. [PMID: 38220131 DOI: 10.1016/j.nbd.2024.106410] [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: 07/14/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024] Open
Abstract
Integrins are receptors that have been linked to various brain disorders, including Alzheimer's disease (AD), the most prevalent neurodegenerative disorder. While Integrin beta-3 (ITGB3) is known to participate in multiple cellular processes such as adhesion, migration, and signaling, its specific role in AD remains poorly understood, particularly in astrocytes, the main glial cell type in the brain. In this study, we investigated alterations in ITGB3 gene and protein expression during aging in different brain regions of the 5xFAD mouse model of AD and assessed the interplay between ITGB3 and astrocytes. Primary cultures from adult mouse brains were used to gain further insight into the connection between ITGB3 and amyloid beta (Aβ) in astrocytes. In vivo studies showed a correlation between ITGB3 and the astrocytic marker GFAP in the 5xFAD brains, indicating its association with reactive astrocytes. In vitro studies revealed increased gene expression of ITGB3 upon Aβ treatment. Our findings underscore the potential significance of ITGB3 in astrocyte reactivity in the context of Alzheimer's disease.
Collapse
Affiliation(s)
- Mariia Ivanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Irina Belaya
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nina Kucháriková
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Izaque de Sousa Maciel
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arto Alatalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilona Juvonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Navjot Thind
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Clarisse Andrès
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riikka Lampinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sweelin Chew
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
14
|
Choi J, Seo S, Lee S, Ko H, Luo Y, Han Y, Shin JH, Cho H, Lee H. Low-Temperature Layer-by-Layer Growth of Semiconducting Few-Layer γ-Graphyne to Exploit Robust Biocompatibility. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41708-41719. [PMID: 37621110 DOI: 10.1021/acsami.3c08446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
The sp-hybridized carbon network in single- or few-layer γ-graphyne (γ-GY) has a polarized electron distribution, which can be crucial in overcoming biosafety issues. Here, we report the low-temperature synthesis, electronic properties, and amyloid fibril nanostructures of electrostatic few-layer γ-GY. ABC stacked γ-GY is synthesized by layer-by-layer growth on a catalytic copper surface, exhibiting intrinsic p-type semiconducting properties in few-layer γ-GY. Thickness-dependent electronic properties of γ-GY elucidate interlayer interactions by electron doping between electrostatic layers and layer stacking-involved modulation of the band gap. Electrostatic few-layer γ-GY induces high electronic sensitivity and intense interaction with amyloid beta (i.e., Aβ40) peptides assembling into elongated mature Aβ40 fibrils. Two-dimensional biocompatible nanostructures of Aβ40 fibrils/few-layer γ-GY enable excellent cell viability and high neuronal differentiation of living cells without external stimulation.
Collapse
Affiliation(s)
- Jungsue Choi
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Centre for Integrated Nanostructure Physics, Institute of Basic Science, Suwon 16419, Republic of Korea
| | - Sohyeon Seo
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Creative Research Institute, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seungeun Lee
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyun Ko
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yongguang Luo
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Centre for Integrated Nanostructure Physics, Institute of Basic Science, Suwon 16419, Republic of Korea
| | - Yeonsu Han
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hee Shin
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hansang Cho
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyoyoung Lee
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Centre for Integrated Nanostructure Physics, Institute of Basic Science, Suwon 16419, Republic of Korea
- Creative Research Institute, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
15
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
16
|
Victorelli FD, Rodero CF, Lutz‐Bueno V, Chorilli M, Mezzenga R. Amyloid Fibrils Enhance the Topical Bio-Adhesivity of Liquid Crystalline Mesophase-Based Drug Formulations. Adv Healthc Mater 2023; 12:e2202720. [PMID: 36681654 PMCID: PMC11468793 DOI: 10.1002/adhm.202202720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/08/2023] [Indexed: 01/23/2023]
Abstract
Despite their distinctive secondary structure based on cross β-strands, amyloid fibrils (AF) are stable fibrous protein aggregates with features similar to collagen, one of the main components of the extracellular matrix, and thus constitute a potential scaffold for enhancing cell adhesion for topical applications. Here, the contribution of AF to skin bio-adhesivity aiming toward topical treatments is investigated. Liquid crystalline mesophase (LCM) based on phytantriol is formulated, with the aqueous phase containing either water or a solution of 4 wt% amyloid fibrils. Then resveratrol is added as a model anti-inflammatory molecule. The developed LCM presents a double gyroid Ia3d mesophase. The incorporation of AF into the LCM increases its bio-adhesive properties. In vitro release and ex vivo permeation and retention confirm the controlled release property of the system, and that resveratrol is retained in epidermis and dermis, but is also permeated through the skin. All formulations are biocompatible with L929 cells. The in vivo assay confirms that systems with AF lead to a higher anti-inflammatory effect of resveratrol. These results confirm the hypothesis that the incorporation of AF in the LCM increases the bio-adhesiveness and efficiency of the system for topical treatment, and consequently, the therapeutical action of the encapsulated drug.
Collapse
Affiliation(s)
| | - Camila Fernanda Rodero
- Department of Drugs and MedicineSchool of Pharmaceutical SciencesSão Paulo State UniversityAraraquaraSão Paulo14800‐903Brazil
| | | | - Marlus Chorilli
- Department of Drugs and MedicineSchool of Pharmaceutical SciencesSão Paulo State UniversityAraraquaraSão Paulo14800‐903Brazil
| | - Raffaele Mezzenga
- Department of Health Sciences & TechnologyETH ZurichZurich8092Switzerland
- Department of MaterialsETH ZurichZurich8093Switzerland
| |
Collapse
|
17
|
Singh N, Patel K, Navalkar A, Kadu P, Datta D, Chatterjee D, Mukherjee S, Shaw R, Gahlot N, Shaw A, Jadhav S, Maji SK. Amyloid fibril-based thixotropic hydrogels for modeling of tumor spheroids in vitro. Biomaterials 2023; 295:122032. [PMID: 36791521 DOI: 10.1016/j.biomaterials.2023.122032] [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: 04/28/2022] [Revised: 12/28/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Biomaterials mimicking extracellular matrices (ECM) for three-dimensional (3D) cultures have gained immense interest in tumor modeling and in vitro organ development. Here, we introduce a new class of amyloid fibril-based peptide hydrogels as a versatile biomimetic ECM scaffold for 3D cell culture and homogenous tumor spheroid modeling. We show that these amyloid fibril-based hydrogels are thixotropic and allow cancer cell adhesion, proliferation, and migration. All seven designed hydrogels support 3D cell culture with five different cancer cell lines forming spheroid with necrotic core and upregulation of the cancer biomarkers. We further developed the homogenous, single spheroid using the drop cast method and the data suggest that all hydrogels support the tumor spheroid formation but with different necrotic core diameters. The detailed gene expression analysis of MCF7 spheroid by microarray suggested the involvement of pro-oncogenes and significant regulatory pathways responsible for tumor spheroid formation. Further, using breast tumor tissue from a mouse xenograft model, we show that selected amyloid hydrogels support the formation of tumor spheroids with a well-defined necrotic core, cancer-associated gene expression, higher drug resistance, and tumor heterogeneity reminiscent of the original tumor. Altogether, we have developed an easy-to-use, rapid, cost-effective, and scalable platform for generating in vitro cancer models for the screening of anti-cancer therapeutics and developing personalized medicine.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Debalina Datta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Debdeep Chatterjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Ranjit Shaw
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Abhishek Shaw
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | | | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
18
|
Zhao J, Zhou Y, Yan J, Liu J, Wang L, Zhang X, Lou Y, Que K. Effects of phase-transited lysozyme on adhesion, migration and odontogenic differentiation of human dental pulp cells: An in vitro study. Int Endod J 2023; 56:475-485. [PMID: 36565046 DOI: 10.1111/iej.13884] [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: 08/27/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
AIM To explore the effects of phase-transited lysozyme (PTL) coated dentine slices on cell adhesion, migration and odontogenic differentiation of human dental pulp cells (HDPCs). METHODOLOGY Cell growth and cell cycle analysis were conducted to verify the biocompatibility of PTL for HDPCs. Cell adhesion, cell morphology and proliferation were explored by DiI staining, Scanning electron microscopy and MTT assay. Cell migration was investigated by Transwell assay. The effects of PTL on odontogenesis and mineralization of HDPCs were assessed by real-time quantitative polymerase chain reaction and Western blot. The mineralization of HDPCs was evaluated by Alizarin red staining. HDPCs were isolated from extracted third molars. The level of statistically significant difference was accepted at p < .05. RESULTS PTL showed no negative effect on cell cycle of HDPCs and compared with the blank group, HDPCs labelled with DiI staining showed significantly more adhered cells at 48 h (p < .05), extending cell processes and more finger-like or reticular pseudopodia on PTL-coated dentine slices. The results of MTT and Transwell assay showed that PTL promoted the proliferation (p < .05) and migration (p < .01) of HDPCs, respectively. Compared with the blank group, the gene expression of dentine sialophosphoprotein (DSPP), osteopontin and bone sialoprotein in HDPCs cultured on PTL was significantly upregulated on day 3 and 7 (p < .05), while the protein expression of DSPP showed no significant change on both day 7 and day 14. Alizarin red staining showed that PTL promoted more mineralization nodules formation of HDPCs (p < .05). CONCLUSIONS PTL promoted the adhesion, proliferation and migration of HDPCs on dentine slices, and positively affected odontogenic differentiation and mineralization of HDPCs.
Collapse
Affiliation(s)
- Jiange Zhao
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Yunjie Zhou
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Jinjie Yan
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Jie Liu
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Linxian Wang
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Xu Zhang
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Yaxin Lou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Kehua Que
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
19
|
Abstract
Amyloid fibrils may serve as building blocks for the preparation of novel hydrogel materials from abundant, low-cost, and biocompatible polypeptides. This work presents the formation of physically cross-linked, self-healing hydrogels based on bovine serum albumin at room temperature through a straightforward disulfide reduction step induced by tris (2-carboxyethyl) phosphine hydrochloride. The structure and surface charge of the amyloid-like fibrils is determined by the pH of the solution during self-assembly, giving rise to hydrogels with distinct physicochemical properties. The hydrogel surface can be readily functionalized with the extracellular matrix protein fibronectin and supports cell adhesion, spreading, and long-term culture. This study offers a simple, versatile, and inexpensive method to prepare amyloid-based albumin hydrogels with potential applications in the biomedical field.
Collapse
Affiliation(s)
- Carolina Diaz
- Department of Cellular BiophysicsMax‐Planck‐Institute for Medical ResearchJahnstr. 2969120HeidelbergGermany
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA)Facultad de Ciencias ExactasUNLP – CONICETCC16 Suc 4 (1900)La PlataBuenos Aires1900Argentina
| | - Dimitris Missirlis
- Department of Cellular BiophysicsMax‐Planck‐Institute for Medical ResearchJahnstr. 2969120HeidelbergGermany
| |
Collapse
|
20
|
Wang WR, Li J, Gu JT, Hu BW, Qin W, Zhu YN, Guo ZX, Ma YX, Tay F, Jiao K, Niu L. Optimization of Lactoferrin-Derived Amyloid Coating for Enhancing Soft Tissue Seal and Antibacterial Activity of Titanium Implants. Adv Healthc Mater 2023; 12:e2203086. [PMID: 36594680 DOI: 10.1002/adhm.202203086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Indexed: 01/04/2023]
Abstract
A poor seal of the titanium implant-soft tissue interface provokes bacterial invasion, aggravates inflammation, and ultimately results in implant failure. To ensure the long-term success of titanium implants, lactoferrin-derived amyloid is coated on the titanium surface to increase the expression of cell integrins and hemidesmosomes, with the goal of promoting soft tissue seal and imparting antibacterial activity to the implants. The lactoferrin-derived amyloid coated titanium structures contain a large number of amino and carboxyl groups on their surfaces, and promote proliferation and adhesion of epithelial cells and fibroblasts via the PI3K/AKT pathway. The amyloid coating also has a strong positive charge and possesses potent antibacterial activities against Staphylococcus aureus and Porphyromonas gingivalis. In a rat immediate implantation model, the amyloid-coated titanium implants form gingival junctional epithelium at the transmucosal region that resembles the junctional epithelium in natural teeth. This provides a strong soft tissue seal to wall off infection. Taken together, lactoferrin-derived amyloid is a dual-function transparent coating that promotes soft tissue seal and possesses antibacterial activity. These unique properties enable the synthesized amyloid to be used as potential biological implant coatings.
Collapse
Affiliation(s)
- Wan-Rong Wang
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Jing Li
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Jun-Ting Gu
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Bo-Wen Hu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, P. R. China
| | - Wen Qin
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Yi-Na Zhu
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Zhen-Xing Guo
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Yu-Xuan Ma
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Franklin Tay
- Department of Endodontics, the Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Lina Niu
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
21
|
Kadu P, Gadhe L, Navalkar A, Patel K, Kumar R, Sastry M, Maji SK. Charge and hydrophobicity of amyloidogenic protein/peptide templates regulate the growth and morphology of gold nanoparticles. NANOSCALE 2022; 14:15021-15033. [PMID: 36194184 DOI: 10.1039/d2nr01942f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Biomolecules are known to interact with metals and produce nanostructured hybrid materials with diverse morphologies and functions. In spite of the great advancement in the principles of biomimetics for designing complex nano-bio structures, the interplay between the physical properties of biomolecules such as sequence, charge, and hydrophobicity with predictable morphology of the resulting nanomaterials is largely unknown. Here, using various amyloidogenic proteins/peptides and their corresponding fibrils in combination with different pH, we show defined principle for gold nanocrystal growth into triangular and supra-spheres with high prediction. Using a combination of different biophysical and structural techniques, we establish the mechanism of nucleation and crystal growth of gold nanostructures and show the effective isolation of intact nanostructures from amyloid templates using protein digestion. This study will significantly advance our design principle for bioinspired materials for specific functions with great predictability.
Collapse
Affiliation(s)
- Pradeep Kadu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Komal Patel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Murali Sastry
- Department of Materials Science and Engineering & Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
22
|
Jia X, Song J, Lv W, Hill JP, Nakanishi J, Ariga K. Adaptive liquid interfaces induce neuronal differentiation of mesenchymal stem cells through lipid raft assembly. Nat Commun 2022; 13:3110. [PMID: 35661107 PMCID: PMC9166733 DOI: 10.1038/s41467-022-30622-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 05/06/2022] [Indexed: 01/02/2023] Open
Abstract
Stem cells and their microenvironment interact cooperatively to dictate their fates. Biomaterials are dynamically remodeled by stem cells, and stem cells sense and translate the changes into cell fate decisions. We have previously reported that adaptive biomaterials composed of fibronectin inserted into protein nanosheets at a liquid interface enhance neuronal differentiation of human mesenchymal stem cells (hMSCs). However, we could not decouple clearly the effect of ligand density from that of fibrillary structure on cellular function and fate. Here we present an adaptive biomaterial based on two-dimensional networks of protein nanofibrils at a liquid–liquid interface. Compared with flat protein nanosheets, this biomaterial enhances neuronal differentiation of hMSCs through a signaling mechanism involving focal adhesion kinase. Lipid raft microdomains in plasma membrane are found to play a central role in which hMSCs rapidly adapt to the dynamic microenvironment at the fluid interface. Our finding has substantial implications for regenerative medicine and tissue engineering. In this work the authors report how human mesenchymal stem cells rapidly adapt to dynamic microenvironment through lipid raft in membrane microdomains that direct neurogenesis.
Collapse
Affiliation(s)
- Xiaofang Jia
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Jingwen Song
- International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.,Department of Advanced Materials Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan
| | - Wenyan Lv
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jonathan P Hill
- International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Jun Nakanishi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Katsuhiko Ariga
- International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan. .,Department of Advanced Materials Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan.
| |
Collapse
|
23
|
Kumari M, Sharma S, Deep S. Tetrabutylammonium based ionic liquids (ILs) inhibit the amyloid aggregation of superoxide dismutase 1 (SOD1). J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.118761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
24
|
Li Y, Fu Y, Zhang H, Wang X, Chen T, Wu Y, Xu X, Yang S, Ji P, Song J. Natural Plant Tissue with Bioinspired Nano Amyloid and Hydroxyapatite as Green Scaffolds for Bone Regeneration. Adv Healthc Mater 2022; 11:e2102807. [PMID: 35285169 DOI: 10.1002/adhm.202102807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Indexed: 12/17/2022]
Abstract
Bone defects have been increasingly prevalent around the globe and traditional bone substitutes are constantly limited by low abundance and biosafety due to their animal-based resources. Plant-based scaffolds are currently studied as a green candidate but the bioinertia of cellulose to mammalian cells leads to uncertain bone regeneration. Inspired by the cross-kingdom adhesion of plants and bacteria, this work proposes a concept of a novel plant bone substitute, involving coating decellularized plant with nano amyloids and nano hydroxyapatites, to bridge the plant scaffold and animal tissue regeneration. Natural microporosity of plants can guide alignment of mammalian cells into various organ-like structures. Taking advantage of the bioactive nano amyloids, the scaffolds drastically promote cell adhesion, viability, and proliferation. The enhanced bio-affinity is elucidated as positively charged nano amyloids and serum deposition on the nanostructure. Nano-hydroxyapatite crystals deposited on amyloid further prompt osteogenic differentiation of pre-osteoblasts. In vivo experiments prove successful trabeculae regeneration in the scaffold. Such a hierarchical design leverages the dedicated microstructure of natural plants and high bioactivity of nano amyloid/hydroxyapatite coatings, and addresses the abundant resource of bone substitutes. Not limited to their current application, plant materials functionalized with nano amyloid/hydroxyapatite coatings allow many cross-kingdom tissue engineering and biomedical applications.
Collapse
Affiliation(s)
- Yuzhou Li
- Stomatological Hospital of Chongqing Medical University Chongqing 401147 P.R. China
| | - Yiru Fu
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education Chongqing 401147 P.R. China
| | - He Zhang
- Stomatological Hospital of Chongqing Medical University Chongqing 401147 P.R. China
| | - Xu Wang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences Chongqing 401147 P.R. China
| | - Tao Chen
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences Chongqing 401147 P.R. China
| | - Yanqiu Wu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences Chongqing 401147 P.R. China
| | - Xinxin Xu
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education Chongqing 401147 P.R. China
| | - Sheng Yang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences Chongqing 401147 P.R. China
| | - Ping Ji
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education Chongqing 401147 P.R. China
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University Chongqing 401147 P.R. China
| |
Collapse
|
25
|
Kadirvelu K, Fathima NN. Deciphering Mechanism of Assembly of Keratin within Nanofibrous Matrix: Expanding the Horizon of Electrospun Polymer/Protein Composites. ChemistrySelect 2021. [DOI: 10.1002/slct.202103018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Kavitha Kadirvelu
- Inorganic and Physical Chemistry Laboratory CSIR- Central Leather Research Institute Chennai 600020 Tamil Nadu India
| | - Nishter Nishad Fathima
- Inorganic and Physical Chemistry Laboratory CSIR- Central Leather Research Institute Chennai 600020 Tamil Nadu India
| |
Collapse
|
26
|
Singh G, Kaur M, Singh M, Kaur H, Kang TS. Spontaneous Fibrillation of Bovine Serum Albumin at Physiological Temperatures Promoted by Hydrolysis-Prone Ionic Liquids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:10319-10329. [PMID: 34407374 DOI: 10.1021/acs.langmuir.1c01350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
This study highlights the role of time-dependent hydrolysis of ionic liquid anion, [BF4]-, of ionic liquid (IL), 1-ethyl-3-methylimidazolium tetrafluoroborate, [C2mim][BF4], which results in ever-changing pH conditions. Such pH changes along with the ionic interactions bring conformational changes in bovine serum albumin (BSA), leading to the formation of amyloid fibers at 37 °C without external control of pH or addition of electrolyte. The fibrillation of BSA occurs spontaneously with the addition of IL; however, the highest growth rate has been observed in aqueous solution of 10% IL (v/v %) among investigated systems. Thioflavin T (ThT) fluorescence emission has been employed to monitor the growth and development of β-sheet content in amyloid fibrils. The structural alterations in BSA have also been investigated using intrinsic fluorescence measurements. Circular dichroism (CD) measurements confirmed the formation of amyloid fibrils. Transmission electron microscopy (TEM) has been explored to establish the morphologies of BSA fibrils at different intervals of time, whereas atomic force microscopy (AFM) has established the helically twisted nature of grown amyloid fibrils. The docking studies have been utilized to understand the insertion of IL ions in different domains of BSA, which along with decreased pH cause the unfolding and growth of BSA into amyloid fibrils. It is expected that the results obtained from this study would help to understand the impact of IL containing [BF4]- anion on protein stability and aggregation along with providing a new platform to control the formation of amyloid fibrils and other biomaterials driven via ionic interactions and alterations in pH.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Chemistry, UGC-Centre for Advance Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| | - Manvir Kaur
- Department of Chemistry, UGC-Centre for Advance Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| | - Manpreet Singh
- Department of Chemistry, UGC-Centre for Advance Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| | - Harmandeep Kaur
- Department of Chemistry, UGC-Centre for Advance Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| | - Tejwant Singh Kang
- Department of Chemistry, UGC-Centre for Advance Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| |
Collapse
|
27
|
Noble Jesus C, Evans R, Forth J, Estarellas C, Gervasio FL, Battaglia G. Amphiphilic Histidine-Based Oligopeptides Exhibit pH-Reversible Fibril Formation. ACS Macro Lett 2021; 10:984-989. [PMID: 34422455 PMCID: PMC8375021 DOI: 10.1021/acsmacrolett.1c00142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022]
Abstract
![]()
We report the design,
simulation, synthesis, and reversible self-assembly
of nanofibrils using polyhistidine-based oligopeptides. The inclusion
of aromatic amino acids in the histidine block produces distinct antiparallel
β-strands that lead to the formation of amyloid-like fibrils.
The structures undergo self-assembly in response to a change in pH.
This creates the potential to produce well-defined fibrils for biotechnological
and biomedical applications that are pH-responsive in a physiologically
relevant range.
Collapse
Affiliation(s)
- Carlos Noble Jesus
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Institute for the Physics of the Living System, University College London, London WC1E 6BT, United Kingdom
| | - Rhys Evans
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Joe Forth
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Institute for the Physics of the Living System, University College London, London WC1E 6BT, United Kingdom
| | - Carolina Estarellas
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Francesco Luigi Gervasio
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Giuseppe Battaglia
- Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Institute for the Physics of the Living System, University College London, London WC1E 6BT, United Kingdom
- Institute for Bioengineering for Catalonia, The Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
28
|
Xuan Q, Wang Y, Chen C, Wang P. Rational Biological Interface Engineering: Amyloidal Supramolecular Microstructure-Inspired Hydrogel. Front Bioeng Biotechnol 2021; 9:718883. [PMID: 34350165 PMCID: PMC8327773 DOI: 10.3389/fbioe.2021.718883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 01/22/2023] Open
Abstract
Amyloidal proteins, which are prone to form fibrillar and ordered aggregates in vivo and in vitro, underlie the mechanism for neurodegenerative disorders and also play essential functions in the process of life. Amyloid fibrils typically adopt a distinctive β-sheet structure, which renders them with inherent extracellular matrix (ECM)-mimicking properties, such as powerful mechanical strength, promising adhesion, and antibacterial activity. Additionally, amyloidal proteins are a category of programmable self-assembled macromolecules, and their assembly and consequent nanostructure can be manipulated rationally. The above advantages motivate researchers to investigate the potential of amyloidal proteins as a novel type of hydrogel material. Currently, the amyloid-inspired hydrogel has become an emerging area and has been widely applied in a variety of biomedical fields, such as tissue repair, cell scaffolds, and drug delivery. In this review, we focus on the discussion of molecular mechanisms underlying the hydrogenation of amyloidal proteins, and introduce the advances achieved in biomedical applications of amyloid-inspired hydrogels.
Collapse
Affiliation(s)
- Qize Xuan
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Yibing Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Chao Chen
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Ping Wang
- Department of Bioproducts and Biosystems Engineering, University of Minnesota, Saint Paul, MN, United States
| |
Collapse
|
29
|
Suveges S, Chamseddine I, Rejniak KA, Eftimie R, Trucu D. Collective Cell Migration in a Fibrous Environment: A Hybrid Multiscale Modelling Approach. FRONTIERS IN APPLIED MATHEMATICS AND STATISTICS 2021; 7:680029. [PMID: 34322539 PMCID: PMC8315487 DOI: 10.3389/fams.2021.680029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The specific structure of the extracellular matrix (ECM), and in particular the density and orientation of collagen fibres, plays an important role in the evolution of solid cancers. While many experimental studies discussed the role of ECM in individual and collective cell migration, there are still unanswered questions about the impact of nonlocal cell sensing of other cells on the overall shape of tumour aggregation and its migration type. There are also unanswered questions about the migration and spread of tumour that arises at the boundary between different tissues with different collagen fibre orientations. To address these questions, in this study we develop a hybrid multi-scale model that considers the cells as individual entities and ECM as a continuous field. The numerical simulations obtained through this model match experimental observations, confirming that tumour aggregations are not moving if the ECM fibres are distributed randomly, and they only move when the ECM fibres are highly aligned. Moreover, the stationary tumour aggregations can have circular shapes or irregular shapes (with finger-like protrusions), while the moving tumour aggregations have elongate shapes (resembling to clusters, strands or files). We also show that the cell sensing radius impacts tumour shape only when there is a low ratio of fibre to non-fibre ECM components. Finally, we investigate the impact of different ECM fibre orientations corresponding to different tissues, on the overall tumour invasion of these neighbouring tissues.
Collapse
Affiliation(s)
| | - Ibrahim Chamseddine
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa Florida, USA
| | - Katarzyna A. Rejniak
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa Florida, USA
| | - Raluca Eftimie
- Laboratoire Mathématiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comté, 16 Route de Gray, Besançon, France
| | - Dumitru Trucu
- Department of Mathematics, University of Dundee, Dundee, UK
| |
Collapse
|
30
|
Aavani F, Biazar E, Heshmatipour Z, Arabameri N, Kamalvand M, Nazbar A. Applications of bacteria and their derived biomaterials for repair and tissue regeneration. Regen Med 2021; 16:581-605. [PMID: 34030458 DOI: 10.2217/rme-2020-0116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microorganisms such as bacteria and their derived biopolymers can be used in biomaterials and tissue regeneration. Various methods have been applied to regenerate damaged tissues, but using probiotics and biomaterials derived from bacteria with improved economic-production efficiency and highly applicable properties can be a new solution in tissue regeneration. Bacteria can synthesize numerous types of biopolymers. These biopolymers possess many desirable properties such as biocompatibility and biodegradability, making them good candidates for tissue regeneration. Here, we reviewed different types of bacterial-derived biopolymers and highlight their applications for tissue regeneration.
Collapse
Affiliation(s)
- Farzaneh Aavani
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), 15916-34311 Tehran, Iran
| | - Esmaeil Biazar
- Department of Biomedical Engineering, Tissue Engineering Group, Tonekabon Branch, Islamic Azad University, 46841-61167 Tonekabon, Iran
| | - Zoheir Heshmatipour
- Department of Microbiology, Tonekabon Branch, Islamic Azad University, 46841-61167 Tonekabon, Iran
| | - Nasibeh Arabameri
- Department of Microbiology, Tonekabon Branch, Islamic Azad University, 46841-61167 Tonekabon, Iran
| | - Mahshad Kamalvand
- Department of Biomedical Engineering, Tissue Engineering Group, Tonekabon Branch, Islamic Azad University, 46841-61167 Tonekabon, Iran
| | - Abolfazl Nazbar
- National Cell Bank, Pasteur Institute of Iran, 13169-43551 Tehran, Iran
| |
Collapse
|
31
|
Jurado R, Gálvez N. Apoferritin Amyloid-Fibril Directed the In Situ Assembly and/or Synthesis of Optical and Magnetic Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:E146. [PMID: 33435618 PMCID: PMC7826742 DOI: 10.3390/nano11010146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 12/25/2022]
Abstract
The coupling of proteins that can assemble, recognise or mineralise specific inorganic species is a promising strategy for the synthesis of nanoscale materials with a controllable morphology and functionality. Herein, we report that apoferritin protein amyloid fibrils (APO) have the ability to assemble and/or synthesise various metal and metal compound nanoparticles (NPs). As such, we prepared metal NP-protein hybrid bioconjugates with improved optical and magnetic properties by coupling diverse gold (AuNPs) and magnetic iron oxide nanoparticles (MNPs) to apoferritin amyloid fibrils and compared them to the well-known β-lactoglobulin (BLG) protein. In a second approach, we used of solvent-exposed metal-binding residues in APO amyloid fibrils as nanoreactors for the in situ synthesis of gold, silver (AgNPs) and palladium nanoparticles (PdNPs). Our results demonstrate, the versatile nature of the APO biotemplate and its high potential for preparing functional hybrid bionanomaterials. Specifically, the use of apoferritin fibrils as vectors to integrate magnetic MNPs or AuNPs is a promising synthetic strategy for the preparation of specific contrast agents for early in vivo detection using various bioimaging techniques.
Collapse
Affiliation(s)
| | - Natividad Gálvez
- Department of Inorganic Chemistry, University of Granada, 18071 Granada, Spain;
| |
Collapse
|
32
|
Khanna S, Singh AK, Behera SP, Gupta S. Thermoresponsive BSA hydrogels with phase tunability. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111590. [PMID: 33321635 DOI: 10.1016/j.msec.2020.111590] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/26/2022]
Abstract
Amyloids are fibrillar structures formed due to protein aggregation or misfolding when the molecules undergo a conformational change from α-helix to β-sheet. Although this self-assembly is associated with many neurodegenerative diseases in vivo, the highly ordered amyloidic structures formed in vitro are ideal scaffolds for many bionanotechnological applications. Amyloid fibrillar networks under specific stimuli can also form stable hydrogels. We have used bovine serum albumin (BSA) as a model amyloidogenic protein to obtain thermally-induced hydrogels that display tunable sol-gel-sol transitions spanning over minutes to days. High concentrations of BSA (14-22% w/v) were heated at 65 °C for less than 3 min without any cross-linking agent to yield soft, injectable gels that were non-toxic to mammalian cells. A detailed investigation of temperature, concentration, incubation time and ionic strength on the formation and reversal of these gels was carried out using visual inspection, rheology, electron microscopy, fluorescence spectroscopy, UV-visible spectroscopy and circular dichroism spectroscopy. The optimum gelation temperature (Tg) for phase reversal of BSA gels was found to lie between 60 and 70 °C. An increase in protein concentration led to a reduction in the gelation time and increase in the gel-to-rev sol transition time. Gels heated for longer duration than their minimum gelation time yielded irreversible gels suggesting that low incubation periods were favourable for partial protein denaturation and hydrogel formation. This was supported by time-resolved secondary and tertiary structural ensemble studies. Further, the hydrogel networks demonstrated a zero-order drug release kinetics and the rev sol was found to be cytocompatible with HaCaT skin cell lines. Overall, our approach demonstrates rapid, crosslinker-free thermoresponsive BSA gelation with wide tunability and control on the time and material property, ideal for topical drug delivery applications.
Collapse
Affiliation(s)
- Shruti Khanna
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Ajay Kumar Singh
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Soumya Prakash Behera
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shalini Gupta
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
33
|
Directionality of Macrophages Movement in Tumour Invasion: A Multiscale Moving-Boundary Approach. Bull Math Biol 2020; 82:148. [PMID: 33211193 PMCID: PMC7677171 DOI: 10.1007/s11538-020-00819-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Invasion of the surrounding tissue is one of the recognised hallmarks of cancer (Hanahan and Weinberg in Cell 100: 57–70, 2000. 10.1016/S0092-8674(00)81683-9), which is accomplished through a complex heterotypic multiscale dynamics involving tissue-scale random and directed movement of the population of both cancer cells and other accompanying cells (including here, the family of tumour-associated macrophages) as well as the emerging cell-scale activity of both the matrix-degrading enzymes and the rearrangement of the cell-scale constituents of the extracellular matrix (ECM) fibres. The involved processes include not only the presence of cell proliferation and cell adhesion (to other cells and to the extracellular matrix), but also the secretion of matrix-degrading enzymes. This is as a result of cancer cells as well as macrophages, which are one of the most abundant types of immune cells in the tumour micro-environment. In large tumours, these tumour-associated macrophages (TAMs) have a tumour-promoting phenotype, contributing to tumour proliferation and spread. In this paper, we extend a previous multiscale moving-boundary mathematical model for cancer invasion, by considering also the multiscale effects of TAMs, with special focus on the influence that their directional movement exerts on the overall tumour progression. Numerical investigation of this new model shows the importance of the interactions between pro-tumour TAMs and the fibrous ECM, highlighting the impact of the fibres on the spatial structure of solid tumour.
Collapse
|
34
|
Zhao X, Wang L, Gao J, Chen X, Wang K. Hyaluronic acid/lysozyme self-assembled coacervate to promote cutaneous wound healing. Biomater Sci 2020; 8:1702-1710. [PMID: 31994544 DOI: 10.1039/c9bm01886g] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Traditional hydrogel dressings are limited in practical applications due to the complexity of the preparation and low biocompatibility. So, there is an urgent need to design wound dressing with simple preparation method, higher biocompatibility, and superior therapeutic effect. Additionally, using a polysaccharide/protein mixture system is an attractive method to prepare the gel. In this study, a simple mixture of hyaluronic acid/lysozyme (HL) was used to obtain the HL coacervate gel. HL coacervate has suitable viscoelasticity and excellent adhesion on the skin tissue. We demonstrated its highly efficient self-healing property to overcome fracture or deformation. HL coacervate showed a significant effect on promoting wound healing in a full-thickness skin defect model. Compared to the commercial 3M dressing, it has faster epithelial tissue regeneration and stronger collagen deposition. In addition, cytotoxicity and organ toxicity tests indicated its high safety. In summary, HL coacervate has broad clinical application prospects as a wound dressing material.
Collapse
Affiliation(s)
- Xiaoye Zhao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jushan Gao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xi Chen
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, China
| | - Ke Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
35
|
Curcumin Inhibits the Primary Nucleation of Amyloid-Beta Peptide: A Molecular Dynamics Study. Biomolecules 2020; 10:biom10091323. [PMID: 32942739 PMCID: PMC7563689 DOI: 10.3390/biom10091323] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/07/2020] [Accepted: 09/12/2020] [Indexed: 12/11/2022] Open
Abstract
The amyloid plaques are a key hallmark of neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. Amyloidogenesis is a complex long-lasting multiphase process starting with the formation of nuclei of amyloid peptides: a process assigned as a primary nucleation. Curcumin (CU) is a well-known inhibitor of the aggregation of amyloid-beta (Aβ) peptides. Even more, CU is able to disintegrate preformed Aβ firbils and amyloid plaques. Here, we simulate by molecular dynamics the primary nucleation process of 12 Aβ peptides and investigate the effects of CU on the process. We found that CU molecules intercalate among the Aβ chains and bind tightly to them by hydrogen bonds, hydrophobic, π–π, and cation–π interactions. In the presence of CU, the Aβ peptides form a primary nucleus of a bigger size. The peptide chains in the nucleus become less flexible and more disordered, and the number of non-native contacts and hydrogen bonds between them decreases. For comparison, the effects of the weaker Aβ inhibitor ferulic acid (FA) on the primary nucleation are also examined. Our study is in good agreement with the observation that taken regularly, CU is able to prevent or at least delay the onset of neurodegenerative disorders.
Collapse
|
36
|
Levin A, Hakala TA, Schnaider L, Bernardes GJL, Gazit E, Knowles TPJ. Biomimetic peptide self-assembly for functional materials. Nat Rev Chem 2020; 4:615-634. [PMID: 39650726 PMCID: PMC7617017 DOI: 10.1038/s41570-020-0215-y] [Citation(s) in RCA: 407] [Impact Index Per Article: 81.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 01/20/2023]
Abstract
Natural biomolecular systems have evolved to form a rich variety of supramolecular materials and machinery fundamental to cellular function. The assembly of these structures commonly involves interactions between specific molecular building blocks, a strategy that can also be replicated in an artificial setting to prepare functional materials. The self-assembly of synthetic biomimetic peptides thus allows the exploration of chemical and sequence space beyond that used routinely by biology. In this Review, we discuss recent conceptual and experimental advances in self-assembling artificial peptidic materials. In particular, we explore how naturally occurring structures and phenomena have inspired the development of functional biomimetic materials that we can harness for potential interactions with biological systems. As our fundamental understanding of peptide self-assembly evolves, increasingly sophisticated materials and applications emerge and lead to the development of a new set of building blocks and assembly principles relevant to materials science, molecular biology, nanotechnology and precision medicine.
Collapse
Affiliation(s)
- Aviad Levin
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Tuuli A Hakala
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Lee Schnaider
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gonçalo J L Bernardes
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Tuomas P J Knowles
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Polypeptides derived from α-Synuclein binding partners to prevent α-Synuclein fibrils interaction with and take-up by cells. PLoS One 2020; 15:e0237328. [PMID: 32790707 PMCID: PMC7425896 DOI: 10.1371/journal.pone.0237328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/23/2020] [Indexed: 11/24/2022] Open
Abstract
α-Synuclein (αSyn) fibrils spread from one neuronal cell to another. This prion-like phenomenon is believed to contribute to the progression of the pathology in Parkinson’s disease and other synucleinopathies. The binding of αSyn fibrils originating from affected cells to the plasma membrane of naïve cells is key in their prion-like propagation propensity. To interfere with this process, we designed polypeptides derived from proteins we previously showed to interact with αSyn fibrils, namely the molecular chaperone Hsc70 and the sodium/potassium pump NaK-ATPase and assessed their capacity to bind αSyn fibrils and/or interfere with their take-up by cells of neuronal origin. We demonstrate here that polypeptides that coat αSyn fibrils surfaces in such a way that they are changed affect αSyn fibrils binding to the plasma membrane components and/or their take-up by cells. Altogether our observations suggest that the rationale design of αSyn fibrils polypeptide binders that interfere with their propagation between neuronal cells holds therapeutic potential.
Collapse
|
38
|
Xing Y, Sun Y, Wang B, Ding F. Morphological Determinants of Carbon Nanomaterial-Induced Amyloid Peptide Self-Assembly. Front Chem 2020; 8:160. [PMID: 32211382 PMCID: PMC7076083 DOI: 10.3389/fchem.2020.00160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/24/2020] [Indexed: 01/12/2023] Open
Abstract
Hybridizing carbon nanomaterials (CNMs) with amyloid fibrils—the ordered nanostructures self-assembled by amyloidogenic peptides—has found promising applications in bionanotechology. Understanding fundamental interactions of CNMs with amyloid peptides and uncovering the determinants of their self-assembly structures and dynamics are, therefore, pivotal for enriching and enabling this novel class of hybrid nanomaterials. Here, we applied atomistic molecular dynamics simulations to investigate the self-assembly of two amyloid peptides—the amyloidogenic core residues 16-22 of amyloid-β (Aβ16−22) and the non-amyloid-β core of α-synuclein (NACore68−78)—on the surface of carbon nanotubes (CNT) with different sizes and chirality. Our computational results showed that with small radial CNTs, both types of peptides could form β-sheets wrapping around the nanotube surface into a supercoiled morphology. The angle between β-strands and nanotube axes in the supercoil structure depended mainly on the peptide sequence and CNT radius, but also weakly on the CNT chirality. Large radial CNTs and the extreme case of the flat graphene nanosheet, on the other hand, could nucleate amyloid fibrils perpendicular to the surface. Our results provided new insights of hybridizing CNMs with amyloid peptides and also offered a novel approach to manipulate the morphology of CNM-induced amyloid assembly by tuning the surface curvature, peptide sequence, and molecular ratio between peptides and available CNM surface area, which may be useful in engineering nanocomposites with high-order structures.
Collapse
Affiliation(s)
- Yanting Xing
- Department of Physics and Astronomy, Clemson University, Clemson, SC, United States
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC, United States
| | - Bo Wang
- Department of Physics and Astronomy, Clemson University, Clemson, SC, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC, United States
| |
Collapse
|
39
|
Amyloid-like peptide nanofibrils as scaffolds for tissue engineering: Progress and challenges (Review). Biointerphases 2019; 14:040801. [PMID: 31284721 DOI: 10.1116/1.5098332] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Networks of amyloid-like nanofibrils assembled from short peptide sequences have the ability to form scaffolds that can encapsulate clinically relevant stem cells encouraging their attachment, growth, and differentiation into various lineages which can be used in tissue engineering applications to treat a range of diseases and traumas. In this review, the author highlights a selection of important proof-of-principle papers that show how this class of self-assembled networks is highly suited to biomaterial scaffold development. The author highlights recent studies which have shown that these scaffolds can be used to promote cell and tissue regeneration both in vitro and in vivo. The author also presents some fundamental knowledge gaps which are preventing the widespread translation of such scaffolds. Finally, the author outlines a selection of studies that elucidate molecular assembly mechanisms and biophysical properties of amyloid-like peptide nanofibrils and suggests how studies like these might lead to the ability to generate nanofibril scaffolds with bespoke properties for tissue engineering.
Collapse
|
40
|
Gosztyla ML, Brothers HM, Robinson SR. Alzheimer's Amyloid-β is an Antimicrobial Peptide: A Review of the Evidence. J Alzheimers Dis 2019; 62:1495-1506. [PMID: 29504537 DOI: 10.3233/jad-171133] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The amyloid-β (Aβ) peptide has long been considered to be the driving force behind Alzheimer's disease (AD). However, clinical trials that have successfully reduced Aβ burden in the brain have not slowed the cognitive decline, and in some instances, have resulted in adverse outcomes. While these results can be interpreted in different ways, a more nuanced picture of Aβ is emerging that takes into account the facts that the peptide is evolutionarily conserved and is present throughout life in cognitively normal individuals. Recent evidence indicates a role for Aβ as an antimicrobial peptide (AMP), a class of innate immune defense molecule that utilizes fibrillation to protect the host from a wide range of infectious agents. In humans and in animal models, infection of the brain frequently leads to increased amyloidogenic processing of the amyloid-β protein precursor (AβPP) and resultant fibrillary aggregates of Aβ. Evidence from in vitro and in vivo studies demonstrates that Aβ oligomers have potent, broad-spectrum antimicrobial properties by forming fibrils that entrap pathogens and disrupt cell membranes. Importantly, overexpression of Aβ confers increased resistance to infection from both bacteria and viruses. The antimicrobial role of Aβ may explain why increased rates of infection have been observed in some of the AD clinical trials that depleted Aβ. Perhaps progress toward a cure for AD will accelerate once treatment strategies begin to take into account the likely physiological functions of this enigmatic peptide.
Collapse
Affiliation(s)
- Maya L Gosztyla
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Holly M Brothers
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Stephen R Robinson
- Discipline of Psychology, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
41
|
α-Synuclein misfolding and aggregation: Implications in Parkinson's disease pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:890-908. [PMID: 30853581 DOI: 10.1016/j.bbapap.2019.03.001] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/03/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022]
Abstract
α-Synuclein (α-Syn) has been extensively studied for its structural and biophysical properties owing to its pathophysiological role in Parkinson's disease (PD). Lewy bodies and Lewy neurites are the pathological hallmarks of PD and contain α-Syn aggregates as their major component. It was therefore hypothesized that α-Syn aggregation is actively associated with PD pathogenesis. The central role of α-Syn aggregation in PD is further supported by the identification of point mutations in α-Syn protein associated with rare familial forms of PD. However, the correlation between aggregation propensities of α-Syn mutants and their association with PD phenotype is not straightforward. Recent evidence suggested that oligomers, formed during the initial stages of aggregation, are the potent neurotoxic species causing cell death in PD. However, the heterogeneous and unstable nature of these oligomers limit their detailed characterization. α-Syn fibrils, on the contrary, are shown to be the infectious agents and propagate in a prion-like manner. Although α-Syn is an intrinsically disordered protein, it exhibits remarkable conformational plasticity by adopting a range of structural conformations under different environmental conditions. In this review, we focus on the structural and functional aspects of α-Syn and role of potential factors that may contribute to the underlying mechanism of synucleinopathies. This information will help to identify novel targets and develop specific therapeutic strategies to combat Parkinson's and other protein aggregation related neurodegenerative diseases.
Collapse
|
42
|
Tan H, Jin D, Qu X, Liu H, Chen X, Yin M, Liu C. A PEG-Lysozyme hydrogel harvests multiple functions as a fit-to-shape tissue sealant for internal-use of body. Biomaterials 2019; 192:392-404. [DOI: 10.1016/j.biomaterials.2018.10.047] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022]
|
43
|
Kirti S, Patel K, Das S, Shrimali P, Samanta S, Kumar R, Chatterjee D, Ghosh D, Kumar A, Tayalia P, Maji SK. Amyloid Fibrils with Positive Charge Enhance Retroviral Transduction in Mammalian Cells. ACS Biomater Sci Eng 2018; 5:126-138. [DOI: 10.1021/acsbiomaterials.8b00248] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
George E, Barai A, Shirke P, Majumder A, Sen S. Engineering interfacial migration by collective tuning of adhesion anisotropy and stiffness. Acta Biomater 2018; 72:82-93. [PMID: 29574184 DOI: 10.1016/j.actbio.2018.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 01/30/2018] [Accepted: 03/07/2018] [Indexed: 02/05/2023]
Abstract
Interfacial migration is central to multiple processes including morphogenesis and wound healing. However, the sensitivity of interfacial migration to properties of the interfacial microenvironment has not been adequately explored. Here, we address this question by tracking motility of 3T3 fibroblasts at the interface of two hydrogels. By sandwiching cells between two adhesive gels (composed of methacrylated gelatin) or between an adhesive and a non-adhesive gel (composed of gellan), we show that cells are more motile in case of the latter. By tuning the bulk stiffness of the gellan gel, we then show that motility is tuned in a stiffness-dependent manner. Fastest motility observed in case of the stiffest gel was associated with increased cell height, suggestive of stiffness-mediated cytoskeletal assembly. Inhibition of cell motility by contractile agonists and actin depolymerizing drugs is indicative of a mode of migration wherein cells combine contractile tractions exerted at their base and actin-based pushing forces on the top surface to propel themselves forward. Together, our results suggest that dorso-ventral adhesion anisotropy and stiffness can be collectively tuned to engineer interfacial migration. STATEMENT OF SIGNIFICANCE It is increasingly understood that cells migrate in vivo through confining spaces which typically occur as pores in the matrix and through naturally occurring interfaces that exist between neighbouring ECM fibers, or between the stroma and the vasculature. Such interfaces are also created when treating wounds on the skin surface by covering the wounds with adhesives. How multiple cues impact interfacial migration has not been adequately addressed. By studying cell migratory behaviour at the interface of two hydrogel substrates, we identify adhesivity and stiffness as two critical factors that can be tuned to maximize cell migration. We foresee a potential use of this knowledge in the design of tissue adhesives for wound healing applications.
Collapse
|
45
|
Das S, Jacob RS, Patel K, Singh N, Maji SK. Amyloid Fibrils: Versatile Biomaterials for Cell Adhesion and Tissue Engineering Applications. Biomacromolecules 2018; 19:1826-1839. [DOI: 10.1021/acs.biomac.8b00279] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Subhadeep Das
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076 Maharashtra, India
| | - Reeba S. Jacob
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076 Maharashtra, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076 Maharashtra, India
| | - Namrata Singh
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076 Maharashtra, India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076 Maharashtra, India
| |
Collapse
|
46
|
Jacob RS, Das S, Singh N, Patel K, Datta D, Sen S, Maji SK. Amyloids Are Novel Cell-Adhesive Matrices. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:79-97. [PMID: 30637692 DOI: 10.1007/978-981-13-3065-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Amyloids are highly ordered peptide/protein aggregates traditionally associated with multiple human diseases including neurodegenerative disorders. However, recent studies suggest that amyloids can also perform several biological functions in organisms varying from bacteria to mammals. In many lower organisms, amyloid fibrils function as adhesives due to their unique surface topography. Recently, amyloid fibrils have been shown to support attachment and spreading of mammalian cells by interacting with the cell membrane and by cell adhesion machinery activation. Moreover, similar to cellular responses on natural extracellular matrices (ECMs), mammalian cells on amyloid surfaces also use integrin machinery for spreading, migration, and differentiation. This has led to the development of biocompatible and implantable amyloid-based hydrogels that could induce lineage-specific differentiation of stem cells. In this chapter, based on adhesion of both lower organisms and mammalian cells on amyloid nanofibrils, we posit that amyloids could have functioned as a primitive extracellular matrix in primordial earth.
Collapse
Affiliation(s)
- Reeba S Jacob
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Subhadeep Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Namrata Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Debalina Datta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India.
| |
Collapse
|
47
|
Ghosh S, Salot S, Sengupta S, Navalkar A, Ghosh D, Jacob R, Das S, Kumar R, Jha NN, Sahay S, Mehra S, Mohite GM, Ghosh SK, Kombrabail M, Krishnamoorthy G, Chaudhari P, Maji SK. p53 amyloid formation leading to its loss of function: implications in cancer pathogenesis. Cell Death Differ 2017; 24. [PMID: 28644435 PMCID: PMC5596421 DOI: 10.1038/cdd.2017.105] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transcriptional regulator p53 has an essential role in tumor suppression. Almost 50% of human cancers are associated with the loss of p53 functions, where p53 often accumulates in the nucleus as well as in cytoplasm. Although it has been previously suggested that amyloid formation could be a cause of p53 loss-of-function in subset of tumors, the characterization of these amyloids and its structure-function relationship is not yet established. In the current study, we provide several evidences for the presence of p53 amyloid formation (in human and animal cancer tissues); along with its isolation from human cancer tissues and the biophysical characterization of these tissue-derived fibrils. Using amyloid seed of p53 fragment (P8, p53(250-257)), we show that p53 amyloid formation in cells not only leads to its functional inactivation but also transforms it into an oncoprotein. The in vitro studies further show that cancer-associated mutation destabilizes the fold of p53 core domain and also accelerates the aggregation and amyloid formation by this protein. Furthermore, we also show evidence of prion-like cell-to-cell transmission of different p53 amyloid species including full-length p53, which is induced by internalized P8 fibrils. The present study suggests that p53 amyloid formation could be one of the possible cause of p53 loss of function and therefore, inhibiting p53 amyloidogenesis could restore p53 tumor suppressor functions.
Collapse
Affiliation(s)
- Saikat Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shimul Salot
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shinjinee Sengupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Dhiman Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Reeba Jacob
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Subhadeep Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
- IITB-Monash Research Academy, Indian Institute of Technology Bombay, Mumbai, India
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Narendra Nath Jha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shruti Sahay
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Ganesh M Mohite
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Santanu K Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Mamata Kombrabail
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Guruswamy Krishnamoorthy
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biotechnology, Anna University, Chennai, India
| | - Pradip Chaudhari
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India. Tel: +91 22 25767774; Fax: +91 2225767760, E-mail:
| |
Collapse
|
48
|
Das S, Kumar R, Jha NN, Maji SK. Controlled Exposure of Bioactive Growth Factor in 3D Amyloid Hydrogel for Stem Cells Differentiation. Adv Healthc Mater 2017; 6. [PMID: 28736995 DOI: 10.1002/adhm.201700368] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/09/2017] [Indexed: 12/21/2022]
Abstract
Amyloid based hydrogels can mimic the extracellular matrix and serve as matrices for tissue engineering both in vitro and in vivo. A pH responsive self-assembled amyloid hydrogel system is used to encapsulate various growth factors for driving stem cell differentiation toward neuronal lineage. Diffusion studies with fluorescence recovery after photobleaching and bulk release with the model protein fluorescein isothiocyanate-bovine serum albumin show that encapsulated protein molecules can be released in a sustained fashion from the hydrogel over a considerable period of time (30 d). Moreover, by modulating the porosity of the hydrogel by the simple addition of salt, the encapsulated protein molecules can be retained for a longer period of time within the hydrogel. Mesenchymal stem cells, when cultured in 3D amyloid hydrogels with growth factors fibroblast growth factor 8 and sonic hedgehog, show more neuron specific differentiation as compared to hydrogel alone. This higher differentiation potential of growth factor encapsulated amyloid hydrogels can be due to concomitant exposure of cells to biomechanical as well as biochemical cues during the course of differentiation. The present study suggests that amyloid based hydrogel can be exploited for controlled growth factor delivery as well as directed stem cell differentiation to neuron.
Collapse
Affiliation(s)
- Subhadeep Das
- IITB‐Monash Research Academy Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
- Department of Materials Science and Engineering Monash University Clayton VIC 3800 Australia
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| | - Narendra Nath Jha
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| |
Collapse
|
49
|
Nyström G, Fong WK, Mezzenga R. Ice-Templated and Cross-Linked Amyloid Fibril Aerogel Scaffolds for Cell Growth. Biomacromolecules 2017; 18:2858-2865. [DOI: 10.1021/acs.biomac.7b00792] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Gustav Nyström
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| | - Wye-Khay Fong
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
- Drug
Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Raffaele Mezzenga
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| |
Collapse
|
50
|
Dharmadana D, Reynolds NP, Conn CE, Valéry C. Molecular interactions of amyloid nanofibrils with biological aggregation modifiers: implications for cytotoxicity mechanisms and biomaterial design. Interface Focus 2017; 7:20160160. [PMID: 28630679 PMCID: PMC5474041 DOI: 10.1098/rsfs.2016.0160] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Amyloid nanofibrils are ubiquitous biological protein fibrous aggregates, with a wide range of either toxic or beneficial activities that are relevant to human disease and normal biology. Protein amyloid fibrillization occurs via nucleated polymerization, through non-covalent interactions. As such, protein nanofibril formation is based on a complex interplay between kinetic and thermodynamic factors. The process entails metastable oligomeric species and a highly thermodynamically favoured end state. The kinetics, and the reaction pathway itself, can be influenced by third party moieties, either molecules or surfaces. Specifically, in the biological context, different classes of biomolecules are known to act as catalysts, inhibitors or modifiers of the generic protein fibrillization process. The biological aggregation modifiers reviewed here include lipid membranes of varying composition, glycosaminoglycans and metal ions, with a final word on xenobiotic compounds. The corresponding molecular interactions are critically analysed and placed in the context of the mechanisms of cytotoxicity of the amyloids involved in diverse pathologies and the non-toxicity of functional amyloids (at least towards their biological host). Finally, the utilization of this knowledge towards the design of bio-inspired and biocompatible nanomaterials is explored.
Collapse
Affiliation(s)
- Durga Dharmadana
- School of Health and Biomedical Sciences, Discipline of Pharmaceutical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3001, Australia
| | - Nicholas P. Reynolds
- ARC Training Centre for Biodevices, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia
| | - Charlotte E. Conn
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3001, Australia
| | - Céline Valéry
- School of Health and Biomedical Sciences, Discipline of Pharmaceutical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| |
Collapse
|