1
|
Taief KA, Nemec S, Middleton IA, Kilian KA, Thordarson P. Scrambled RGD Hexameric Peptide Hydrogel Supports Efficient Self-Assembly and Cell Activity. Chemistry 2025; 31:e202404410. [PMID: 40192287 PMCID: PMC12080297 DOI: 10.1002/chem.202404410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
The amino acid sequence is crucial in controlling peptide-based hydrogel formation, whereby changing the position of a single amino acid can significantly alter the gel's properties. Herein, we report the gelation kinetics and cell viability of scrFmoc-GFFRDG (where we have scrambled the RGD-based gel hexapeptide; Fmoc-GFFRGD). The scrambled sequence showed improved gelation properties compared to the original Fmoc-GFFRGD sequence, with scrFmoc-GFFRDG forming a gel in under 10 min, significantly faster than the 2-h gelation time, and at a concentration eight times lower than the original Fmoc-GFFRGD sequence. We also examined the combination of the two gelators in a ratio of 1:1, final concentration of 0.4% (w/v). Interestingly, the stiffness of the hybrid hydrogel was ∼3 kPa, whereas individually, neither gelator at the same concentration exceeded 0.5 kPa. The cell-adhesion motif RGD improves the ability of the peptides to promote attachment of cells due to integrin recognition. However, when fibroblasts were cultured on the hydrogels, scrFmoc-GFFRDG yielded a higher level of α-SMA expression in cells than those cultured on Fmoc-GFFRGD, suggesting a microenvironment conducive to myofibroblast transitions. This study provides a new outlook on how a well-known scrambled peptide motif (RDG) can fine-tune hydrogel assembly and cell culture applications.
Collapse
Affiliation(s)
- Karrar Al Taief
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
| | - Stephanie Nemec
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Isis A. Middleton
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
| | - Kristopher A. Kilian
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Pall Thordarson
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
2
|
Perez-Chirinos L, Almonte L, Cortés-Ossa JD, Solano E, Calvo MR, Sasselli IR, Cortajarena AL. Tuning the Dimensionality of Protein-Peptide Coassemblies to Build 2D Conductive Nanomaterials. ACS NANO 2025; 19:16500-16516. [PMID: 40277076 PMCID: PMC12060649 DOI: 10.1021/acsnano.4c18613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
The natural self-assembly tendency of proteins to build complex structural architectures has kindled inspiration in developing supramolecular structures through the rational design of biomacromolecules. While there has been significant progress in achieving precise control over the morphology of self-assembled structures, combining different molecules within assemblies enables the design of materials with increased complexity, sophisticated structures, and a broad spectrum of functionalities. Here, the development of 1D and 2D peptide-protein coassembled systems based on the design of amphiphilic peptides and engineered proteins is described. The peptide was optimized to form stable self-assembled fibers by evaluating, computationally and experimentally, the assembling tendencies and the supramolecular features of peptides with different lengths and negative charges. A superhelical repeat protein was engineered by fusing one or two amphiphilic peptides into one or both termini. This modification drove the coassembly between the self-assembled fibers and the protein with one or two peptides, resulting in 1D or 2D coassembled systems. The protein films and the 2D coassembled system exhibited high ionic conductivity for a biomolecular system, attributed to their high content of charged residues, positioning these materials as promising candidates for developing bioelectronic devices. Thus, this work provides a versatile framework for developing coassembled materials with tunable dimensionality by using biocompatible building blocks without any additional chemical moieties, highlighting the potential for their use in biocompatible electronics.
Collapse
Affiliation(s)
- Laura Perez-Chirinos
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research
and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián 20014, Spain
| | - Lisa Almonte
- Instituto
Universitario de Materiales de Alicante (IUMA), Universidad de Alicante, Alicante 03690, Spain
- BCMaterials,
Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Vizcaya 48940, Spain
| | - Juan David Cortés-Ossa
- Instituto
Universitario de Materiales de Alicante (IUMA), Universidad de Alicante, Alicante 03690, Spain
- BCMaterials,
Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Vizcaya 48940, Spain
| | - Eduardo Solano
- NCD-SWEET
Beamline, ALBA Synchrotron Light Source, Cerdanyola del Vallès, Barcelona 08290, Spain
| | - M. Reyes Calvo
- Instituto
Universitario de Materiales de Alicante (IUMA), Universidad de Alicante, Alicante 03690, Spain
- BCMaterials,
Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Vizcaya 48940, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Ivan R. Sasselli
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research
and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián 20014, Spain
- Centro
de Física de Materiales (CFM), CSIC-UPV/EHU, Paseo Manuel de Lardizabal 5, Donostia-San Sebastián 20018, Spain
| | - Aitziber L. Cortajarena
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research
and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián 20014, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| |
Collapse
|
3
|
Marjan T, Lafuente-Gómez N, Rampal A, Mooney DJ, Peyton SR, Qazi TH. Cell-Instructive Biomaterials with Native-Like Biochemical Complexity. Annu Rev Biomed Eng 2025; 27:185-209. [PMID: 39874600 PMCID: PMC12045723 DOI: 10.1146/annurev-bioeng-120823-020209] [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] [Indexed: 01/30/2025]
Abstract
Biochemical signals in native tissue microenvironments instruct cell behavior during many biological processes ranging from developmental morphogenesis and tissue regeneration to tumor metastasis and disease progression. The detection and characterization of these signals using spatial and highly resolved quantitative methods have revealed their existence as matricellular proteins in the matrisome, some of which are bound to the extracellular matrix while others are freely diffusing. Including these biochemical signals in engineered biomaterials can impart enhanced functionality and native-like complexity, ultimately benefiting efforts to understand, model, and treat various diseases. In this review, we discuss advances in characterizing, mimicking, and harnessing biochemical signals in developing advanced engineered biomaterials. An overview of the diverse forms in which these biochemical signals exist and their effects on intracellular signal transduction is also provided. Finally, we highlight the application of biochemically complex biomaterials in the three broadly defined areas of tissue regeneration, immunoengineering, and organoid morphogenesis.
Collapse
Affiliation(s)
- Tuba Marjan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Nuria Lafuente-Gómez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Akaansha Rampal
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
4
|
Li L, Zheng R, Sun R. Hierarchical Self-Assembly of Short Peptides: Nanostructure Formation, Function Tailoring, and Applications. Macromol Biosci 2025; 25:e2400523. [PMID: 39887542 DOI: 10.1002/mabi.202400523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/20/2024] [Indexed: 02/01/2025]
Abstract
This article explores the hierarchical self-assembly of short peptides, which refers to the structured spatial arrangements of these molecules over long distances. This phenomenon is commonly found in nature and has important implications for biological structure and function. Short peptides are preferred for self-assembly because they have the ability to spontaneously create various nanostructures. This process, known as bottom-up assembly, allows for the addition of functional groups at the carboxyl or amine ends of the peptides. These functional groups enable specific functions that are extremely valuable in the fields of biotechnology and biomedicine. This text discusses the basic processes involved in the self-assembly of short peptides, such as the characteristics of amino acid side chains, the categorization of short peptides according to their chemical structure, the influence of intermolecular forces, and the dynamic nature of the self-assembly process. In addition, the paper discusses the various uses of short peptides in the disciplines of biomedicine and optoelectronics, including stimulus-responsive hydrogels, tissue engineering, and drug delivery. The article also suggests rational design principles for controlling the hierarchical self-assembly of short peptides, creating new commercial applications, particularly with functional hydrogels, and offers insights into the future of the discipline.
Collapse
Affiliation(s)
- Liangchun Li
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| | - Renlin Zheng
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| | - Rongqin Sun
- School of Materials Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| |
Collapse
|
5
|
Moreno-Vargas LM, Prada-Gracia D. Exploring the Chemical Features and Biomedical Relevance of Cell-Penetrating Peptides. Int J Mol Sci 2024; 26:59. [PMID: 39795918 PMCID: PMC11720145 DOI: 10.3390/ijms26010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/13/2025] Open
Abstract
Cell-penetrating peptides (CPPs) are a diverse group of peptides, typically composed of 4 to 40 amino acids, known for their unique ability to transport a wide range of substances-such as small molecules, plasmid DNA, small interfering RNA, proteins, viruses, and nanoparticles-across cellular membranes while preserving the integrity of the cargo. CPPs exhibit passive and non-selective behavior, often requiring functionalization or chemical modification to enhance their specificity and efficacy. The precise mechanisms governing the cellular uptake of CPPs remain ambiguous; however, electrostatic interactions between positively charged amino acids and negatively charged glycosaminoglycans on the membrane, particularly heparan sulfate proteoglycans, are considered the initial crucial step for CPP uptake. Clinical trials have highlighted the potential of CPPs in diagnosing and treating various diseases, including cancer, central nervous system disorders, eye disorders, and diabetes. This review provides a comprehensive overview of CPP classifications, potential applications, transduction mechanisms, and the most relevant algorithms to improve the accuracy and reliability of predictions in CPP development.
Collapse
|
6
|
Tyagi G, Sengupta S. Unveiling the multifaceted potential of amyloid fibrils: from pathogenic myths to biotechnological marvels. Biophys Rev 2024; 16:737-751. [PMID: 39830121 PMCID: PMC11735760 DOI: 10.1007/s12551-024-01232-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/18/2024] [Indexed: 01/22/2025] Open
Abstract
Amyloid fibrils, historically stigmatized due to their association with diseases like Alzheimer's and Parkinson's, are now recognized as a distinct class of functional proteins with extraordinary potential. These highly ordered, cross-β-sheet protein aggregates are found across all domains of life, playing crucial physiological roles. In bacteria, functional amyloids like curli fibers are essential for surface adhesion, biofilm formation, and viral DNA packaging. Fungal prions exploit amyloid conformations to regulate translation, metabolism, and virulence, while mammalian amyloids are integral to melanin synthesis, hormone storage, and antimicrobial defense. The stability and hydrophobic nature of amyloid scaffolds underpin these diverse biological functions. Beyond their natural roles, amyloid fibrils offer unique capabilities in biomedicine, nanotechnology, and materials science. Their exceptional mechanical strength and biocompatibility make them ideal for controlled drug delivery, tissue engineering scaffolds, and enzyme immobilization. The intrinsic fluorescence and optical properties of certain amyloids open up innovative applications in biosensors, molecular probes, and optoelectronic devices. Furthermore, amyloid fibrils can template metal nanowires, enhance conducting materials, and form nanocomposites by integrating with polymers. This newfound appreciation for the functional diversity of amyloids has ignited intense research efforts to elucidate their molecular mechanisms, stability, and tunable properties. By unraveling the structural intricacies of functional amyloids, researchers aim to harness their remarkable attributes for groundbreaking biomedical therapies, advanced nanomaterials, and sustainable biotechnological innovations. This review explores the transformative journey of amyloids from pathological entities to biotechnological marvels, highlighting their vast potential across agriculture, environmental remediation, and industrial processes.
Collapse
Affiliation(s)
- Gauri Tyagi
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, 201313 Noida, India
| | - Shinjinee Sengupta
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, 201313 Noida, India
| |
Collapse
|
7
|
Mashweu AR, Azov VA. Nanotechnology in Drug Delivery: Anatomy and Molecular Insight into the Self-Assembly of Peptide-Based Hydrogels. Molecules 2024; 29:5654. [PMID: 39683812 PMCID: PMC11643151 DOI: 10.3390/molecules29235654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
The bioavailability, release, and stability of pharmaceuticals under physicochemical conditions is the major cause of drug candidates failing during their clinical trials. Therefore, extensive efforts have been invested in the development of novel drug delivery systems that are able to transport drugs to a desired site and improve bioavailability. Hydrogels, and peptide hydrogels in particular, have been extensively investigated due to their excellent biocompatibility and biodegradability properties. However, peptide hydrogels often have weak mechanical strength, which limits their therapeutic efficacy. Therefore, a number of methods for improving their rheological properties have been established. This review will cover the broad area of drug delivery, focusing on the recent developments in this research field. We will discuss the variety of different types of nanocarrier drug delivery systems and then, more specifically, the significance and perspectives of peptide-based hydrogels. In particular, the interplay of intermolecular forces that govern the self-assembly of peptide hydrogels, progress made in understanding the distinct morphologies of hydrogels, and applications of non-canonical amino acids in hydrogel design will be discussed in more detail.
Collapse
Affiliation(s)
- Adelaide R. Mashweu
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Vladimir A. Azov
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| |
Collapse
|
8
|
Ligorio C, Martinez-Espuga M, Laurenza D, Hartley A, Rodgers CB, Kotowska AM, Scurr DJ, Dalby MJ, Ordóñez-Morán P, Mata A. Disassembly of self-assembling peptide hydrogels as a versatile method for cell extraction and manipulation. J Mater Chem B 2024; 12:11939-11952. [PMID: 39449374 PMCID: PMC11502993 DOI: 10.1039/d4tb01575d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Self-assembling peptide hydrogels (SAPHs) are increasingly being used as two-dimensional (2D) cell culture substrates and three-dimensional (3D) matrices due to their tunable properties and biomimicry of native tissues. Despite these advantages, SAPHs often represent an end-point in cell culture, as isolating cells from them leads to low yields and disruption of cells, limiting their use and post-culture analyses. Here, we report on a protocol designed to easily and effectively disassemble peptide amphiphile (PA) SAPHs to retrieve 3D encapsulated cells with high viability and minimal disruption. Due to the pivotal role played by salt ions in SAPH gelation, tetrasodium ethylenediaminetetraacetic acid (Na4EDTA) was used as metal chelator to sequester ions participating in PA self-assembly and induce a rapid, efficient, clean, and gentle gel-to-sol transition. We characterise PA disassembly from the nano- to the macro-scale, provide mechanistic and practical insights into the PA disassembly mechanism, and assess the potential use of the process. As proof-of-concept, we isolated different cell types from cell-laden PA hydrogels and demonstrated the possibility to perform downstream biological analyses including cell re-plating, gene analysis, and flow cytometry with high reproducibility and no material interference. Our work offers new opportunities for the use of SAPHs in cell culture and the potential use of cells cultured on SAPHs, in applications such as cell expansion, analysis of in vitro models, cell therapies, and regenerative medicine.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Magda Martinez-Espuga
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Domenico Laurenza
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Alex Hartley
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Chloe B Rodgers
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow G11 6EW, UK
| | - Anna M Kotowska
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David J Scurr
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow G11 6EW, UK
| | - Paloma Ordóñez-Morán
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| |
Collapse
|
9
|
Xie C, Chen Y, Wang L, Liao K, Xue B, Han Y, Li L, Jiang Q. Recent research of peptide-based hydrogel in nervous regeneration. Bioact Mater 2024; 40:503-523. [PMID: 39040568 PMCID: PMC11261279 DOI: 10.1016/j.bioactmat.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Neurological disorders exert significantly affect the quality of life for patients, necessitating effective strategies for nerve regeneration. Both traditional autologous nerve transplantation and emerging therapeutic approaches encounter scientific challenges due to the complex nature of the nervous system and the unsuitability of the surrounding environment for cell transplantation. Tissue engineering techniques offer a promising path for neurotherapy. Successful neural tissue engineering relies on modulating cell differentiation behavior and tissue repair by developing biomaterials that mimic the natural extracellular matrix (ECM) and establish a three-dimensional microenvironment. Peptide-based hydrogels have emerged as a potent option among these biomaterials due to their ability to replicate the structure and complexity of the ECM. This review aims to explore the diverse range of peptide-based hydrogels used in nerve regeneration with a specific focus on dipeptide hydrogels, tripeptide hydrogels, oligopeptide hydrogels, multidomain peptides (MDPs), and amphiphilic peptide hydrogels (PAs). Peptide-based hydrogels offer numerous advantages, including biocompatibility, structural diversity, adjustable mechanical properties, and degradation without adverse effects. Notably, hydrogels formed from self-assembled polypeptide nanofibers, derived from amino acids, show promising potential in engineering neural tissues, outperforming conventional materials like alginate, poly(ε-caprolactone), and polyaniline. Additionally, the simple design and cost-effectiveness of dipeptide-based hydrogels have enabled the creation of various functional supramolecular structures, with significant implications for nervous system regeneration. These hydrogels are expected to play a crucial role in future neural tissue engineering research. This review aims to highlight the benefits and potential applications of peptide-based hydrogels, contributing to the advancement of neural tissue engineering.
Collapse
Affiliation(s)
- Chunmei Xie
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Yueyang Chen
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Lang Wang
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Kin Liao
- Advanced Digital and Additive Manufacturing Center, Khalifa University of Science and Technology, Po Box 127788, Abu Dhabi, United Arab Emirates
| | - Bin Xue
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing, China
| | - Yulong Han
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Bonde S, Osmani RAM, Trivedi R, Patravale V, Angolkar M, Prasad AG, Ravikumar AA. Harnessing DNA origami's therapeutic potential for revolutionizing cardiovascular disease treatment: A comprehensive review. Int J Biol Macromol 2024; 270:132246. [PMID: 38735608 DOI: 10.1016/j.ijbiomac.2024.132246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
DNA origami is a cutting-edge nanotechnology approach that creates precise and detailed 2D and 3D nanostructures. The crucial feature of DNA origami is how it is created, which enables precise control over its size and shape. Biocompatibility, targetability, programmability, and stability are further advantages that make it a potentially beneficial technique for a variety of applications. The preclinical studies of sophisticated programmable nanomedicines and nanodevices that can precisely respond to particular disease-associated triggers and microenvironments have been made possible by recent developments in DNA origami. These stimuli, which are endogenous to the targeted disorders, include protein upregulation, pH, redox status, and small chemicals. Oncology has traditionally been the focus of the majority of past and current research on this subject. Therefore, in this comprehensive review, we delve into the intricate world of DNA origami, exploring its defining features and capabilities. This review covers the fundamental characteristics of DNA origami, targeting DNA origami to cells, cellular uptake, and subcellular localization. Throughout the review, we emphasised on elucidating the imperative for such a therapeutic platform, especially in addressing the complexities of cardiovascular disease (CVD). Moreover, we explore the vast potential inherent in DNA origami technology, envisioning its promising role in the realm of CVD treatment and beyond.
Collapse
Affiliation(s)
- Smita Bonde
- Department of Pharmaceutics, SSR College of Pharmacy, Silvassa 396230, UT of Dadra and Nagar Haveli, India.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Rashmi Trivedi
- Department of Pharmaceutics, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, Maharashtra, India.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga (E), Mumbai 400019, Maharashtra, India.
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Aprameya Ganesh Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Akhila Akkihebbal Ravikumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| |
Collapse
|
11
|
Dranseike D, Ota Y, Edwardson TGW, Guzzi EA, Hori M, Nakic ZR, Deshmukh DV, Levasseur MD, Mattli K, Tringides CM, Zhou J, Hilvert D, Peters C, Tibbitt MW. Designed modular protein hydrogels for biofabrication. Acta Biomater 2024; 177:107-117. [PMID: 38382830 DOI: 10.1016/j.actbio.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
Designing proteins that fold and assemble over different length scales provides a way to tailor the mechanical properties and biological performance of hydrogels. In this study, we designed modular proteins that self-assemble into fibrillar networks and, as a result, form hydrogel materials with novel properties. We incorporated distinct functionalities by connecting separate self-assembling (A block) and cell-binding (B block) domains into single macromolecules. The number of self-assembling domains affects the rigidity of the fibers and the final storage modulus G' of the materials. The mechanical properties of the hydrogels could be tuned over a broad range (G' = 0.1 - 10 kPa), making them suitable for the cultivation and differentiation of multiple cell types, including cortical neurons and human mesenchymal stem cells. Moreover, we confirmed the bioavailability of cell attachment domains in the hydrogels that can be further tailored for specific cell types or other biological applications. Finally, we demonstrate the versatility of the designed proteins for application in biofabrication as 3D scaffolds that support cell growth and guide their function. STATEMENT OF SIGNIFICANCE: Designed proteins that enable the decoupling of biophysical and biochemical properties within the final material could enable modular biomaterial engineering. In this context, we present a designed modular protein platform that integrates self-assembling domains (A blocks) and cell-binding domains (B blocks) within a single biopolymer. The linking of assembly domains and cell-binding domains this way provided independent tuning of mechanical properties and inclusion of biofunctional domains. We demonstrate the use of this platform for biofabrication, including neural cell culture and 3D printing of scaffolds for mesenchymal stem cell culture and differentiation. Overall, this work highlights how informed design of biopolymer sequences can enable the modular design of protein-based hydrogels with independently tunable biophysical and biochemical properties.
Collapse
Affiliation(s)
- Dalia Dranseike
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland
| | - Yusuke Ota
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland
| | | | - Elia A Guzzi
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland
| | - Mao Hori
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Kevin Mattli
- Biosystems Technology, ZHAW, Wädenswil, Switzerland
| | | | - Jiangtao Zhou
- Laboratory of Food and Soft Materials, ETH Zurich, Switzerland
| | - Donald Hilvert
- Organic Chemistry Laboratory, ETH Zurich, Zurich, Switzerland.
| | | | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Luo R, Wan Y, Liu G, Chen J, Luo X, Li Z, Su D, Lu N, Luo Z. Engineering Self-Assembling Peptide Hydrogel to Enhance the Capacity of Dendritic Cells to Activate In Vivo T-Cell Immunity. Biomacromolecules 2024; 25:1408-1428. [PMID: 38236703 DOI: 10.1021/acs.biomac.3c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The efficacy of the dendritic cell (DC) has failed to meet expectations thus far, and crucial problems such as the immature state of DCs, low targeting efficiency, insufficient number of dendritic cells, and microenvironment are still the current focus. To address these problems, we developed two self-assembling peptides, RLDI and RQDT, that mimic extracellular matrix (ECM). These peptides can be self-assembled into highly ordered three-dimensional nanofiber scaffold structures, where RLDI can form gelation immediately. In addition, we found that RLDI and RQDT enhance the biological function of DCs, including releasing antigens sustainably, adhering to DCs, promoting the maturation of DCs, and increasing the ability of DC antigen presentation. Moreover, peptide hydrogel-based DC treatment significantly achieved prophylactic and treatment effects on colon cancer. These results have certain implications for the design of new broad-spectrum vaccines in the future.
Collapse
Affiliation(s)
- Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa 52242, United States
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhaoxu Li
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
13
|
Chavda VP, Teli D, Balar PC, Davidson M, Bojarska J, Vaghela DA, Apostolopoulos V. Self-assembled peptide hydrogels for the treatment of diabetes and associated complications. Colloids Surf B Biointerfaces 2024; 235:113761. [PMID: 38281392 DOI: 10.1016/j.colsurfb.2024.113761] [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: 07/25/2023] [Revised: 11/27/2023] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Diabetes is a widespread epidemic that includes a number of comorbid conditions that greatly increase the chance of acquiring other chronic illnesses. Every year, there are significantly more people with diabetes because of the rise in type-2 diabetes prevalence. The primary causes of illness and mortality worldwide are, among these, hyperglycemia and its comorbidities. There has been a lot of interest in the creation of peptide-based hydrogels as a potentially effective platform for the treatment of diabetes and its consequences. Here, we emphasize the use of self-assembled hydrogel formulations and their unique potential for the treatment/management of type-2 diabetes and its consequences. (i.e., wounds). Key aspects covered include the characteristics of self-assembled peptide hydrogels, methods for their preparation, and their pre-clinical and clinical applications in addressing metabolic disorders such as type-2 diabetes.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, India.
| | - Divya Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Pankti C Balar
- Pharmacy section, L.M. College of Pharmacy, Ahmedabad, India
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Joanna Bojarska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromski S.t, 90-924 Lodz, Poland.
| | - Dixa A Vaghela
- Pharmacy section, L.M. College of Pharmacy, Ahmedabad, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia; Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Lee ALZ, Balakrishnan N, Ng JY, Liu S, Ong ZY, Wang Y, Gao S, Yang YY. Injectable Hydrogels Prepared Using Novel Synthetic Short Peptides with Defined Structure and Gelatin as Scaffolds to Support Cell and Tissue Growth. Adv Healthc Mater 2024; 13:e2302786. [PMID: 37837308 DOI: 10.1002/adhm.202302786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Indexed: 10/15/2023]
Abstract
Animal-derived basement-membrane matrices such as Geltrex are used to grow cells and tissues. Particularly, these are commonly applied to support tumor growth in animals for cancer research. However, a material derived from an animal source has an undefined composition, and may thus have unavoidable batch-to-batch variation in properties. To overcome these issues, a series of synthetic short peptides to form hydrogels is designed in combination with gelatin to promote cell adhesion and growth. The peptides have sequences of (X1Y1X2Y2)2 , where X1 and X2 are hydrophobic residues, while Y1 and Y2 are hydrophilic residues. The peptides spontaneously fold and self-assemble into a β-sheet secondary structure upon contact with salts, and then aggregate to form hydrophilic networks of hydrogels. Hybrid hydrogels formed by mixing the peptide IEVEIRVK (IVK8) with gelatin are injectable and enzymatically degradable. The hybrid hydrogels at optimal compositions support SW480 and HepG2 tumor spheroid growth in vitro as effectively as Geltrex. More importantly, the peptide/gelatin hydrogels support tumor growth in a SW480 human colorectal adenocarcinoma xenograft mouse model. Altogether, the results illustrate that the synthetic peptide/gelatin hybrid hydrogel is a promising scaffold that can be used to support cell and tissue growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Ashlynn Ling Zhi Lee
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Nithiyaa Balakrishnan
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Jian Yao Ng
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Shaoqiong Liu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Zhan Yuin Ong
- School of Physics and Astronomy, St. James's University of Leeds, Leeds, LS2 9JT, UK
- Leeds Institute of Medical Research, St. James's University of Leeds, Leeds, LS2 9JT, UK
| | - Yanming Wang
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| |
Collapse
|
15
|
Giorgi Z, Veneruso V, Petillo E, Veglianese P, Perale G, Rossi F. Biomaterials and Cell Therapy Combination in Central Nervous System Treatments. ACS APPLIED BIO MATERIALS 2024; 7:80-98. [PMID: 38158393 PMCID: PMC10792669 DOI: 10.1021/acsabm.3c01058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Current pharmacological and surgical therapies for the central nervous system (CNS) show a limited capacity to reduce the damage progression; that together with the intrinsic limited capability of the CNS to regenerate greatly reduces the hopes of recovery. Among all the therapies proposed, the tissue engineering strategies supplemented with therapeutic stem cells remain the most promising. Neural tissue engineering strategies are based on the development of devices presenting optimal physical, chemical, and mechanical properties which, once inserted in the injured site, can support therapeutic cells, limiting the effect of a hostile environment and supporting regenerative processes. Thus, this review focuses on the employment of hydrogel and nanofibrous scaffolds supplemented with stem cells as promising therapeutic tools for the central and peripheral nervous systems in preclinical and clinical applications.
Collapse
Affiliation(s)
- Zoe Giorgi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Valeria Veneruso
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Emilia Petillo
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Pietro Veglianese
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Giuseppe Perale
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
- Ludwig
Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Filippo Rossi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| |
Collapse
|
16
|
Yu P, Duan L, Yan Z, Li J, Cai DZ. RADA-16-based Self-assembled Peptide Nanofiber Scaffolds Loaded with TGF-β1 Enhance the Chondrogenic Differentiation Potential of BMSCs In vitro. Curr Stem Cell Res Ther 2024; 19:257-266. [PMID: 36927429 DOI: 10.2174/1574888x18666230316112847] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVE At present, cartilage repair does not offer ideal efficacy. Fortunately, recent studies have claimed that RADA-16 peptide is an attractive therapeutic strategy for repairing cartilage defects. Therefore, this study tried to explore the effect of RADA-16 loaded with transforming growth factor-beta (TGF-β) 1 on cartilage differentiation of bone marrow mesenchymal stem cells (BMSCs). METHODS First, the RADA-16 peptide was synthesized by solid phase peptide, and a well-defined hydrogel was formed by supramolecular peptide self-assembly. Then, TGF-β1 (loading concentration of 10 ng/mL) was loaded into RADA-16, with scanning electron microscopy to observe the morphology of the TGF-β1/RADA-16 hydrogel and detect its related properties. Next, BMSCs were isolated from bone marrow samples and identified. TGF-β1/RADA-16 was co-cultured with L929, BMSCs, and C28/I2 cells, respectively, and the survival and proliferation ability of the cells was determined by live/dead cell staining and MTT assay. Chondrogenic differentiation and sGAG production of BMSCs were determined by Alcian blue staining and Blyscan assay, the expression of cartilage-associated genes by qRT-PCR, and the levels of inflammatory factors by ELISA. As for mechanism investigation, the Smad and ERK/MAPK signaling pathways were detected by western blot. RESULTS RADA-16 hydrogel exhibited a well-distributed and interconnected porous surface structure, with a loading rate of 91.9% for TGF-β1. The TGF-β1/RADA-16 hydrogel had good release and degradation properties, and had no negative effect on the survival and proliferation ability of BMSCs, L929, and C28/I2 cells. Importantly, TGF-β1/RADA-16 hydrogel significantly accelerated chondrogenic differentiation and sGAG generation in BMSCs, and decreased pro-inflammatory factor production. In addition, the hydrogel also significantly activated the Smad and ERK/MAPK pathways of BMSCs. CONCLUSION RADA-16 loaded with TGF-β1 has good biological properties and can enhance the chondrogenic differentiation ability of BMSCs.
Collapse
Affiliation(s)
- Peng Yu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Department of Joint Surgery, The First Affiliated Hospital of Hainan Medical University. Haikou, 570102, China
| | - Lian Duan
- Department of Joint Surgery, The First Affiliated Hospital of Hainan Medical University. Haikou, 570102, China
| | - Zhen Yan
- Department of Joint Surgery, The First Affiliated Hospital of Hainan Medical University. Haikou, 570102, China
| | - Jun Li
- Department of Joint Surgery, The First Affiliated Hospital of Hainan Medical University. Haikou, 570102, China
| | - Dao-Zhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
17
|
Aqel S, Al-Thani N, Haider MZ, Abdelhady S, Al Thani AA, Kobeissy F, Shaito AA. Biomaterials in Traumatic Brain Injury: Perspectives and Challenges. BIOLOGY 2023; 13:21. [PMID: 38248452 PMCID: PMC10813103 DOI: 10.3390/biology13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term impairment globally. TBI has a dynamic pathology, encompassing a variety of metabolic and molecular events that occur in two phases: primary and secondary. A forceful external blow to the brain initiates the primary phase, followed by a secondary phase that involves the release of calcium ions (Ca2+) and the initiation of a cascade of inflammatory processes, including mitochondrial dysfunction, a rise in oxidative stress, activation of glial cells, and damage to the blood-brain barrier (BBB), resulting in paracellular leakage. Currently, there are no FDA-approved drugs for TBI, but existing approaches rely on delivering micro- and macromolecular treatments, which are constrained by the BBB, poor retention, off-target toxicity, and the complex pathology of TBI. Therefore, there is a demand for innovative and alternative therapeutics with effective delivery tactics for the diagnosis and treatment of TBI. Tissue engineering, which includes the use of biomaterials, is one such alternative approach. Biomaterials, such as hydrogels, including self-assembling peptides and electrospun nanofibers, can be used alone or in combination with neuronal stem cells to induce neurite outgrowth, the differentiation of human neural stem cells, and nerve gap bridging in TBI. This review examines the inclusion of biomaterials as potential treatments for TBI, including their types, synthesis, and mechanisms of action. This review also discusses the challenges faced by the use of biomaterials in TBI, including the development of biodegradable, biocompatible, and mechanically flexible biomaterials and, if combined with stem cells, the survival rate of the transplanted stem cells. A better understanding of the mechanisms and drawbacks of these novel therapeutic approaches will help to guide the design of future TBI therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Asmaa A. Al Thani
- Biomedical Research Center and Department of Biomedical Sciences, College of Health Science, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
18
|
Nguyen AK, Molley TG, Kardia E, Ganda S, Chakraborty S, Wong SL, Ruan J, Yee BE, Mata J, Vijayan A, Kumar N, Tilley RD, Waters SA, Kilian KA. Hierarchical assembly of tryptophan zipper peptides into stress-relaxing bioactive hydrogels. Nat Commun 2023; 14:6604. [PMID: 37872151 PMCID: PMC10593748 DOI: 10.1038/s41467-023-41907-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/22/2023] [Indexed: 10/25/2023] Open
Abstract
Soft materials in nature are formed through reversible supramolecular assembly of biological polymers into dynamic hierarchical networks. Rational design has led to self-assembling peptides with structural similarities to natural materials. However, recreating the dynamic functional properties inherent to natural systems remains challenging. Here we report the discovery of a short peptide based on the tryptophan zipper (trpzip) motif, that shows multiscale hierarchical ordering that leads to emergent dynamic properties. Trpzip hydrogels are antimicrobial and self-healing, with tunable viscoelasticity and unique yield-stress properties that allow immediate harvest of embedded cells through a flick of the wrist. This characteristic makes Trpzip hydrogels amenable to syringe extrusion, which we demonstrate with examples of cell delivery and bioprinting. Trpzip hydrogels display innate bioactivity, allowing propagation of human intestinal organoids with apical-basal polarization. Considering these extensive attributes, we anticipate the Trpzip motif will prove a versatile building block for supramolecular assembly of soft materials for biotechnology and medicine.
Collapse
Affiliation(s)
- Ashley K Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thomas G Molley
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Materials Science and Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Egi Kardia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sylvia Ganda
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sudip Chakraborty
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sharon L Wong
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Juanfang Ruan
- Electron Microscopy Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Bethany E Yee
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jitendra Mata
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organization, Lucas Heights, NSW, 2234, Australia
| | - Abhishek Vijayan
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Richard D Tilley
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Electron Microscopy Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shafagh A Waters
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Randwick, NSW, 2031, Australia
| | - Kristopher A Kilian
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- School of Materials Science and Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
19
|
Kurokawa M, Ohtsu T, Chatani E, Tamura A. Hyper Thermostability and Liquid-Crystal-Like Properties of Designed α-Helical Peptide Nanofibers. J Phys Chem B 2023; 127:8331-8343. [PMID: 37751540 DOI: 10.1021/acs.jpcb.3c03833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Structural and thermodynamic transitions of artificially designed α-helical nanofibers were investigated using eight peptide variants, including four peptides with amide-modified carboxyl termini (CB peptides) and four unmodified peptides (CF peptides). Temperature-dependent circular dichroism spectroscopy and differential scanning calorimetry showed that CB peptides exhibit thermostability up to 50 °C higher than CF peptides. As a result, one of the denaturation temperatures approached nearly 130 °C, which is exceptionally high for a biomacromolecule. Thermodynamic analysis and microscopy observations also showed that CB peptides undergo a thermal transition similar to the phase transition in liquid crystals. In addition, one of the peptides showed a sharp and highly cooperative transition with a small enthalpy change at around 25 °C, which was ascribed to a giga-bundle burst of the molecular assembly. These macroscopic changes in the thermostability and crystallinity of CB peptides may be attributed to an increased amphiphilicity of the molecule in the direction of the helix axis, originating from the microscopic modification of the carboxyl-terminus.
Collapse
Affiliation(s)
- Minami Kurokawa
- Graduate School of Science, Kobe University, 1-1 Rokkoudai, Nada, Kobe, 657-8501, Japan
| | - Tomoya Ohtsu
- Graduate School of Science, Kobe University, 1-1 Rokkoudai, Nada, Kobe, 657-8501, Japan
| | - Eri Chatani
- Graduate School of Science, Kobe University, 1-1 Rokkoudai, Nada, Kobe, 657-8501, Japan
| | - Atsuo Tamura
- Graduate School of Science, Kobe University, 1-1 Rokkoudai, Nada, Kobe, 657-8501, Japan
| |
Collapse
|
20
|
Emamyari S, Mirzaei M, Mohammadinejad S, Fazli D, Fazli H. Impact of flexibility on the aggregation of polymeric macromolecules. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:66. [PMID: 37522950 DOI: 10.1140/epje/s10189-023-00324-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
Dependence of the dimerization probability and the aggregation behavior of polymeric macromolecules on their flexibility is studied using Langevin dynamics simulations. It is found that the dimerization probability is a non-monotonic function of the polymers persistence length. For a given value of inter-polymer attraction strength, semiflexible polymers have lower dimerization probability relative to flexible and rigid polymers of the same length. The threshold temperature of the formation of aggregates in a many-polymer system and its dependence on the polymers persistence length is also investigated. The simulation results of two- and many-polymer systems are in good agreement and show how the amount of flexibility affects the dimerization and the aggregation behaviors of polymeric macromolecules.
Collapse
Affiliation(s)
- Soheila Emamyari
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran
| | - Masoud Mirzaei
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran
| | - Sarah Mohammadinejad
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran
| | - Davood Fazli
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran
| | - Hossein Fazli
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran.
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Prof. Yousef Sobouti Blvd., Zanjan, 45137-66731, Iran.
| |
Collapse
|
21
|
Bertouille J, Kasas S, Martin C, Hennecke U, Ballet S, Willaert RG. Fast Self-Assembly Dynamics of a β-Sheet Peptide Soft Material. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206795. [PMID: 36807731 DOI: 10.1002/smll.202206795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/28/2022] [Indexed: 05/18/2023]
Abstract
Peptide-based hydrogels are promising biocompatible materials for wound healing, drug delivery, and tissue engineering applications. The physical properties of these nanostructured materials depend strongly on the morphology of the gel network. However, the self-assembly mechanism of the peptides that leads to a distinct network morphology is still a subject of ongoing debate, since complete assembly pathways have not yet been resolved. To unravel the dynamics of the hierarchical self-assembly process of the model β-sheet forming peptide KFE8 (Ac-FKFEFKFE-NH2 ), high-speed atomic force microscopy (HS-AFM) in liquid is used. It is demonstrated that a fast-growing network, based on small fibrillar aggregates, is formed at a solid-liquid interface, while in bulk solution, a distinct, more prolonged nanotube network emerges from intermediate helical ribbons. Moreover, the transformation between these morphologies has been visualized. It is expected that this new in situ and in real-time methodology will set the path for the in-depth unravelling of the dynamics of other peptide-based self-assembled soft materials, as well as gaining advanced insights into the formation of fibers involved in protein misfolding diseases.
Collapse
Affiliation(s)
- Jolien Bertouille
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Sandor Kasas
- Laboratory of Biological Electron Microscopy, Ecole Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
- International Joint Research Group VUB-EPFL BioNanotechnology & NanoMedicine, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Ulrich Hennecke
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Ronnie G Willaert
- International Joint Research Group VUB-EPFL BioNanotechnology & NanoMedicine, Vrije Universiteit Brussel, Brussels, 1050, Belgium
- Research Group Structural Biology Brussels, Alliance Research Group VUB-UGent NanoMicrobiology, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| |
Collapse
|
22
|
Duarte AC, Costa EC, Filipe HAL, Saraiva SM, Jacinto T, Miguel SP, Ribeiro MP, Coutinho P. Animal-derived products in science and current alternatives. BIOMATERIALS ADVANCES 2023; 151:213428. [PMID: 37146527 DOI: 10.1016/j.bioadv.2023.213428] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
More than fifty years after the 3Rs definition and despite the continuous implementation of regulatory measures, animals continue to be widely used in basic research. Their use comprises not only in vivo experiments with animal models, but also the production of a variety of supplements and products of animal origin for cell and tissue culture, cell-based assays, and therapeutics. The animal-derived products most used in basic research are fetal bovine serum (FBS), extracellular matrix proteins such as Matrigel™, and antibodies. However, their production raises several ethical issues regarding animal welfare. Additionally, their biological origin is associated with a high risk of contamination, resulting, frequently, in poor scientific data for clinical translation. These issues support the search for new animal-free products able to replace FBS, Matrigel™, and antibodies in basic research. In addition, in silico methodologies play an important role in the reduction of animal use in research by refining the data previously to in vitro and in vivo experiments. In this review, we depicted the current available animal-free alternatives in in vitro research.
Collapse
Affiliation(s)
- Ana C Duarte
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Elisabete C Costa
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Hugo A L Filipe
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sofia M Saraiva
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Telma Jacinto
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sónia P Miguel
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Maximiano P Ribeiro
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
23
|
Dzierżyńska M, Sawicka J, Deptuła M, Sosnowski P, Sass P, Peplińska B, Pietralik-Molińska Z, Fularczyk M, Kasprzykowski F, Zieliński J, Kozak M, Sachadyn P, Pikuła M, Rodziewicz-Motowidło S. Release systems based on self-assembling RADA16-I hydrogels with a signal sequence which improves wound healing processes. Sci Rep 2023; 13:6273. [PMID: 37072464 PMCID: PMC10113214 DOI: 10.1038/s41598-023-33464-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
Self-assembling peptides can be used for the regeneration of severely damaged skin. They can act as scaffolds for skin cells and as a reservoir of active compounds, to accelerate scarless wound healing. To overcome repeated administration of peptides which accelerate healing, we report development of three new peptide biomaterials based on the RADA16-I hydrogel functionalized with a sequence (AAPV) cleaved by human neutrophil elastase and short biologically active peptide motifs, namely GHK, KGHK and RDKVYR. The peptide hybrids were investigated for their structural aspects using circular dichroism, thioflavin T assay, transmission electron microscopy, and atomic force microscopy, as well as their rheological properties and stability in different fluids such as water or plasma, and their susceptibility to digestion by enzymes present in the wound environment. In addition, the morphology of the RADA-peptide hydrogels was examined with a unique technique called scanning electron cryomicroscopy. These experiments enabled us to verify if the designed peptides increased the bioactivity of the gel without disturbing its gelling processes. We demonstrate that the physicochemical properties of the designed hybrids were similar to those of the original RADA16-I. The materials behaved as expected, leaving the active motif free when treated with elastase. XTT and LDH tests on fibroblasts and keratinocytes were performed to assess the cytotoxicity of the RADA16-I hybrids, while the viability of cells treated with RADA16-I hybrids was evaluated in a model of human dermal fibroblasts. The hybrid peptides revealed no cytotoxicity; the cells grew and proliferated better than after treatment with RADA16-I alone. Improved wound healing following topical delivery of RADA-GHK and RADA-KGHK was demonstrated using a model of dorsal skin injury in mice and histological analyses. The presented results indicate further research is warranted into the engineered peptides as scaffolds for wound healing and tissue engineering.
Collapse
Affiliation(s)
- Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł Sosnowski
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Piotr Sass
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | | | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | | | - Jacek Zieliński
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
24
|
Sun W, Gregory DA, Zhao X. Designed peptide amphiphiles as scaffolds for tissue engineering. Adv Colloid Interface Sci 2023; 314:102866. [PMID: 36898186 DOI: 10.1016/j.cis.2023.102866] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David Alexander Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; Department of Material Science and Engineering, University of Sheffield, Sheffield S3 7HQ, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
25
|
Bashir MH, Korany NS, Farag DBE, Abbass MMS, Ezzat BA, Hegazy RH, Dörfer CE, Fawzy El-Sayed KM. Polymeric Nanocomposite Hydrogel Scaffolds in Craniofacial Bone Regeneration: A Comprehensive Review. Biomolecules 2023; 13:biom13020205. [PMID: 36830575 PMCID: PMC9953024 DOI: 10.3390/biom13020205] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
Nanocomposite biomaterials combine a biopolymeric matrix structure with nanoscale fillers. These bioactive and easily resorbable nanocomposites have been broadly divided into three groups, namely natural, synthetic or composite, based on the polymeric origin. Preparing such nanocomposite structures in the form of hydrogels can create a three-dimensional natural hydrophilic atmosphere pivotal for cell survival and new tissue formation. Thus, hydrogel-based cell distribution and drug administration have evolved as possible options for bone tissue engineering and regeneration. In this context, nanogels or nanohydrogels, created by cross-linking three-dimensional polymer networks, either physically or chemically, with high biocompatibility and mechanical properties were introduced as promising drug delivery systems. The present review highlights the potential of hydrogels and nanopolymers in the field of craniofacial tissue engineering and bone regeneration.
Collapse
Affiliation(s)
- Maha H. Bashir
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Nahed S. Korany
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Dina B. E. Farag
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Marwa M. S. Abbass
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Bassant A. Ezzat
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Radwa H. Hegazy
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Correspondence: ; Tel.: +49-431-500-26210
| |
Collapse
|
26
|
Haq-Siddiqi NA, Britton D, Kim Montclare J. Protein-engineered biomaterials for cartilage therapeutics and repair. Adv Drug Deliv Rev 2023; 192:114647. [PMID: 36509172 DOI: 10.1016/j.addr.2022.114647] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Cartilage degeneration and injury are major causes of pain and disability that effect millions, and yet treatment options for conditions like osteoarthritis (OA) continue to be mainly palliative or involve complete replacement of injured joints. Several biomaterial strategies have been explored to address cartilage repair either by the delivery of therapeutics or as support for tissue repair, however the complex structure of cartilage tissue, its mechanical needs, and lack of regenerative capacity have hindered this goal. Recent advances in synthetic biology have opened new possibilities for engineered proteins to address these unique needs. Engineered protein and peptide-based materials benefit from inherent biocompatibility and nearly unlimited tunability as they utilize the body's natural building blocks to fabricate a variety of supramolecular structures. The pathophysiology and needs of OA cartilage are presented here, along with an overview of the current state of the art and next steps for protein-engineered repair strategies for cartilage.
Collapse
Affiliation(s)
- Nada A Haq-Siddiqi
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Dustin Britton
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States; Department of Chemistry, New York University, New York 10003, United States; Department of Radiology, New York University Grossman School of Medicine, New York 10016, United States; Department of Biomaterials, NYU College of Dentistry, New York, NY 10010, United States; Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States.
| |
Collapse
|
27
|
Ouyang J, Sheng Y, Wang W. Recent Advances of Studies on Cell-Penetrating Peptides Based on Molecular Dynamics Simulations. Cells 2022; 11:cells11244016. [PMID: 36552778 PMCID: PMC9776715 DOI: 10.3390/cells11244016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
With the ability to transport cargo molecules across cell membranes with low toxicity, cell-penetrating peptides (CPPs) have become promising candidates for next generation peptide-based drug delivery vectors. Over the past three decades since the first CPP was discovered, a great deal of work has been done on the cellular uptake mechanisms and the applications for the delivery of therapeutic molecules, and significant advances have been made. But so far, we still do not have a precise and unified understanding of the structure-activity relationship of the CPPs. Molecular dynamics (MD) simulations provide a method to reveal peptide-membrane interactions at the atomistic level and have become an effective complement to experiments. In this paper, we review the progress of the MD simulations on CPP-membrane interactions, including the computational methods and technical improvements in the MD simulations, the research achievements in the CPP internalization mechanism, CPP decoration and coupling, and the peptide-induced membrane reactions during the penetration process, as well as the comparison of simulated and experimental results.
Collapse
Affiliation(s)
- Jun Ouyang
- School of Public Courses, Bengbu Medical College, Bengbu 233030, China
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Yuebiao Sheng
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- High Performance Computing Center, Nanjing University, Nanjing 210093, China
- Correspondence: (Y.S.); (W.W.)
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- Correspondence: (Y.S.); (W.W.)
| |
Collapse
|
28
|
Le X, Gao T, Wang L, Wei F, Chen C, Zhao Y. Self-Assembly of Short Amphiphilic Peptides and Their Biomedical Applications. Curr Pharm Des 2022; 28:3546-3562. [PMID: 36424793 DOI: 10.2174/1381612829666221124103526] [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: 05/30/2022] [Revised: 10/22/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022]
Abstract
A series of functional biomaterials with different sizes and morphologies can be constructed through self-assembly, among which amphiphilic peptide-based materials have received intense attention. One main possible reason is that the short amphiphilic peptides can facilitate the formation of versatile materials and promote their further applications in different fields. Another reason is that the simple structure of amphiphilic peptides can help establish the structure-function relationship. This review highlights the recent advances in the self-assembly of two typical peptide species, surfactant-like peptides (SLPs) and peptides amphiphiles (PAs). These peptides can self-assemble into diverse nanostructures. The formation of these different nanostructures resulted from the delicate balance of varied non-covalent interactions. This review embraced each non-covalent interaction and then listed the typical routes for regulating these non-covalent interactions, then realized the morphologies modulation of the self-assemblies. Finally, their applications in some biomedical fields, such as the stabilization of membrane proteins, templating for nanofabrication and biomineralization, acting as the antibacterial and antitumor agents, hemostasis, and synthesis of melanin have been summarized. Further advances in the self-assembly of SLPs and PAs may focus on the design of functional materials with targeted properties and exploring their improved properties.
Collapse
Affiliation(s)
- Xiaosong Le
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| | - Tianwen Gao
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| | - Li Wang
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| | - Feng Wei
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| | - Cuixia Chen
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| | - Yurong Zhao
- State Key Laboratory of Heavy Oil Processing and the Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao266580, China
| |
Collapse
|
29
|
Advancements in Hydrogel Application for Ischemic Stroke Therapy. Gels 2022; 8:gels8120777. [PMID: 36547301 PMCID: PMC9778209 DOI: 10.3390/gels8120777] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability worldwide. There is almost no effective treatment for this disease. Therefore, developing effective treatment for ischemic stroke is urgently needed. Efficient delivery of therapeutic drugs to ischemic sites remained a great challenge for improved treatment of strokes. In recent years, hydrogel-based strategies have been widely investigated for new and improved therapies. They have the advantage of delivering therapeutics in a controlled manner to the poststroke sites, aiming to enhance the intrinsic repair and regeneration. In this review, we discuss the pathophysiology of stroke and the development of injectable hydrogels in the application of both stroke treatment and neural tissue engineering. We also discuss the prospect and the challenges of hydrogels in the treatment of ischemic strokes.
Collapse
|
30
|
Xiao X, Robang AS, Sarma S, Le JV, Helmicki ME, Lambert MJ, Guerrero-Ferreira R, Arboleda-Echavarria J, Paravastu AK, Hall CK. Sequence patterns and signatures: Computational and experimental discovery of amyloid-forming peptides. PNAS NEXUS 2022; 1:pgac263. [PMID: 36712347 PMCID: PMC9802472 DOI: 10.1093/pnasnexus/pgac263] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Screening amino acid sequence space via experiments to discover peptides that self-assemble into amyloid fibrils is challenging. We have developed a computational peptide assembly design (PepAD) algorithm that enables the discovery of amyloid-forming peptides. Discontinuous molecular dynamics (DMD) simulation with the PRIME20 force field combined with the FoldAmyloid tool is used to examine the fibrilization kinetics of PepAD-generated peptides. PepAD screening of ∼10,000 7-mer peptides resulted in twelve top-scoring peptides with two distinct hydration properties. Our studies revealed that eight of the twelve in silico discovered peptides spontaneously form amyloid fibrils in the DMD simulations and that all eight have at least five residues that the FoldAmyloid tool classifies as being aggregation-prone. Based on these observations, we re-examined the PepAD-generated peptides in the sequence pool returned by PepAD and extracted five sequence patterns as well as associated sequence signatures for the 7-mer amyloid-forming peptides. Experimental results from Fourier transform infrared spectroscopy (FTIR), thioflavin T (ThT) fluorescence, circular dichroism (CD), and transmission electron microscopy (TEM) indicate that all the peptides predicted to assemble in silico assemble into antiparallel β-sheet nanofibers in a concentration-dependent manner. This is the first attempt to use a computational approach to search for amyloid-forming peptides based on customized settings. Our efforts facilitate the identification of β-sheet-based self-assembling peptides, and contribute insights towards answering a fundamental scientific question: "What does it take, sequence-wise, for a peptide to self-assemble?".
Collapse
Affiliation(s)
| | | | | | - Justin V Le
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Michael E Helmicki
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Matthew J Lambert
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ricardo Guerrero-Ferreira
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Johana Arboleda-Echavarria
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
31
|
Chowdhary S, Schmidt RF, Sahoo AK, Tom Dieck T, Hohmann T, Schade B, Brademann-Jock K, Thünemann AF, Netz RR, Gradzielski M, Koksch B. Rational design of amphiphilic fluorinated peptides: evaluation of self-assembly properties and hydrogel formation. NANOSCALE 2022; 14:10176-10189. [PMID: 35796261 DOI: 10.1039/d2nr01648f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Advanced peptide-based nanomaterials composed of self-assembling peptides (SAPs) are of emerging interest in pharmaceutical and biomedical applications. The introduction of fluorine into peptides, in fact, offers unique opportunities to tune their biophysical properties and intermolecular interactions. In particular, the degree of fluorination plays a crucial role in peptide engineering as it can be used to control the characteristics of fluorine-specific interactions and, thus, peptide conformation and self-assembly. Here, we designed and explored a series of amphipathic peptides by incorporating the fluorinated amino acids (2S)-4-monofluoroethylglycine (MfeGly), (2S)-4,4-difluoroethylglycine (DfeGly) and (2S)-4,4,4-trifluoroethylglycine (TfeGly) as hydrophobic components. This approach enabled studying the impact of fluorination on secondary structure formation and peptide self-assembly on a systematic basis. We show that the interplay between polarity and hydrophobicity, both induced differentially by varying degrees of side chain fluorination, does affect peptide folding significantly. A greater degree of fluorination promotes peptide fibrillation and subsequent formation of physical hydrogels in physiological conditions. Molecular simulations revealed the key role played by electrostatically driven intra-chain and inter-chain contact pairs that are modulated by side chain fluorination and give insights into the different self-organization behaviour of selected peptides. Our study provides a systematic report about the distinct features of fluorinated oligomeric peptides with potential applications as peptide-based biomaterials.
Collapse
Affiliation(s)
- Suvrat Chowdhary
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 20, 14195 Berlin, Germany.
| | - Robert Franz Schmidt
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 124, 10623 Berlin, Germany
| | - Anil Kumar Sahoo
- Department of Physics, Freie Universität Berlin, Arnimallee 14, 14195 Berlin, Germany
- Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Tiemo Tom Dieck
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 20, 14195 Berlin, Germany.
| | - Thomas Hohmann
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 20, 14195 Berlin, Germany.
| | - Boris Schade
- Institute of Chemistry and Biochemistry and Core Facility BioSupraMol, Freie Universität Berlin, Fabeckstraße 36a, 14195 Berlin, Germany
| | - Kerstin Brademann-Jock
- Federal Institute for Materials Research and Testing (BAM), Unter den Eichen 87, 12205 Berlin, Germany
| | - Andreas F Thünemann
- Federal Institute for Materials Research and Testing (BAM), Unter den Eichen 87, 12205 Berlin, Germany
| | - Roland R Netz
- Department of Physics, Freie Universität Berlin, Arnimallee 14, 14195 Berlin, Germany
| | - Michael Gradzielski
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 124, 10623 Berlin, Germany
| | - Beate Koksch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 20, 14195 Berlin, Germany.
| |
Collapse
|
32
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
33
|
Tanaka T, Kuroiwa K. Supramolecular Hybrids from Cyanometallate Complexes and Diblock Copolypeptide Amphiphiles in Water. Molecules 2022; 27:3262. [PMID: 35630738 PMCID: PMC9143414 DOI: 10.3390/molecules27103262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022] Open
Abstract
The self-assembly of discrete cyanometallates has attracted significant interest due to the potential of these materials to undergo soft metallophilic interactions as well as their optical properties. Diblock copolypeptide amphiphiles have also been investigated concerning their capacity for self-assembly into morphologies such as nanostructures. The present work combined these two concepts by examining supramolecular hybrids comprising cyanometallates with diblock copolypeptide amphiphiles in aqueous solutions. Discrete cyanometallates such as [Au(CN)2]-, [Ag(CN)2]-, and [Pt(CN)4]2- dispersed at the molecular level in water cannot interact with each other at low concentrations. However, the results of this work demonstrate that the addition of diblock copolypeptide amphiphiles such as poly-(L-lysine)-block-(L-cysteine) (Lysm-b-Cysn) to solutions of these complexes induces the supramolecular assembly of the discrete cyanometallates, resulting in photoluminescence originating from multinuclear complexes with metal-metal interactions. Electron microscopy images confirmed the formation of nanostructures of several hundred nanometers in size that grew to form advanced nanoarchitectures, including those resembling the original nanostructures. This concept of combining diblock copolypeptide amphiphiles with discrete cyanometallates allows the design of flexible and functional supramolecular hybrid systems in water.
Collapse
Affiliation(s)
| | - Keita Kuroiwa
- Department of Nanoscience, Faculty of Engineering, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan;
| |
Collapse
|
34
|
Li T, Lu XM, Zhang MR, Hu K, Li Z. Peptide-based nanomaterials: Self-assembly, properties and applications. Bioact Mater 2022; 11:268-282. [PMID: 34977431 PMCID: PMC8668426 DOI: 10.1016/j.bioactmat.2021.09.029] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 11/24/2022] Open
Abstract
Peptide-based materials that have diverse structures and functionalities are an important type of biomaterials. In former times, peptide-based nanomaterials with excellent stability were constructed through self-assembly. Compared with individual peptides, peptide-based self-assembly nanomaterials that form well-ordered superstructures possess many advantages such as good thermo- and mechanical stability, semiconductivity, piezoelectricity and optical properties. Moreover, due to their excellent biocompatibility and biological activity, peptide-based self-assembly nanomaterials have been vastly used in different fields. In this review, we provide the advances of peptide-based self-assembly nanostructures, focusing on the driving forces that dominate peptide self-assembly and assembly mechanisms of peptides. After that, we outline the synthesis and properties of peptide-based nanomaterials, followed by the applications of functional peptide nanomaterials. Finally, we provide perspectives on the challenges and future of peptide-based nanomaterials.
Collapse
Affiliation(s)
- Tong Li
- College of Chemistry and Chemical Engineering, Center of Nanoenergy Research, Guangxi University, Nanning, 530004, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Xian-Mao Lu
- College of Chemistry and Chemical Engineering, Center of Nanoenergy Research, Guangxi University, Nanning, 530004, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, The National Institute of Radiological Sciences, The National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Kuan Hu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Department of Advanced Nuclear Medicine Sciences, The National Institute of Radiological Sciences, The National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Zhou Li
- College of Chemistry and Chemical Engineering, Center of Nanoenergy Research, Guangxi University, Nanning, 530004, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 101400, China
| |
Collapse
|
35
|
Yang Y, Hao S, Lei X, Chen J, Fang G, Liu J, Wang S, He X. Design of metalloenzyme mimics based on self-assembled peptides for organophosphorus pesticides detection. JOURNAL OF HAZARDOUS MATERIALS 2022; 428:128262. [PMID: 35051771 DOI: 10.1016/j.jhazmat.2022.128262] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Organophosphorus pesticides (OPs) detection has attracted considerable attention because of the extensive application of OPs. In this research, non-toxic and high-performance metalloenzyme mimics of Zn2+-bonding peptides were developed by obtaining inspiration from phosphotriesterase (PTE) and nanofiber formation. Furthermore, based on the electrochemical activity of p-nitrophenol (PNP), the electrochemical sensor of metalloenzyme mimics was developed. By examining the effect of the active sites of peptides and fibril formation on the degradation of OPs, the optimal metalloenzyme mimic was selected. Furthermore, optimal metalloenzyme mimics were combined with NiCo2O4 to develop an electrochemical sensor of OPs. By monitoring square wave voltammetry (SWV) signals of PNP degraded from OPs, the amounts of OPs in actual samples could be determined in 15 min. We discovered that both the active sites of α metal and β metal were required for metalloenzyme mimics; Zn2+ promoted peptide fibrosis and especially acted as a cofactor for degrading OPs. Compared to traditional methods, the electrochemical sensor of metalloenzyme mimics was sensitive, reliable, and non-toxic; furthermore, the detection limit of methyl paraoxon was as low as 0.08 µM. The metalloenzyme mimics will be a promising material for detecting OPs in the food industry and environment fields.
Collapse
Affiliation(s)
- Yayu Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Sijia Hao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xiangmin Lei
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Jianan Chen
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Guozhen Fang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Jifeng Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China.
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China; Research Center of Food Science and Human Health, School of Medicine, Nankai University, Tianjin 300071, PR China.
| | - Xingxing He
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, PR China
| |
Collapse
|
36
|
Acidic and basic self-assembling peptide and peptide-graphene oxide hydrogels: characterisation and effect on encapsulated nucleus pulposus cells. Acta Biomater 2022; 143:145-158. [PMID: 35196554 DOI: 10.1016/j.actbio.2022.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 12/28/2022]
Abstract
Extracellular pH can have a profound effect on cell metabolism, gene and protein expression. Nucleus pulposus (NP) cells, for example, under acidic conditions accelerate the production of degradative enzymes and pro-inflammatory cytokines, leading ultimately to intervertebral disc degeneration, a major cause of back pain. Self-assembling peptide hydrogels constitute a well-established class of biomaterials that could be exploited as pH-tunable platform to investigate cell behaviour under normal and non-physiological pH. In this paper we formulated acidic (pH = 4) and basic (pH = 9) hydrogels, from the same octapeptide FEFKFEFK (F8) (F = phenyalanine, E = glutamic acid, K = lysine), to test the effect of non-physiological pH on encapsulated NP cells. Similarly, graphene oxide-containing F8 hydrogels (GO-F8) were formulated as stiffer analogues. Acidic and basic hydrogels showed peculiar morphologies and rheological properties, with all systems able to buffer within 30 minutes of exposure to cell culture media. NP cells seeded in acidic F8 hydrogels showed a more catabolic phenotype compared to basic hydrogels, with increased gene expression of degradative enzymes (MMP-3, ADAMTS-4), neurotrophic factors (NGF and BDNF) and NF-κB p65 phosphorylation. Acidic GO-F8 hydrogels also induced a catabolic response, although milder than basic counterparts and with the highest gene expression of characteristic NP-matrix components, aggrecan and collagen II. In all systems, the cellular response had a peak within 3 days of encapsulation, thereafter decreasing over 7 days, suggesting a 'transitory' effect of hydrogel pH on encapsulated cells. This work gives an insight on the effect of pH (and pH buffering) on encapsulated NP cells and offers new designs of low and high pH peptide hydrogels for 3D cell culture studies. STATEMENT OF SIGNIFICANCE: We have recently shown the potential of graphene oxide - self-assembling peptide hybrid hydrogels for NP cell culture and regeneration. Alongside cell carrier, self-assembling peptide hydrogels actually provide a versatile pH-tunable platform for biological studies. In this work we decided to explore the effect of non-physiological pH (and pH buffering) on encapsulated NP cells. Our approach allows the formulation of both acidic and basic hydrogels, starting from the same peptide sequence. We showed that the initial pH of the scaffold does not affect significantly cell response to encapsulation, but the presence of GO results in lower inflammatory levels and higher NP matrix protein production. This platform could be exploited to study the effect of pH on different cell types whose behaviour can be pH-dependent.
Collapse
|
37
|
Wang XJ, Cheng J, Zhang LY, Zhang JG. Self-assembling peptides-based nano-cargos for targeted chemotherapy and immunotherapy of tumors: recent developments, challenges, and future perspectives. Drug Deliv 2022; 29:1184-1200. [PMID: 35403517 PMCID: PMC9004497 DOI: 10.1080/10717544.2022.2058647] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xue-Jun Wang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jian Cheng
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| | - Le-Yi Zhang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jun-Gang Zhang
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
38
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
39
|
Ligorio C, Hoyland JA, Saiani A. Self-Assembling Peptide Hydrogels as Functional Tools to Tackle Intervertebral Disc Degeneration. Gels 2022; 8:gels8040211. [PMID: 35448112 PMCID: PMC9028266 DOI: 10.3390/gels8040211] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
Low back pain (LBP), caused by intervertebral disc (IVD) degeneration, is a major contributor to global disability. In its healthy state, the IVD is a tough and well-hydrated tissue, able to act as a shock absorber along the spine. During degeneration, the IVD is hit by a cell-driven cascade of events, which progressively lead to extracellular matrix (ECM) degradation, chronic inflammation, and pain. Current treatments are divided into palliative care (early stage degeneration) and surgical interventions (late-stage degeneration), which are invasive and poorly efficient in the long term. To overcome these limitations, alternative tissue engineering and regenerative medicine strategies, in which soft biomaterials are used as injectable carriers of cells and/or biomolecules to be delivered to the injury site and restore tissue function, are currently being explored. Self-assembling peptide hydrogels (SAPHs) represent a promising class of de novo synthetic biomaterials able to merge the strengths of both natural and synthetic hydrogels for biomedical applications. Inherent features, such as shear-thinning behaviour, high biocompatibility, ECM biomimicry, and tuneable physiochemical properties make these hydrogels appropriate and functional tools to tackle IVD degeneration. This review will describe the pathogenesis of IVD degeneration, list biomaterials requirements to attempt IVD repair, and focus on current peptide hydrogel materials exploited for this purpose.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 3BB, UK;
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PG, UK;
- Correspondence:
| | - Judith A. Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PG, UK;
| | - Alberto Saiani
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 3BB, UK;
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
40
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
41
|
Boosted Cross-Linking and Characterization of High-Performing Self-Assembling Peptides. NANOMATERIALS 2022; 12:nano12030320. [PMID: 35159664 PMCID: PMC8838902 DOI: 10.3390/nano12030320] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/10/2022]
Abstract
Tissue engineering (TE) strategies require the design and characterization of novel biomaterials capable of mimicking the physiological microenvironments of the tissues to be regenerated. As such, implantable materials should be biomimetic, nanostructured and with mechanical properties approximating those of the target organ/tissue. Self-assembling peptides (SAPs) are biomimetic nanomaterials that can be readily synthesized and customized to match the requirements of some TE applications, but the weak interactions involved in the self-assembling phenomenon make them soft hydrogels unsuited for the regeneration of medium-to-hard tissues. In this work, we moved significant steps forward in the field of chemical cross-linked SAPs towards the goal of stiff peptidic materials suited for the regeneration of several tissues. Novel SAPs were designed and characterized to boost the 4-(N-Maleimidomethyl) cyclohexane-1-carboxylic acid 3-sulpho-N-hydroxysuccinimide ester (Sulfo-SMCC) mediated cross-linking reaction, where they reached G′ values of ~500 kPa. An additional orthogonal cross-linking was also effective and allowed to top remarkable G′ values of 840 kPa. We demonstrated that cross-linking fastened the pre-existing self-aggregated nanostructures, and at the same time, a strong presence of ß-structures is necessary for an effective cross-linking of (LKLK)3-based SAPs. Combining strong SAP design and orthogonal cross-linking reactions, we brought SAP stiffness closer to the MPa threshold, and as such, we opened the door of the regeneration of skin, muscle and lung to biomimetic SAP technology.
Collapse
|
42
|
Trubelja A, Kasper FK, Farach-Carson MC, Harrington DA. Bringing hydrogel-based craniofacial therapies to the clinic. Acta Biomater 2022; 138:1-20. [PMID: 34743044 PMCID: PMC9234983 DOI: 10.1016/j.actbio.2021.10.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 01/17/2023]
Abstract
This review explores the evolution of the use of hydrogels for craniofacial soft tissue engineering, ranging in complexity from acellular injectable fillers to fabricated, cell-laden constructs with complex compositions and architectures. Addressing both in situ and ex vivo approaches, tissue restoration secondary to trauma or tumor resection is discussed. Beginning with relatively simple epithelia of oral mucosa and gingiva, then moving to more functional units like vocal cords or soft tissues with multilayer branched structures, such as salivary glands, various approaches are presented toward the design of function-driven architectures, inspired by native tissue organization. Multiple tissue replacement paradigms are presented here, including the application of hydrogels as structural materials and as delivery platforms for cells and/or therapeutics. A practical hierarchy is proposed for hydrogel systems in craniofacial applications, based on their material and cellular complexity, spatial order, and biological cargo(s). This hierarchy reflects the regulatory complexity dictated by the Food and Drug Administration (FDA) in the United States prior to commercialization of these systems for use in humans. The wide array of available biofabrication methods, ranging from simple syringe extrusion of a biomaterial to light-based spatial patterning for complex architectures, is considered within the history of FDA-approved commercial therapies. Lastly, the review assesses the impact of these regulatory pathways on the translational potential of promising pre-clinical technologies for craniofacial applications. STATEMENT OF SIGNIFICANCE: While many commercially available hydrogel-based products are in use for the craniofacial region, most are simple formulations that either are applied topically or injected into tissue for aesthetic purposes. The academic literature previews many exciting applications that harness the versatility of hydrogels for craniofacial soft tissue engineering. One of the most exciting developments in the field is the emergence of advanced biofabrication methods to design complex hydrogel systems that can promote the functional or structural repair of tissues. To date, no clinically available hydrogel-based therapy takes full advantage of current pre-clinical advances. This review surveys the increasing complexity of the current landscape of available clinical therapies and presents a framework for future expanded use of hydrogels with an eye toward translatability and U.S. regulatory approval for craniofacial applications.
Collapse
Affiliation(s)
- Alen Trubelja
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - F Kurtis Kasper
- Department of Orthodontics, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States; Department of BioSciences, Rice University, Houston, TX 77005, United States
| | - Daniel A Harrington
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States; Department of BioSciences, Rice University, Houston, TX 77005, United States.
| |
Collapse
|
43
|
Tian H, Guo A, Li K, Tao B, Lei D, Deng Z. Effects of a novel self-assembling peptide scaffold on bone regeneration and controlled release of two growth factors. J Biomed Mater Res A 2021; 110:943-953. [PMID: 34873824 DOI: 10.1002/jbm.a.37342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/12/2021] [Accepted: 11/28/2021] [Indexed: 12/16/2022]
Abstract
RADA16 is a self-assembling peptide material with good bioactivity. To improve the bioactivity of a material, some specific functional motifs can be added to its peptide sequence. Here, we report a self-assembling peptide nanogel, RADA16-RGD, that has better bioactivity than RADA16 and can simultaneously carry and control the release of two growth factors, VEGF and BMP-2, which have synergistic effects on bone formation. The peptide materials were characterized by transmission electron microscopy and scanning electron microscopy. The mechanical properties of the peptides were evaluated by the rheology test. The biocompatibility of the materials was evaluated via the use of the CCK-8 test, live/dead staining and confocal laser scanning microscopy. Osteogenesis capability in vitro was evaluated by means of ALP staining, extracellular matrix mineralization and detection of osteogenic markers. The controlled release of growth factors was examined by ELISA. The results showed that RADA16-RGD exhibited a better ability than RADA16 to promote cell proliferation, adhesion and bone formation. In addition, RADA16-RGD had good biocompatibility and exhibited effective controlled release of VEGF and BMP-2. More importantly, compared with RADA16-RGD loaded with single growth factor or without growth factors, RADA16-RGD loaded with two growth factors exhibited a stronger ability to promote cell proliferation and osteogenesis. This study provides a promising strategy for the application of self-assembling peptides to promote osteogenesis and controlled release of proteins.
Collapse
Affiliation(s)
- Hongchuan Tian
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ai Guo
- Department of Orthopaedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Li
- Department of Orthopaedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dengliang Lei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongliang Deng
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Das S, Das D. Rational Design of Peptide-based Smart Hydrogels for Therapeutic Applications. Front Chem 2021; 9:770102. [PMID: 34869218 PMCID: PMC8635208 DOI: 10.3389/fchem.2021.770102] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Peptide-based hydrogels have captivated remarkable attention in recent times and serve as an excellent platform for biomedical applications owing to the impressive amalgamation of unique properties such as biocompatibility, biodegradability, easily tunable hydrophilicity/hydrophobicity, modular incorporation of stimuli sensitivity and other functionalities, adjustable mechanical stiffness/rigidity and close mimicry to biological molecules. Putting all these on the same plate offers smart soft materials that can be used for tissue engineering, drug delivery, 3D bioprinting, wound healing to name a few. A plethora of work has been accomplished and a significant progress has been realized using these peptide-based platforms. However, designing hydrogelators with the desired functionalities and their self-assembled nanostructures is still highly serendipitous in nature and thus a roadmap providing guidelines toward designing and preparing these soft-materials and applying them for a desired goal is a pressing need of the hour. This review aims to provide a concise outline for that purpose and the design principles of peptide-based hydrogels along with their potential for biomedical applications are discussed with the help of selected recent reports.
Collapse
Affiliation(s)
- Saurav Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| | - Debapratim Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
45
|
Sidorova A, Bystrov V, Lutsenko A, Shpigun D, Belova E, Likhachev I. Quantitative Assessment of Chirality of Protein Secondary Structures and Phenylalanine Peptide Nanotubes. NANOMATERIALS 2021; 11:nano11123299. [PMID: 34947648 PMCID: PMC8707344 DOI: 10.3390/nano11123299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 01/25/2023]
Abstract
In this study we consider the features of spatial-structure formation in proteins and their application in bioengineering. Methods for the quantitative assessment of the chirality of regular helical and irregular structures of proteins are presented. The features of self-assembly of phenylalanine (F) into peptide nanotubes (PNT), which form helices of different chirality, are also analyzed. A method is proposed for calculating the magnitude and sign of the chirality of helix-like peptide nanotubes using a sequence of vectors for the dipole moments of individual peptides.
Collapse
Affiliation(s)
- Alla Sidorova
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.L.); (D.S.); (E.B.)
- Correspondence:
| | - Vladimir Bystrov
- Institute of Mathematical Problems of Biology, The Branch of Keldysh Institute of Applied Mathematics, RAS, 142290 Pushchino, Russia; (V.B.); (I.L.)
| | - Aleksey Lutsenko
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.L.); (D.S.); (E.B.)
| | - Denis Shpigun
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.L.); (D.S.); (E.B.)
| | - Ekaterina Belova
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.L.); (D.S.); (E.B.)
| | - Ilya Likhachev
- Institute of Mathematical Problems of Biology, The Branch of Keldysh Institute of Applied Mathematics, RAS, 142290 Pushchino, Russia; (V.B.); (I.L.)
| |
Collapse
|
46
|
Root-Bernstein R, Churchill B. Co-Evolution of Opioid and Adrenergic Ligands and Receptors: Shared, Complementary Modules Explain Evolution of Functional Interactions and Suggest Novel Engineering Possibilities. Life (Basel) 2021; 11:life11111217. [PMID: 34833093 PMCID: PMC8623292 DOI: 10.3390/life11111217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Cross-talk between opioid and adrenergic receptors is well-characterized and involves second messenger systems, the formation of receptor heterodimers, and the presence of extracellular allosteric binding regions for the complementary ligand; however, the evolutionary origins of these interactions have not been investigated. We propose that opioid and adrenergic ligands and receptors co-evolved from a common set of modular precursors so that they share binding functions. We demonstrate the plausibility of this hypothesis through a review of experimental evidence for molecularly complementary modules and report unexpected homologies between the two receptor types. Briefly, opioids form homodimers also bind adrenergic compounds; opioids bind to conserved extracellular regions of adrenergic receptors while adrenergic compounds bind to conserved extracellular regions of opioid receptors; opioid-like modules appear in both sets of receptors within key ligand-binding regions. Transmembrane regions associated with homodimerization of each class of receptors are also highly conserved across receptor types and implicated in heterodimerization. This conservation of multiple functional modules suggests opioid–adrenergic ligand and receptor co-evolution and provides mechanisms for explaining the evolution of their crosstalk. These modules also suggest the structure of a primordial receptor, providing clues for engineering receptor functions.
Collapse
|
47
|
Muraoka T. Amphiphilic Peptides with Flexible Chains for Tuning Supramolecular Morphologies, Macroscopic Properties and Biological Functions. J SYN ORG CHEM JPN 2021. [DOI: 10.5059/yukigoseikyokaishi.79.1033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
48
|
The biological applications of DNA nanomaterials: current challenges and future directions. Signal Transduct Target Ther 2021; 6:351. [PMID: 34620843 PMCID: PMC8497566 DOI: 10.1038/s41392-021-00727-9] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/24/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023] Open
Abstract
DNA, a genetic material, has been employed in different scientific directions for various biological applications as driven by DNA nanotechnology in the past decades, including tissue regeneration, disease prevention, inflammation inhibition, bioimaging, biosensing, diagnosis, antitumor drug delivery, and therapeutics. With the rapid progress in DNA nanotechnology, multitudinous DNA nanomaterials have been designed with different shape and size based on the classic Watson-Crick base-pairing for molecular self-assembly. Some DNA materials could functionally change cell biological behaviors, such as cell migration, cell proliferation, cell differentiation, autophagy, and anti-inflammatory effects. Some single-stranded DNAs (ssDNAs) or RNAs with secondary structures via self-pairing, named aptamer, possess the ability of targeting, which are selected by systematic evolution of ligands by exponential enrichment (SELEX) and applied for tumor targeted diagnosis and treatment. Some DNA nanomaterials with three-dimensional (3D) nanostructures and stable structures are investigated as drug carrier systems to delivery multiple antitumor medicine or gene therapeutic agents. While the functional DNA nanostructures have promoted the development of the DNA nanotechnology with innovative designs and preparation strategies, and also proved with great potential in the biological and medical use, there is still a long way to go for the eventual application of DNA materials in real life. Here in this review, we conducted a comprehensive survey of the structural development history of various DNA nanomaterials, introduced the principles of different DNA nanomaterials, summarized their biological applications in different fields, and discussed the current challenges and further directions that could help to achieve their applications in the future.
Collapse
|
49
|
Marchini A, Gelain F. Synthetic scaffolds for 3D cell cultures and organoids: applications in regenerative medicine. Crit Rev Biotechnol 2021; 42:468-486. [PMID: 34187261 DOI: 10.1080/07388551.2021.1932716] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional (3D) cell cultures offer an unparalleled platform to recreate spatial arrangements of cells in vitro. 3D cell culture systems have gained increasing interest due to their evident advantages in providing more physiologically relevant information and more predictive data compared to their two-dimensional (2D) counterpart. Design and well-established fabrication of organoids (a particular type of 3D cell culture system) are nowadays considered a pivotal achievement for their ability to replicate in vitro cytoarchitecture and the functionalities of an organ. In this condition, pluripotent stem cells self-organize into 3D structures mimicking the in vivo microenvironments, architectures and multi-lineage differentiation. Scaffolds are key supporting elements in these 3D constructs, and Matrigel is the most commonly used matrix despite its relevant translation limitations including animal-derived sources, non-defined composition, batch-to-batch variability and poorly tailorable properties. Alternatively, 3D synthetic scaffolds, including self-assembling peptides, show promising biocompatibility and biomimetic properties, and can be tailored on specific target tissue/cells. In this review, we discuss the recent advances on 3D cell culture systems and organoids, promising tools for tissue engineering and regenerative medicine applications. For this purpose, we will describe the current state-of-the-art on 3D cell culture systems and organoids based on currently available synthetic-based biomaterials (including tailored self-assembling peptides) either tested in in vivo animal models or developed in vitro with potential application in the field of tissue engineering, with the aim to inspire researchers to take on such promising platforms for clinical applications in the near future.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
50
|
Di Foggia M, Tugnoli V, Ottani S, Dettin M, Zamuner A, Sanchez-Cortes S, Cesini D, Torreggiani A. SERS Investigation on Oligopeptides Used as Biomimetic Coatings for Medical Devices. Biomolecules 2021; 11:959. [PMID: 34209793 PMCID: PMC8301923 DOI: 10.3390/biom11070959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 11/22/2022] Open
Abstract
The surface-enhanced Raman scattering (SERS) spectra of three amphiphilic oligopeptides derived from EAK16 (AEAEAKAK)2 were examined to study systematic amino acid substitution effects on the corresponding interaction with Ag colloidal nanoparticles. Such self-assembling molecular systems, known as "molecular Lego", are of particular interest for their uses in tissue engineering and as biomimetic coatings for medical devices because they can form insoluble macroscopic membranes under physiological conditions. Spectra were collected for both native and gamma-irradiated samples. Quantum mechanical data on two of the examined oligopeptides were also obtained to clarify the assignment of the prominent significative bands observed in the spectra. In general, the peptide-nanoparticles interaction occurs through the COO- groups, with the amide bond and the aliphatic chain close to the colloid surface. After gamma irradiation, mimicking a free oxidative radical attack, the SERS spectra of the biomaterials show that COO- groups still provide the main peptide-nanoparticle interactions. However, the spatial arrangement of the peptides is different, exhibiting a systematic decrease in the distance between aliphatic chains and colloid nanoparticles.
Collapse
Affiliation(s)
- Michele Di Foggia
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40126 Bologna, Italy;
| | - Vitaliano Tugnoli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40126 Bologna, Italy;
| | - Stefano Ottani
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche (ISOF-CNR), 40129 Bologna, Italy; (S.O.); (A.T.)
| | - Monica Dettin
- Dipartimento di Ingegneria Industriale, Università di Padova, 35131 Padova, Italy; (M.D.); (A.Z.)
| | - Annj Zamuner
- Dipartimento di Ingegneria Industriale, Università di Padova, 35131 Padova, Italy; (M.D.); (A.Z.)
| | - Santiago Sanchez-Cortes
- Instituto de Estructura de la Materia, Consejo Superior de Investigaciones Cientificas (CSIC), 28006 Madrid, Spain;
| | - Daniele Cesini
- Dipartimento CNAF, Istituto Nazionale di Fisica Nucleare (INFN-CNAF), 40127 Bologna, Italy;
| | - Armida Torreggiani
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche (ISOF-CNR), 40129 Bologna, Italy; (S.O.); (A.T.)
| |
Collapse
|