1
|
Peura A, Turpin R, Liu R, Heilala M, Salmela M, Aung J, Mikkonen P, Mutka M, Kovanen PE, Niinikoski L, Meretoja T, Mattson J, Heikkilä P, Palanne P, Kantanen T, Kilpeläinen M, Ukkonen O, Hollmén M, Tervonen TA, Klefström J, Munne PM. Soft matrix promotes immunosuppression in tumor-resident immune cells via COX-FGF2 signaling. Nat Commun 2025; 16:4908. [PMID: 40425576 PMCID: PMC12116891 DOI: 10.1038/s41467-025-60092-x] [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: 04/26/2024] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanical forces of the tumor microenvironment change dynamically during key events of tumorigenesis such as invasion and metastasis. These changes in compressive forces often affect the breast cancer cell phenotype. However, it is lesser known how these dynamic mechanical forces in the tumor microenvironment affect the phenotypes of tumor infiltrated leukocytes (TIL) and their subsequent anticancer activities. Here we find, in primary patient-derived explant cultures (PDEC) containing resident TILs, that low compression promotes a change in the original identity of breast cancer cells from luminal to a more mesenchymal and undifferentiated state. These altered tumor cells induce an upregulation of immunosuppressive cytokines such as interleukin-10 (IL-10) and Transforming Growth Factor Beta (TGF-β), as well as polarization of macrophages towards pro-tumor M2(Gc)-type and depletion of CD8+ effector memory T-cells. These immunosuppressive events are mediated by tumor cell derived fibroblast growth factor 2 (FGF2) and prostaglandin E2 (PGE2). We also find that FGF2 rich areas in primary tumors show enrichment in M2-like-macrophages and diminished numbers of CD8 + T and B-cells. Our results suggest that low compressive forces in the tumor microenvironment induce local immunosuppression via FGF2 secretion arising from phenotypic plasticity of tumor cells.
Collapse
Affiliation(s)
- Aino Peura
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Rita Turpin
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, Turku, Finland
| | - Ruixian Liu
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Maria Heilala
- Department of Applied Physics, Aalto University, Espoo, Finland
| | - Maria Salmela
- Finnish Genome Editing Center, HiLIFE infrastructures, University of Helsinki and Biocenter Finland, Helsinki, Finland
| | - July Aung
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
| | - Piia Mikkonen
- UPM Biomedicals, UPM-Kymmene Corporation, Helsinki, Finland
| | - Minna Mutka
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Panu E Kovanen
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Laura Niinikoski
- Breast Surgery Unit, Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tuomo Meretoja
- Breast Surgery Unit, Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Johanna Mattson
- Comprehensive Cancer Center, University of Helsinki & Helsinki University Hospital, Helsinki, Finland
| | - Päivi Heikkilä
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Palanne
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Tiina Kantanen
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko Kilpeläinen
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Outi Ukkonen
- Department of Surgery, Kymenlaakso Central Hospital, KYMSOTE, Kotka, Finland
| | - Maija Hollmén
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, Turku, Finland
| | - Topi A Tervonen
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland
- Finnish Genome Editing Center, HiLIFE infrastructures, University of Helsinki and Biocenter Finland, Helsinki, Finland
| | - Juha Klefström
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland.
- Finnish Cancer Institute, Helsinki, Finland.
- FICAN South, Helsinki University Hospital, Helsinki, Finland.
- Department of Cell & Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue, UCSF Campus, San Francisco, CA, USA.
| | - Pauliina M Munne
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, & Medicum, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Castillo-Díaz LA, Gough JE, Miller AF, Saiani A. RGD-functionalised self-assembling peptide hydrogel induces a proliferative profile in human osteoblasts in vitro. J Pept Sci 2025; 31:e3653. [PMID: 39329311 DOI: 10.1002/psc.3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024]
Abstract
Self-assembling peptide hydrogels (SAPHs) have been used in the past decade as reliable three-dimensional (3D) synthetic scaffolds for the culture of a variety of mammalian cells in vitro. Thanks to their versatile physicochemical properties, they allow researchers to tailor the hydrogel properties, including stiffness and functionality to the targeted cells and cells' behaviour. One of the advantages of using SAPH scaffolds is the ease of functionalisation. In the present work, we discuss the effect that functionalising the FEFEFKFK (F, phenylalanine; K, lysine; and E, glutamic acid) hydrogel scaffold using the cell-binding RGDS (fibronectin - R, arginine; G, glycine; D, aspartic acid; S, serine) epitope affects the material properties as well as the function of encapsulated human osteoblast cells. RGDS functionalisation resulted in cells adopting an elongated morphology, suggesting attachment and increased proliferation. While this led to higher cell viability, it also resulted in a decrease in extra-cellular matrix (ECM) protein production as well as a decrease in calcium ion deposition, suggesting lower mineralisation capabilities. The work clearly shows that SAPHs are a flexible platform that allow the modification of scaffolds in a controlled manner to investigate cell-material interactions.
Collapse
Affiliation(s)
- Luis A Castillo-Díaz
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdisciplinaria de Ciencias Biológicas y de la Salud, Universidad de Sonora, Hermosillo, Sonora, Mexico
| | - Julie E Gough
- School of Materials & Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Aline F Miller
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2025; 14:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
4
|
Xin Y, Ligorio C, O'brien M, Collins R, Dong S, Miller AF, Saiani A, Gough JE. Effect of supramolecular peptide hydrogel scaffold charge on HepG2 viability and spheroid formation. J Mater Chem B 2024; 12:12553-12566. [PMID: 39502032 DOI: 10.1039/d4tb01701c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Supramolecular bioinspired self-assembling peptide hydrogel (SAPH) scaffolds represent a class of fully defined synthetic materials whose chemical and mechanical properties can be finely engineered. In this study, the relationship between SAPHs physicochemical properties and HepG2 cells viability, spheroid formation and function are discussed. We first report that negatively charged SAPHs promote hepatocyte proliferation and spheroids formation in vitro 3D culture while positively charged SAPHs lead to hepatocyte death irrespective of the hydrogel mechanical properties. More specifically HepG2 cultured in 3D in E(FKFE)2 negatively charged SAPH maintained a differentiated phenotype and assembled into well-defined spheroids with strong cell-cell interactions. Furthermore, HepG2 spheroids responded to acetaminophen exposure with upregulation of key CYP450 enzymes expression clearly showing their potential for drug toxicity testing. These findings demonstrate how fine-tuned functional SAPH scaffolds can be used to identify key scaffolds parameters affecting cells. In this case we demonstrated the potential of negatively charged SAPHs for the 3D culture of HepG2 with potential applications in drug screening.
Collapse
Affiliation(s)
- Yu Xin
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Cosimo Ligorio
- Department of Materials & Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Marie O'brien
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Richard Collins
- Electron Microscopy Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Siyuan Dong
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Aline F Miller
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Julie E Gough
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| |
Collapse
|
5
|
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
|
6
|
Wychowaniec JK, Šrejber M, Zeng N, Smith AM, Miller AF, Otyepka M, Saiani A. Effects of proline substitution/inclusion on the nanostructure of a self-assembling β-sheet-forming peptide. RSC Adv 2024; 14:37419-37430. [PMID: 39606779 PMCID: PMC11601148 DOI: 10.1039/d4ra07065h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Self-assembling peptides remain persistently interesting objects for building nanostructures and further assemble into macroscopic structures, e.g. hydrogels, at sufficiently high concentrations. The modulation of self-assembling β-sheet-forming peptide sequences, with a selection from the full library of amino acids, offers unique possibility for rational tuning of the resulting nanostructured morphology and topology of the formed hydrogel networks. In the present work, we explored how a known β-sheet-disassembling amino acid, proline (P), affects the self-assembly and gelation properties of amphipathic peptides. For this purpose, we modified the backbone of a known β-sheet-forming peptide, FEFKFEFK (F8, F = phenylalanine, E = glutamic acid, and K = lysine), with P to form three sequences: FEFKPEFK (FP), FEFKPEFKF (KPE) and FEFEPKFKF (EPK). The replacement of F by P in the hydrophobic face resulted in the loss of the extended β-sheet conformation of the FP peptide and no gelation at concentration as high as 100 mg mL-1, compared to typical 5 mg mL-1 concentration corresponding to F8. However, by retaining four hydrophobic phenylalanine amino acids in the sequences, hydrogels containing a partial β-sheet structure were still formed at 30 mg mL-1 for KPE (pH 4-10) and EPK (pH 2-5). TEM, AFM, small-angle X-ray scattering (SAXS) and wide-angle X-ray scattering (WAXS) revealed that KPE and EPK peptides self-assemble into nanoribbons and twisted nanofibers, respectively. Molecular dynamics confirmed that the single amino acid replacement of F by P prevented the assembly of the FP peptide with respect to the stable β-sheet-forming F8 variant. Moreover, additional prolongation by F in the KPE variant and shuffling of the polar amino acid sequence in the EPK peptide supported aggregation capabilities of both variants in forming distinct shapes of individual aggregates. Although the overall number of amino acids is the same in both KPE and EPK, their shifted charge density (i.e., the chemical environment in which ionic groups reside) drives self-assembly into distinct nanostructures. The investigated structural changes can contribute to new material designs for biomedical applications and provide better understanding in the area of protein folding.
Collapse
Affiliation(s)
- Jacek K Wychowaniec
- Department of Materials, Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester UK
- AO Research Institute Davos Clavadelerstrasse 8 Davos 7270 Switzerland
| | - Martin Šrejber
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc 779 00 Olomouc Czech Republic
| | - Niting Zeng
- Department of Materials, Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester UK
| | - Andrew M Smith
- Department of Materials, Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester UK
| | - Aline F Miller
- Department of Chemical Engineering, Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester UK
| | - Michal Otyepka
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc 779 00 Olomouc Czech Republic
- IT4Innovations, VSB-Technical University of Ostrava 708 00 Ostrava-Poruba Czech Republic
| | - Alberto Saiani
- Division of Pharmacy and Optometry, Manchester Institute of Biotechnology, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester UK
| |
Collapse
|
7
|
D’Andrea LD, Romanelli A. Morphology and Applications of Self-Assembled Peptide Nucleic Acids. Int J Mol Sci 2024; 25:12435. [PMID: 39596501 PMCID: PMC11594392 DOI: 10.3390/ijms252212435] [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: 10/03/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Obtaining new materials by exploiting the self-assembly of biomolecules is a very challenging field. In recent years, short peptides and nucleic acids have been used as scaffolds to produce supramolecular structures for different applications in the biomedical and technological fields. In this review, we will focus on the self-assembly of peptide nucleic acids (PNAs), their conjugates with peptides, or other molecules. We will describe the physical properties of the assembled systems and, where described, the application they were designed for.
Collapse
Affiliation(s)
- Luca Domenico D’Andrea
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, via M. Bianco 9, 20131 Milano, Italy;
| | - Alessandra Romanelli
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Venezian 21, 20133 Milan, Italy
| |
Collapse
|
8
|
Peleg-Evron O, Wirzeberger D, Davidovich-Pinhas M, Cometa S, De Giglio E, Bianco-Peled H. Comparative analysis of classic network vs. nanogel junction network in konjac glucomannan/kappa carrageenan hybrid hydrogels. Int J Biol Macromol 2024; 279:135244. [PMID: 39270886 DOI: 10.1016/j.ijbiomac.2024.135244] [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: 06/04/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
The three-dimensional network architecture of hydrogels significantly influences their mechanical and physical properties; therefore, understanding them is essential for designing optimized hydrogel-based biomaterials. This study presents a comparative analysis of two hybrid hydrogels composed of konjac glucomannan (KGM) and kappa carrageenan (KCAR) with the same stiffness (5.2-5.7 kPa and 1.6-1.7 kPa) thus similar cross-linking density but different network architectures: a classic network formed by extended polysaccharide interactions and a nanogel junction network where nanoscale cross-linked KCAR (KCAR-NGs) links KGM chains. The mechanical behavior, dissolution, and diffusion characteristics were examined, revealing that the classic network demonstrates superior tensile resistance, elongation, and solvent-induced swelling resistance, leading to slower dissolution rates and higher viscosity. Conversely, the nanogel junction network offers higher permeability for small molecules and faster dissolution, suggesting a more open network structure. These findings highlight the nanogel-based hydrogels' advantages for biomedical applications requiring stability, permeability, and rapid dissolution without high temperatures or chelating agents. This study underscores the potential of nanogel junction networks to balance hydrogel stiffness and permeability, advancing the design of hydrogel-based biomaterials.
Collapse
Affiliation(s)
- Or Peleg-Evron
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| | - Dana Wirzeberger
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| | - Maya Davidovich-Pinhas
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| | | | - Elvira De Giglio
- Department of Chemistry, University of Bari Aldo Moro, Via E. Orabona 4, Bari 70126, Italy.
| | - Havazelet Bianco-Peled
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
9
|
Guiotto M, Clayton A, Morgan R, Raffoul W, Hart A, Riehle M, di Summa P. Biogelx-IKVAV Is An Innovative Human Platelet Lysate-Adipose-Derived Stem Cells Delivery Strategy to Improve Peripheral Nerve Repair. Tissue Eng Part A 2024; 30:681-692. [PMID: 38482791 DOI: 10.1089/ten.tea.2023.0307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2024] Open
Abstract
Adipose-derived stem cells (ADSC) are nowadays one of the most exploited cells in regenerative medicine. They are fast growing, capable of enhancing axonal elongation, support and locally stimulate Schwann cells (SCs), and protect de-innervated muscles from atrophy after a peripheral nerve injury. With the aim of developing a bio-safe, clinically translatable cell-therapy, we assessed the effect of ADSC pre-expanded with human platelet lysate in an in vivo rat model, delivering the cells into a 15 mm critical-size sciatic nerve defect embedded within a laminin-peptide-functionalized hydrogel (Biogelx-IKVAV) wrapped by a poly-ɛ-caprolactone (PCL) nerve conduit. ADSC retained their stemness, their immunophenotype and proliferative activity when tested in vitro. At 6 weeks post-implantation, robust regeneration was observed across the critical-size gap as evaluated by both the axonal elongation (anti-NF 200) and SC proliferation (anti-S100) within the human ADSC-IKVAV filled PCL conduit. All the other experimental groups manifested significantly lower levels of growth cone elongation. The histological gastrocnemius muscle analysis was comparable with no quantitative significant differences among the experimental groups. Taken together, these results suggest that ADSC encapsulated in Biogelx-IKVAV are a potential path to improve the efficacy of nerve regeneration. New perspectives can be pursued for the development of a fully synthetic bioengineered nerve graft for the treatment of peripheral nerve injury. Impact statement Human adipose-derived stem cells pre-expanded in vitro with human platelet lysate culture medium additive and encapsulated into BiogelX-IKVAV are a promising strategy to improve nerve regeneration through a critical nerve gap in rat model.
Collapse
Affiliation(s)
- Martino Guiotto
- Department of Plastic, Reconstructive and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, United Kingdom
| | | | | | - Wassim Raffoul
- Department of Plastic, Reconstructive and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Andrew Hart
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, United Kingdom
- Canniesburn Plastic Surgery Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Mathis Riehle
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, United Kingdom
| | - Pietro di Summa
- Department of Plastic, Reconstructive and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
10
|
De Maeseneer T, Cauwenbergh T, Gardiner J, White JF, Thielemans W, Martin C, Moldenaers P, Ballet S, Cardinaels R. Peptide Sequence Variations Govern Hydrogel Stiffness: Insights from a Multi-Scale Structural Analysis of H-FQFQFK-NH 2 Peptide Derivatives. Macromol Biosci 2024; 24:e2300579. [PMID: 38552257 DOI: 10.1002/mabi.202300579] [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: 12/18/2023] [Revised: 03/14/2024] [Indexed: 04/26/2024]
Abstract
Throughout the past decades, amphipathic peptide-based hydrogels have proven to be promising materials for biomedical applications. Amphipathic peptides are known to adopt β-sheet configurations that self-assemble into fibers that then interact to form a hydrogel network. A fundamental understanding of how the peptide sequence alters the structural properties of the hydrogels would allow for a more rational design of novel peptides for a variety of biomedical applications in the future. Therefore, the current work investigates how changing the type of amino acid, the amphipathic pattern, and the peptide length affects the secondary structure, fiber characteristics, and stiffness of peptide-based hydrogels. Hereto, seven amphipathic peptides of different sequence and length, four of which have not been previously reported, based on and including the hexapeptide H-Phe-Gln-Phe-Gln-Phe-Lys-NH2, are synthesized and thoroughly characterized by circular dichroism (CD), Fourier Transform Infrared (FTIR) spectroscopy, Wide Angle X-ray Scattering (WAXS), Small Angle X-ray Scattering (SAXS), Transmission Electron Microscopy (TEM), and Thioflavin T (ThT) fibrillization assays. The results show that a high amount of regularly spaced β-sheets, a high amount of fibers, and fiber bundling contribute to the stiffness of the hydrogel. Furthermore, a study of the time-dependent fibril formation process reveals complex transient dynamics. The peptide strands structure through an intermediate helical state prior to β-sheet formation, which is found to be concentration- and time-dependent.
Collapse
Affiliation(s)
- Tess De Maeseneer
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J, Box 2424, Leuven, 3001, Belgium
| | - Thibault Cauwenbergh
- Research Group of Organic Chemistry, Department of Chemistry, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, 1050, Belgium
| | - James Gardiner
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3169, Australia
| | - Jacinta F White
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3169, Australia
| | - Wim Thielemans
- Sustainable Materials Lab, Department of Chemical Engineering, KU Leuven, campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk, 8500, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Department of Chemistry, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, 1050, Belgium
| | - Paula Moldenaers
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J, Box 2424, Leuven, 3001, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Department of Chemistry, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, 1050, Belgium
| | - Ruth Cardinaels
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J, Box 2424, Leuven, 3001, Belgium
- Processing and Performance of Materials, Department of Mechanical Engineering, TU Eindhoven, P.O. Box 513, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
11
|
Veloso SRS, Rosa M, Diaferia C, Fernandes C. A Review on the Rheological Properties of Single Amino Acids and Short Dipeptide Gels. Gels 2024; 10:507. [PMID: 39195036 DOI: 10.3390/gels10080507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Self-assembled peptide-based hydrogels have attracted considerable interest from the research community. Particularly, low molecular weight gelators (LMWGs) consisting of amino acids and short peptides are highly suitable for biological applications owing to their facile synthesis and scalability, as well as their biocompatibility, biodegradability, and stability in physiological conditions. However, challenges in understanding the structure-property relationship and lack of design rules hinder the development of new gelators with the required properties for several applications. Hereby, in the plethora of peptide-based gelators, this review discusses the mechanical properties of single amino acid and dipeptide-based hydrogels. A mutual analysis of these systems allows us to highlight the relationship between the gel mechanical properties and amino acid sequence, preparation methods, or N capping groups. Additionally, recent advancements in the tuning of the gels' rheological properties are reviewed. In this way, the present review aims to help bridge the knowledge gap between structure and mechanical properties, easing the selection or design of peptides with the required properties for biological applications.
Collapse
Affiliation(s)
- Sérgio R S Veloso
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Laboratory of Physics for Materials and Emergent Technologies (LaPMET), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Mariangela Rosa
- Department of Pharmacy, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi "Carlo Pedone" (CIRPeB), University of Naples "Federico II", Via Tommaso de Amicis 95, 80131 Naples, Italy
| | - Carlo Diaferia
- Department of Pharmacy, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi "Carlo Pedone" (CIRPeB), University of Naples "Federico II", Via Tommaso de Amicis 95, 80131 Naples, Italy
| | - Célio Fernandes
- Transport Phenomena Research Centre (CEFT), Department of Mechanical Engineering, Faculty of Engineering, University of Porto (FEUP), Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Centre of Mathematics (CMAT), School of Sciences, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
12
|
Cimmino L, Diaferia C, Rosa M, Morelli G, Rosa E, Accardo A. Hybrid peptide-PNA monomers as building blocks for the fabrication of supramolecular aggregates. J Pept Sci 2024; 30:e3573. [PMID: 38471735 DOI: 10.1002/psc.3573] [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: 01/17/2024] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/14/2024]
Abstract
Advantages like biocompatibility, biodegradability and tunability allowed the exploitation of peptides and peptidomimetics as versatile therapeutic or diagnostic agents. Because of their selectivity towards transmembrane receptors or cell membranes, peptides have also been identified as suitable molecules able to deliver in vivo macromolecules, proteins or nucleic acids. However, after the identification of the homodimer diphenylalanine (FF) as an aggregative motif inside the Aβ1-42 polypeptide, short and ultrashort peptides have been studied as building blocks for the fabrication of supramolecular, ordered nanostructures for applications in biotechnological, biomedical and industrial fields. In this perspective, many hybrid molecules that combine FF with other chemical entities have been synthesized and characterized. Two novel hybrid derivatives (tFaF and cFgF), in which the FF homodimer is alternated with the peptide-nucleic acid (PNA) heterodimer "g-c" (guanine-cytosine) or "a-t" (adenine-thymine) and their dimeric forms (tFaF)2 and (cFgF)2 were synthesized. The structural characterization performed by circular dichroism (CD), Fourier transform infrared (FTIR) and fluorescence spectroscopies highlighted the capability of all the FF-PNA derivatives to self-assemble into β-sheet structures. As a consequence of this supramolecular organization, the resulting aggregates also exhibit optoelectronic properties already reported for other similar nanostructures. This photoemissive behavior is promising for their potential applications in bioimaging.
Collapse
Affiliation(s)
| | - Carlo Diaferia
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Mariangela Rosa
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Giancarlo Morelli
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Elisabetta Rosa
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Antonella Accardo
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| |
Collapse
|
13
|
Krupa I, Treacy NJ, Clerkin S, Davis JL, Miller AF, Saiani A, Wychowaniec JK, Reynaud EG, Brougham DF, Crean J. Protocol for the Growth and Maturation of hiPSC-Derived Kidney Organoids using Mechanically Defined Hydrogels. Curr Protoc 2024; 4:e1096. [PMID: 38984433 DOI: 10.1002/cpz1.1096] [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: 07/11/2024]
Abstract
With recent advances in the reprogramming of somatic cells into induced Pluripotent Stem Cells (iPSCs), gene editing technologies, and protocols for the directed differentiation of stem cells into heterogeneous tissues, iPSC-derived kidney organoids have emerged as a useful means to study processes of renal development and disease. Considerable advances guided by knowledge of fundamental renal developmental signaling pathways have been made with the use of exogenous morphogens to generate more robust kidney-like tissues in vitro. However, both biochemical and biophysical microenvironmental cues are major influences on tissue development and self-organization. In the context of engineering the biophysical aspects of the microenvironment, the use of hydrogel extracellular scaffolds for organoid studies has been gaining interest. Two families of hydrogels have recently been the subject of significant attention: self-assembling peptide hydrogels (SAPHs), which are fully synthetic and chemically defined, and gelatin methacryloyl (GelMA) hydrogels, which are semi-synthetic. Both can be used as support matrices for growing kidney organoids. Based on our recently published work, we highlight methods describing the generation of human iPSC (hiPSC)-derived kidney organoids and their maturation within SAPHs and GelMA hydrogels. We also detail protocols required for the characterization of such organoids using immunofluorescence imaging. Together, these protocols should enable the user to grow hiPSC-derived kidney organoids within hydrogels of this kind and evaluate the effects that the biophysical microenvironment provided by the hydrogels has on kidney organoid maturation. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Directed differentiation of human induced pluripotent stem cells (hiPSCs) into kidney organoids and maturation within mechanically tunable self-assembling peptide hydrogels (SAPHs) Alternate Protocol: Encapsulation of day 9 nephron progenitor aggregates in gelatin methacryloyl (GelMA) hydrogels. Support Protocol 1: Human induced pluripotent stem cell (hiPSC) culture. Support Protocol 2: Organoid fixation with paraformaldehyde (PFA) Basic Protocol 2: Whole-mount immunofluorescence imaging of kidney organoids. Basic Protocol 3: Immunofluorescence of organoid cryosections.
Collapse
Affiliation(s)
- Ivan Krupa
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Niall J Treacy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Shane Clerkin
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Jessica L Davis
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Aline F Miller
- Department of Chemical Engineering & Manchester Institute of Biotechnology (MIB), School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, United Kingdom
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology (MIB), School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom
| | - Jacek K Wychowaniec
- UCD School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
- Current address: AO Research Institute Davos, Davos, Switzerland
| | - Emmanuel G Reynaud
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| | - Dermot F Brougham
- UCD School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
| | - John Crean
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
14
|
Dong S, Chapman SL, Pluen A, Richardson SM, Miller AF, Saiani A. Effect of Peptide-Polymer Host-Guest Electrostatic Interactions on Self-Assembling Peptide Hydrogels Structural and Mechanical Properties and Polymer Diffusivity. Biomacromolecules 2024; 25:3628-3641. [PMID: 38771115 PMCID: PMC11170954 DOI: 10.1021/acs.biomac.4c00232] [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: 02/19/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024]
Abstract
Peptide-based supramolecular hydrogels are an attractive class of soft materials for biomedical applications when biocompatibility is a key requirement as they exploit the physical self-assembly of short self-assembling peptides avoiding the need for chemical cross-linking. Based on the knowledge developed through our previous work, we designed two novel peptides, E(FKFE)2 and K(FEFK)2, that form transparent hydrogels at pH 7. We characterized the phase behavior of these peptides and showed the clear link that exists between the charge carried by the peptides and the physical state of the samples. We subsequently demonstrate the cytocompatibility of the hydrogel and its suitability for 3D cell culture using 3T3 fibroblasts and human mesenchymal stem cells. We then loaded the hydrogels with two polymers, poly-l-lysine and dextran. When polymer and peptide fibers carry opposite charges, the size of the elemental fibril formed decreases, while the overall level of fiber aggregation and fiber bundle formation increases. This overall network topology change, and increase in cross-link stability and density, leads to an overall increase in the hydrogel mechanical properties and stability, i.e., resistance to swelling when placed in excess media. Finally, we investigate the diffusion of the polymers out of the hydrogels and show how electrostatic interactions can be used to control the release of large molecules. The work clearly shows how polymers can be used to tailor the properties of peptide hydrogels through guided intermolecular interactions and demonstrates the potential of these new soft hydrogels for use in the biomedical field in particular for delivery or large molecular payloads and cells as well as scaffolds for 3D cell culture.
Collapse
Affiliation(s)
- Siyuan Dong
- Department
of Chemical Engineering, School of Engineering, Faculty of Science
and Engineering, The University of Manchester, Oxford Road, M13 9PL Manchester, U.K.
- Manchester
Institute of Biotechnology (MIB), Faculty of Science and Engineering, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Sam L. Chapman
- Division
of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology,
Medicine and Health, The University of Manchester, Oxford Road, M13 9PL Manchester, U.K.
| | - Alain Pluen
- Division
of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology,
Medicine and Health, The University of Manchester, Oxford Road, M13 9PL Manchester, U.K.
| | - Stephen M. Richardson
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, The University of
Manchester, Manchester M13 9PT, U.K.
| | - Aline F. Miller
- Department
of Chemical Engineering, School of Engineering, Faculty of Science
and Engineering, The University of Manchester, Oxford Road, M13 9PL Manchester, U.K.
- Manchester
Institute of Biotechnology (MIB), Faculty of Science and Engineering, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Alberto Saiani
- Manchester
Institute of Biotechnology (MIB), Faculty of Science and Engineering, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
- Division
of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology,
Medicine and Health, The University of Manchester, Oxford Road, M13 9PL Manchester, U.K.
| |
Collapse
|
15
|
Lingard E, Dong S, Hoyle A, Appleton E, Hales A, Skaria E, Lawless C, Taylor-Hearn I, Saadati S, Chu Q, Miller AF, Domingos M, Saiani A, Swift J, Gilmore AP. Optimising a self-assembling peptide hydrogel as a Matrigel alternative for 3-dimensional mammary epithelial cell culture. BIOMATERIALS ADVANCES 2024; 160:213847. [PMID: 38657288 DOI: 10.1016/j.bioadv.2024.213847] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/10/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
Three-dimensional (3D) organoid models have been instrumental in understanding molecular mechanisms responsible for many cellular processes and diseases. However, established organic biomaterial scaffolds used for 3D hydrogel cultures, such as Matrigel, are biochemically complex and display significant batch variability, limiting reproducibility in experiments. Recently, there has been significant progress in the development of synthetic hydrogels for in vitro cell culture that are reproducible, mechanically tuneable, and biocompatible. Self-assembling peptide hydrogels (SAPHs) are synthetic biomaterials that can be engineered to be compatible with 3D cell culture. Here we investigate the ability of PeptiGel® SAPHs to model the mammary epithelial cell (MEC) microenvironment in vitro. The positively charged PeptiGel®Alpha4 supported MEC viability, but did not promote formation of polarised acini. Modifying the stiffness of PeptiGel® Alpha4 stimulated changes in MEC viability and changes in protein expression associated with altered MEC function, but did not fully recapitulate the morphologies of MECs grown in Matrigel. To supply the appropriate biochemical signals for MEC organoids, we supplemented PeptiGels® with laminin. Laminin was found to require negatively charged PeptiGel® Alpha7 for functionality, but was then able to provide appropriate signals for correct MEC polarisation and expression of characteristic proteins. Thus, optimisation of SAPH composition and mechanics allows tuning to support tissue-specific organoids.
Collapse
Affiliation(s)
- Eliana Lingard
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Siyuan Dong
- School of Materials, Faculty of Science and Engineering, The University of Manchester, Oxford Road, Manchester, UK
| | - Anna Hoyle
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Ellen Appleton
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Alis Hales
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Eldhose Skaria
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK
| | - Craig Lawless
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK
| | - Isobel Taylor-Hearn
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Simon Saadati
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK
| | - Qixun Chu
- School of Materials, Faculty of Science and Engineering, The University of Manchester, Oxford Road, Manchester, UK; Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Aline F Miller
- School of Materials, Faculty of Science and Engineering, The University of Manchester, Oxford Road, Manchester, UK
| | - Marco Domingos
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, United Kingdom, M13 9PL, UK
| | - Alberto Saiani
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK; Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Andrew P Gilmore
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester M13 9PT, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK.
| |
Collapse
|
16
|
Singh K, Wychowaniec JK, Edwards-Gayle CJC, Reynaud EG, Rodriguez BJ, Brougham DF. Structure-dynamics correlations in composite PF127-PEG-based hydrogels; cohesive/hydrophobic interactions determine phase and rheology and identify the role of micelle concentration in controlling 3D extrusion printability. J Colloid Interface Sci 2024; 660:302-313. [PMID: 38244497 DOI: 10.1016/j.jcis.2023.12.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024]
Abstract
A library of composite polymer networks (CPNs) were formed by combining Pluronic F127, as the primary gelator, with a range of di-acrylate functionalised PEG polymers, which tune the rheological properties and provide UV crosslinkability. A coarse-grained sol-gel room temperature phase diagram was constructed for the CPN library, which identifies PEG-dependent disruption of micelles as leading to liquefication. Small angle X-ray scattering and rheological measurements provide detailed insight into; (i) micelle-micelle ordering; (ii) micelle-micelle disruption, and; (iii) acrylate-micelle disruption; with contributions that depend on composition, including weak PEG chain length and end group effects. The influence of composition on 3D extrusion printability through modulation of the cohesive/hydrophobic interactions was assessed. It was found that only micelle content provides consistent changes in printing fidelity, controlled largely by printing conditions (pressure and feed rate). Finally, the hydrogels were shown to be UV photo-crosslinkable, which further improves fidelity and structural integrity, and usefully reduces the mesh size. Our results provide a guide for design of 3D-printable CPN inks for future biomedical applications.
Collapse
Affiliation(s)
- Krutika Singh
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland; AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland.
| | | | - Emmanuel G Reynaud
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Brian J Rodriguez
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; School of Physics, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
17
|
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
|
18
|
Dodd-O J, Roy A, Siddiqui Z, Jafari R, Coppola F, Ramasamy S, Kolloli A, Kumar D, Kaundal S, Zhao B, Kumar R, Robang AS, Li J, Azizogli AR, Pai V, Acevedo-Jake A, Heffernan C, Lucas A, McShan AC, Paravastu AK, Prasad BVV, Subbian S, Král P, Kumar V. Antiviral fibrils of self-assembled peptides with tunable compositions. Nat Commun 2024; 15:1142. [PMID: 38326301 PMCID: PMC10850501 DOI: 10.1038/s41467-024-45193-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
The lasting threat of viral pandemics necessitates the development of tailorable first-response antivirals with specific but adaptive architectures for treatment of novel viral infections. Here, such an antiviral platform has been developed based on a mixture of hetero-peptides self-assembled into functionalized β-sheets capable of specific multivalent binding to viral protein complexes. One domain of each hetero-peptide is designed to specifically bind to certain viral proteins, while another domain self-assembles into fibrils with epitope binding characteristics determined by the types of peptides and their molar fractions. The self-assembled fibrils maintain enhanced binding to viral protein complexes and retain high resilience to viral mutations. This method is experimentally and computationally tested using short peptides that specifically bind to Spike proteins of SARS-CoV-2. This platform is efficacious, inexpensive, and stable with excellent tolerability.
Collapse
Affiliation(s)
- Joseph Dodd-O
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Roya Jafari
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Francesco Coppola
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Santhamani Ramasamy
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Dilip Kumar
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Soni Kaundal
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Boyang Zhao
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jeffrey Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Varun Pai
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
- SAPHTx Inc, Newark, NJ, 07104, USA
| | - Alexandra Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E, Tempe, AZ, USA
| | - Andrew C McShan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - B V Venkataram Prasad
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Physics, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- SAPHTx Inc, Newark, NJ, 07104, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103, USA.
| |
Collapse
|
19
|
Laurent H, Hughes MDG, Walko M, Brockwell DJ, Mahmoudi N, Youngs TGA, Headen TF, Dougan L. Visualization of Self-Assembly and Hydration of a β-Hairpin through Integrated Small and Wide-Angle Neutron Scattering. Biomacromolecules 2023; 24:4869-4879. [PMID: 37874935 PMCID: PMC10646990 DOI: 10.1021/acs.biomac.3c00583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/03/2023] [Indexed: 10/26/2023]
Abstract
Fundamental understanding of the structure and assembly of nanoscale building blocks is crucial for the development of novel biomaterials with defined architectures and function. However, accessing self-consistent structural information across multiple length scales is challenging. This limits opportunities to exploit atomic scale interactions to achieve emergent macroscale properties. In this work we present an integrative small- and wide-angle neutron scattering approach coupled with computational modeling to reveal the multiscale structure of hierarchically self-assembled β hairpins in aqueous solution across 4 orders of magnitude in length scale from 0.1 Å to 300 nm. Our results demonstrate the power of this self-consistent cross-length scale approach and allows us to model both the large-scale self-assembly and small-scale hairpin hydration of the model β hairpin CLN025. Using this combination of techniques, we map the hydrophobic/hydrophilic character of this model self-assembled biomolecular surface with atomic resolution. These results have important implications for the multiscale investigation of aqueous peptides and proteins, for the prediction of ligand binding and molecular associations for drug design, and for understanding the self-assembly of peptides and proteins for functional biomaterials.
Collapse
Affiliation(s)
- Harrison Laurent
- School
of Physics and Astronomy, University of
Leeds, Leeds, United Kingdom, LS2
9JT
| | - Matt D. G. Hughes
- School
of Physics and Astronomy, University of
Leeds, Leeds, United Kingdom, LS2
9JT
- Astbury
Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom LS2
9JT
| | - Martin Walko
- School
of Chemistry, University of Leeds, Leeds, United
Kingdom, LS2 9JT
| | - David J. Brockwell
- Astbury
Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom LS2
9JT
| | - Najet Mahmoudi
- ISIS
Neutron and Muon Source, Rutherford Appleton
Laboratory, Harwell Oxford, Didcot, United Kingdom, OX11 0QX
| | - Tristan G. A. Youngs
- ISIS
Neutron and Muon Source, Rutherford Appleton
Laboratory, Harwell Oxford, Didcot, United Kingdom, OX11 0QX
| | - Thomas F. Headen
- ISIS
Neutron and Muon Source, Rutherford Appleton
Laboratory, Harwell Oxford, Didcot, United Kingdom, OX11 0QX
| | - Lorna Dougan
- School
of Physics and Astronomy, University of
Leeds, Leeds, United Kingdom, LS2
9JT
- Astbury
Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom LS2
9JT
| |
Collapse
|
20
|
Hansda B, Mondal B, Hazra S, Das KS, Castelletto V, Hamley IW, Banerjee A. Effect of molar ratio and concentration on the rheological properties of two-component supramolecular hydrogels: tuning of the morphological and drug releasing behaviour. SOFT MATTER 2023; 19:8264-8273. [PMID: 37869972 DOI: 10.1039/d3sm00883e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Self-assembled supramolecular hydrogels offer great potential as biomaterials and drug delivery systems. Specifically, peptide-based multicomponent hydrogels are promising materials due to their advantage that their mechanical and physical properties can be tuned to enhance their functionalities and broaden their applications. Herein, we report two-component assembly and formation of hydrogels containing inexpensive complementary anionic, BUVV-OH (A), and cationic, KFFC12 (B), peptide amphiphiles. Individually, neither of these components formed a hydrogel, while mixtures with compositions 1 : 1, 1 : 2, and 2 : 1 (molar ratio) as A : B show hydrogel formation (Milli-Q water, at pH = 6.79). These hydrogels displayed a good shear-thinning behaviour with different mechanical stabilities and nano-fibrous network structures. The 1 : 1 hydrogel shows good cell viability for human embryonic kidney (HEK-293) cells and CHO cells indicating its non-cytotoxicity. The biocompatible, thixotropic 1 : 1 hydrogel with a nanofiber network structure shows the highest mechanical strength with a storage modulus of 3.4 × 103 Pa. The hydrogel is able to encapsulate drugs including antibiotics amoxicillin and rifampicin, and anticancer drug doxorubicin, and it exhibits sustainable release of 76%, 70%, and 81% respectively in vitro after 3 days. The other two mixtures (composition 1 : 2 and 2 : 1) are unable to form a hydrogel when they are loaded with these drugs. Interestingly, it is noticed that with an increase in concentration, the mechanical strength of a 1 : 1 hydrogel is significantly enhanced, showing potential that may act as a scaffold for tissue engineering. The two-component gel offers tunable mechanical properties, thixotropy, injectability, and biocompatibility and has great potential as a scaffold for sustained drug release and tissue engineering.
Collapse
Affiliation(s)
- Biswanath Hansda
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Biplab Mondal
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Soumyajit Hazra
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Krishna Sundar Das
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India
| | | | - Ian W Hamley
- Department of Chemistry, University of Reading, Reading RG6 6AD, UK
| | - Arindam Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| |
Collapse
|
21
|
Altuntaş E, Özkan B, Güngör S, Özsoy Y. Biopolymer-Based Nanogel Approach in Drug Delivery: Basic Concept and Current Developments. Pharmaceutics 2023; 15:1644. [PMID: 37376092 DOI: 10.3390/pharmaceutics15061644] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Due to their increased surface area, extent of swelling and active substance-loading capacity and flexibility, nanogels made from natural and synthetic polymers have gained significant interest in scientific and industrial areas. In particular, the customized design and implementation of nontoxic, biocompatible, and biodegradable micro/nano carriers makes their usage very feasible for a range of biomedical applications, including drug delivery, tissue engineering, and bioimaging. The design and application methodologies of nanogels are outlined in this review. Additionally, the most recent advancements in nanogel biomedical applications are discussed, with particular emphasis on applications for the delivery of drugs and biomolecules.
Collapse
Affiliation(s)
- Ebru Altuntaş
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Burcu Özkan
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220 Istanbul, Türkiye
| | - Sevgi Güngör
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Yıldız Özsoy
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| |
Collapse
|
22
|
Treherne JM, Miller AF. Novel hydrogels: are they poised to transform 3D cell-based assay systems in early drug discovery? Expert Opin Drug Discov 2023; 18:335-346. [PMID: 36722285 DOI: 10.1080/17460441.2023.2175813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Success in drug discovery remains unpredictable. However, more predictive and relevant disease models are becoming pivotal to demonstrating the clinical benefits of new drugs earlier in the lengthy drug discovery process. Novel hydrogel scaffolds are being developed to transform the relevance of such 3D cell-based in vitro assay systems. AREAS COVERED Most traditional hydrogels are still of unknown composition and suffer significant batch-to-batch variations, which lead to technical constraints. This article looks at how a new generation of novel synthetic hydrogels that are based on self-assembling peptides are poised to transform 3D cell-based assay systems by improving their relevance, reproducibility and scalability. EXPERT OPINION The emerging advantages of using these novel hydrogels for human 3D screening assays should enable the discovery of more cost-effective drugs, leading to improved patient benefits. Such a disruptive change could also reduce the considerable time lag from obtaining in vitro assay data to initiating clinical trials. There is now a sufficient body of data available in the literature to enable this ambition to become a reality by significantly improving the predictive validity of 3D cell-based assays in early drug discovery. Novel hydrogels are key to unlocking the full potential of these assay systems.
Collapse
Affiliation(s)
- J Mark Treherne
- Talisman Therapeutics Ltd, Jonas Webb Building and Cell Guidance Sysyems Ltd, Babraham Research Campus, Cambridge, UK
| | - Aline F Miller
- Manchester Institute of Biotechnology, School of Engineering, The University of Manchester, Oxford Road, Manchester, UK
| |
Collapse
|
23
|
Treacy NJ, Clerkin S, Davis JL, Kennedy C, Miller AF, Saiani A, Wychowaniec JK, Brougham DF, Crean J. Growth and differentiation of human induced pluripotent stem cell (hiPSC)-derived kidney organoids using fully synthetic peptide hydrogels. Bioact Mater 2023; 21:142-156. [PMID: 36093324 PMCID: PMC9420433 DOI: 10.1016/j.bioactmat.2022.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/27/2022] [Accepted: 08/01/2022] [Indexed: 11/15/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived kidney organoids have prospective applications ranging from basic disease modelling to personalised medicine. However, there remains a necessity to refine the biophysical and biochemical parameters that govern kidney organoid formation. Differentiation within fully-controllable and physiologically relevant 3D growth environments will be critical to improving organoid reproducibility and maturation. Here, we matured hiPSC-derived kidney organoids within fully synthetic self-assembling peptide hydrogels (SAPHs) of variable stiffness (storage modulus, G'). The resulting organoids contained complex structures comparable to those differentiated within the animal-derived matrix, Matrigel. Single-cell RNA sequencing (scRNA-seq) was then used to compare organoids matured within SAPHs to those grown within Matrigel or at the air-liquid interface. A total of 13,179 cells were analysed, revealing 14 distinct clusters. Organoid compositional analysis revealed a larger proportion of nephron cell types within Transwell-derived organoids, while SAPH-derived organoids were enriched for stromal-associated cell populations. Notably, differentiation within a higher G' SAPH generated podocytes with more mature gene expression profiles. Additionally, maturation within a 3D microenvironment significantly reduced the derivation of off-target cell types, which are a known limitation of current kidney organoid protocols. This work demonstrates the utility of synthetic peptide-based hydrogels with a defined stiffness, as a minimally complex microenvironment for the selected differentiation of kidney organoids.
Collapse
Affiliation(s)
- Niall J Treacy
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Shane Clerkin
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Jessica L Davis
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Ciarán Kennedy
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Aline F Miller
- Department of Materials & Manchester Institute of Biotechnology (MIB), School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Alberto Saiani
- Department of Materials & Manchester Institute of Biotechnology (MIB), School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Jacek K Wychowaniec
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - John Crean
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| |
Collapse
|
24
|
Hardan L, Chedid JCA, Bourgi R, Cuevas-Suárez CE, Lukomska-Szymanska M, Tosco V, Monjarás-Ávila AJ, Jabra M, Salloum-Yared F, Kharouf N, Mancino D, Haikel Y. Peptides in Dentistry: A Scoping Review. Bioengineering (Basel) 2023; 10:214. [PMID: 36829708 PMCID: PMC9952573 DOI: 10.3390/bioengineering10020214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Currently, it remains unclear which specific peptides could be appropriate for applications in different fields of dentistry. The aim of this scoping review was to scan the contemporary scientific papers related to the types, uses and applications of peptides in dentistry at the moment. Literature database searches were performed in the following databases: PubMed/MEDLINE, Scopus, Web of Science, Embase, and Scielo. A total of 133 articles involving the use of peptides in dentistry-related applications were included. The studies involved experimental designs in animals, microorganisms, or cells; clinical trials were also identified within this review. Most of the applications of peptides included caries management, implant osseointegration, guided tissue regeneration, vital pulp therapy, antimicrobial activity, enamel remineralization, periodontal therapy, the surface modification of tooth implants, and the modification of other restorative materials such as dental adhesives and denture base resins. The in vitro and in vivo studies included in this review suggested that peptides may have beneficial effects for treating early carious lesions, promoting cell adhesion, enhancing the adhesion strength of dental implants, and in tissue engineering as healthy promotors of the periodontium and antimicrobial agents. The lack of clinical trials should be highlighted, leaving a wide space available for the investigation of peptides in dentistry.
Collapse
Affiliation(s)
- Louis Hardan
- Department of Restorative Dentistry, School of Dentistry, Saint Joseph University, Beirut 1107 2180, Lebanon
| | - Jean Claude Abou Chedid
- Department of Pediatric Dentistry, Faculty of Dentistry, Saint Joseph University, Beirut 1107 2180, Lebanon
| | - Rim Bourgi
- Department of Restorative Dentistry, School of Dentistry, Saint Joseph University, Beirut 1107 2180, Lebanon
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France
| | - Carlos Enrique Cuevas-Suárez
- Dental Materials Laboratory, Academic Area of Dentistry, Autonomous University of Hidalgo State, San Agustín Tlaxiaca 42160, Mexico
| | | | - Vincenzo Tosco
- Department of Clinical Sciences and Stomatology (DISCO), Polytechnic University of Marche, 60126 Ancona, Italy
| | - Ana Josefina Monjarás-Ávila
- Dental Materials Laboratory, Academic Area of Dentistry, Autonomous University of Hidalgo State, San Agustín Tlaxiaca 42160, Mexico
| | - Massa Jabra
- Faculty of Medicine, Damascus University, Damascus 0100, Syria
| | | | - Naji Kharouf
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Davide Mancino
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Youssef Haikel
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
25
|
Wu T, Wu Y, Cao Z, Zhao L, Lv J, Li J, Xu Y, Zhang P, Liu X, Sun Y, Cheng M, Tang K, Jiang X, Ling C, Yao Q, Zhu Y. Cell-free and cytokine-free self-assembling peptide hydrogel-polycaprolactone composite scaffolds for segmental bone defects. Biomater Sci 2023; 11:840-853. [PMID: 36512317 DOI: 10.1039/d2bm01609e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Segmental bone defects over the self-healing threshold are a major challenge for orthopedics. Despite the advancements in clinical practice, traditional tissue engineering methods are limited by the addition of heterogeneous cells and cytokines, leading to carcinoma or other adverse effects. Here, we present a cell-free and cytokine-free strategy using an ECM-mimetic self-assembling peptide hydrogel (SAPH)- polycaprolactone (PCL) composite scaffold. The hydrophilic SAPH endows the rigid PCL scaffold with excellent biocompatibility and preference for osteogenesis induction. The autologous cells around the bone defect site immediately grew, proliferated, and secreted ECM and cytokines after contacting the implanted SAPH-PCL composite scaffold, and the bone repair of rabbit ulnar segmental bone defect was achieved in just six months. Quantitative proteomic analysis reveals that the SAPH-PCL composite scaffold accelerates osteoblastogenesis, osteoclastogenesis, and angiogenesis with moderate immune responses and negligible effects on pathological fibrosis. These findings have important implications for the potential clinical applications of the SAPH-PCL composite scaffold in patients with segmental bone defects and identify the mechanisms of action for accelerated segmental bone defect repair.
Collapse
Affiliation(s)
- Tong Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Lulu Zhao
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Jiayi Li
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Yue Xu
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Xu Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Min Cheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| | - Kexin Tang
- College of Pharmaceutical Sciences, Nanjing Tech University, 211816, Nanjing, China
| | - Xiao Jiang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Chen Ling
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 211816, Nanjing, China.
| |
Collapse
|
26
|
Luan X, Kong H, He P, Yang G, Zhu D, Guo L, Wei G. Self-Assembled Peptide-Based Nanodrugs: Molecular Design, Synthesis, Functionalization, and Targeted Tumor Bioimaging and Biotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205787. [PMID: 36440657 DOI: 10.1002/smll.202205787] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/14/2022] [Indexed: 06/16/2023]
Abstract
Functional nanomaterials as nanodrugs based on the self-assembly of inorganics, polymers, and biomolecules have showed wide applications in biomedicine and tissue engineering. Ascribing to the unique biological, chemical, and physical properties of peptide molecules, peptide is used as an excellent precursor material for the synthesis of functional nanodrugs for highly effective cancer therapy. Herein, recent progress on the design, synthesis, functional regulation, and cancer bioimaging and biotherapy of peptide-based nanodrugs is summarized. For this aim, first molecular design and controllable synthesis of peptide nanodrugs with 0D to 3D structures are presented, and then the functional customization strategies for peptide nanodrugs are presented. Then, the applications of peptide-based nanodrugs in bioimaging, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT) are demonstrated and discussed in detail. Furthermore, peptide-based drugs in preclinical, clinical trials, and approved are briefly described. Finally, the challenges and potential solutions are pointed out on addressing the questions of this promising research topic. This comprehensive review can guide the motif design and functional regulation of peptide nanomaterials for facile synthesis of nanodrugs, and further promote their practical applications for diagnostics and therapy of diseases.
Collapse
Affiliation(s)
- Xin Luan
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Hao Kong
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Peng He
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Guozheng Yang
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Danzhu Zhu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Lei Guo
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| |
Collapse
|
27
|
Flexibility, size and hydrophobicity of alkyl side groups in methoxy-poly (ethylene glycol)-polypeptide for the nano-assembly and thermo-sensitivity. POLYMER 2022. [DOI: 10.1016/j.polymer.2022.125499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
28
|
Slawinski M, Kaeek M, Rajmiel Y, Khoury LR. Acetic Acid Enables Precise Tailoring of the Mechanical Behavior of Protein-Based Hydrogels. NANO LETTERS 2022; 22:6942-6950. [PMID: 36018622 PMCID: PMC9479135 DOI: 10.1021/acs.nanolett.2c01558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Engineering viscoelastic and biocompatible materials with tailored mechanical and microstructure properties capable of mimicking the biological stiffness (<17 kPa) or serving as bioimplants will bring protein-based hydrogels to the forefront in the biomaterials field. Here, we introduce a method that uses different concentrations of acetic acid (AA) to control the covalent tyrosine-tyrosine cross-linking interactions at the nanoscale level during protein-based hydrogel synthesis and manipulates their mechanical and microstructure properties without affecting protein concentration and (un)folding nanomechanics. We demonstrated this approach by adding AA as a precursor to the preparation buffer of a photoactivated protein-based hydrogel mixture. This strategy allowed us to synthesize hydrogels made from bovine serum albumin (BSA) and eight repeats protein L structure, with a fine-tailored wide range of stiffness (2-35 kPa). Together with protein engineering technologies, this method will open new routes in developing and investigating tunable protein-based hydrogels and extend their application toward new horizons.
Collapse
Affiliation(s)
- Marina Slawinski
- Department
of Physics, University of Wisconsin—Milwaukee, 3135 N. Maryland Ave, Milwaukee, Wisconsin 53211, United States
| | - Maria Kaeek
- Department
of Materials Science and Engineering, Technion
Israel Institute of Technology, Haifa 32000, Israel
| | - Yair Rajmiel
- Department
of Materials Science and Engineering, Technion
Israel Institute of Technology, Haifa 32000, Israel
| | - Luai R. Khoury
- Department
of Materials Science and Engineering, Technion
Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
29
|
El Hamoui O, Saydé T, Svahn I, Gudin A, Gontier E, Le Coustumer P, Verget J, Barthélémy P, Gaudin K, Battu S, Lespes G, Alies B. Nucleoside-Derived Low-Molecular-Weight Gelators as a Synthetic Microenvironment for 3D Cell Culture. ACS Biomater Sci Eng 2022; 8:3387-3398. [PMID: 35772731 DOI: 10.1021/acsbiomaterials.2c00308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For the last few decades, many efforts have been made in developing cell culture methods in order to overcome the biological limitations of the conventional two-dimensional culture. This paradigm shift is driven by a large amount of new hydrogel-based systems for three-dimensional culture, among other systems, since they are known to mimic some living tissue properties. One class of hydrogel precursors has received interest in the field of biomaterials, low-molecular-weight gelators (LMWGs). In comparison to polymer gels, LMWG gels are formed by weak interactions upon an external trigger between the molecular subunits, giving them the ability to reverse the gelation, thus showing potential for many applications of practical interest. This study presents the use of the nucleoside derivative subclass of LMWGs, which are glyco-nucleo-bola-amphiphiles, as a proof of concept of a 3D cell culture scaffold. Physicochemical characterization was performed in order to reach the optimal features to fulfill the requirements of the cell culture microenvironment, in terms of the mechanical properties, architecture, molecular diffusion, porosity, and experimental practicality. The retained conditions were tested by culturing glioblastoma cells for over a month. The cell viability, proliferation, and spatial organization showed during the experiments demonstrate the proof of concept of nucleoside-derived LMWGs as a soft 3D cell culture scaffold. One of the hydrogels tested permits cell proliferation and spheroidal organization over the entire culture time. These systems offer many advantages as they consume very few matters within the optimal range of viscoelasticity for cell culture, and the thermoreversibility of these hydrogels permits their use with few instruments. The LMWG-based scaffold for the 3D cell culture presented in this study unlocked the ability to grow spheroids from patient cells to reach personalized therapies by dramatically reducing the variability of the lattice used.
Collapse
Affiliation(s)
- Omar El Hamoui
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.,Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), Université de Pau et des Pays de l'Adour (E2S/UPPA) CNRS UMR 5254, 2 Avenue Pierre Angot, 64053 Pau Cedex, France
| | - Tarek Saydé
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.,Université de Limoges, UMR INSERM 1308 CAPTuR, Faculté de Médecine, 87025 Limoges, France
| | - Isabelle Svahn
- Université de Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France
| | - Antoine Gudin
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Etienne Gontier
- Université de Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France
| | - Philippe Le Coustumer
- Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), Université de Pau et des Pays de l'Adour (E2S/UPPA) CNRS UMR 5254, 2 Avenue Pierre Angot, 64053 Pau Cedex, France.,Université de Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France
| | - Julien Verget
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Philippe Barthélémy
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Karen Gaudin
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Serge Battu
- Université de Limoges, UMR INSERM 1308 CAPTuR, Faculté de Médecine, 87025 Limoges, France
| | - Gaëtane Lespes
- Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), Université de Pau et des Pays de l'Adour (E2S/UPPA) CNRS UMR 5254, 2 Avenue Pierre Angot, 64053 Pau Cedex, France
| | - Bruno Alies
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| |
Collapse
|
30
|
Elsawy MA, Wychowaniec JK, Castillo Díaz LA, Smith AM, Miller AF, Saiani A. Controlling Doxorubicin Release from a Peptide Hydrogel through Fine-Tuning of Drug-Peptide Fiber Interactions. Biomacromolecules 2022; 23:2624-2634. [PMID: 35543610 PMCID: PMC9198986 DOI: 10.1021/acs.biomac.2c00356] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
![]()
Hydrogels are versatile
materials that have emerged in the last
few decades as promising candidates for a range of applications in
the biomedical field, from tissue engineering and regenerative medicine
to controlled drug delivery. In the drug delivery field, in particular,
they have been the subject of significant interest for the spatially
and temporally controlled delivery of anticancer drugs and therapeutics.
Self-assembling peptide-based hydrogels, in particular, have recently
come to the fore as potential candidate vehicles for the delivery
of a range of drugs. In order to explore how drug–peptide interactions
influence doxorubicin (Dox) release, five β-sheet-forming self-assembling
peptides with different physicochemical properties were used for the
purpose of this study, namely: FEFKFEFK (F8), FKFEFKFK (FK), FEFEFKFE
(FE), FEFKFEFKK (F8K), and KFEFKFEFKK (KF8K) (F: phenylalanine; E:
glutamic acid; K: lysine). First, Dox-loaded hydrogels were characterized
to ensure that the incorporation of the drug did not significantly
affect the hydrogel properties. Subsequently, Dox diffusion out of
the hydrogels was investigated using UV absorbance. The amount of
drug retained in F8/FE composite hydrogels was found to be directly
proportional to the amount of charge carried by the peptide fibers.
When cation−π interactions were used, the position and
number of end-lysine were found to play a key role in the retention
of Dox. In this case, the amount of Dox retained in F8/KF8K composite
hydrogels was linked to the amount of end-lysine introduced, and an
end-lysine/Dox interaction stoichiometry of 3/1 was obtained. For
pure FE and KF8K hydrogels, the maximum amount of Dox retained was
also found to be related to the overall concentration of the hydrogels
and, therefore, to the overall fiber surface area available for interaction
with the drug. For 14 mM hydrogel, ∼170–200 μM
Dox could be retained after 24 h. This set of peptides also showed
a broad range of susceptibilities to enzymatic degradation opening
the prospect of being able to control also the rate of degradation
of these hydrogels. Finally, the Dox released from the hydrogel was
shown to be active and affect 3T3 mouse fibroblasts viability in vitro.
Our study clearly shows the potential of this peptide design as a
platform for the formulation of injectable or sprayable hydrogels
for controlled drug delivery.
Collapse
Affiliation(s)
- Mohamed A Elsawy
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.,Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K
| | - Jacek K Wychowaniec
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.,Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K
| | - Luis A Castillo Díaz
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.,Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K
| | - Andrew M Smith
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.,Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K
| | - Aline F Miller
- Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K.,Department of Chemical Engineering and Analytical Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Alberto Saiani
- Department of Materials, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.,Manchester Institute of Biotechnology, Oxford Road, Manchester M13 9PL, U.K
| |
Collapse
|
31
|
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
|
32
|
Yu M, Lin S, Ge R, Xiong C, Xu L, Zhao M, Fan J. Buckwheat self-assembling peptide-based hydrogel: Preparation, characteristics and forming mechanism. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2021.107378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Giraud T, Hoschtettler P, Pickaert G, Averlant-Petit MC, Stefan L. Emerging low-molecular weight nucleopeptide-based hydrogels: state of the art, applications, challenges and perspectives. NANOSCALE 2022; 14:4908-4921. [PMID: 35319034 DOI: 10.1039/d1nr06131c] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last twenty years, low-molecular weight gelators and, in particular, peptide-based hydrogels, have drawn great attention from scientists thanks to both their inherent advantages in terms of properties and their high modularity (e.g., number and nature of the amino acids). These supramolecular hydrogels originate from specific peptide self-assembly processes that can be driven, modulated and optimized via specific chemical modifications brought to the peptide sequence. Among them, the incorporation of nucleobases, another class of biomolecules well-known for their abilities to self-assemble, has recently appeared as a new promising and burgeoning approach to finely design supramolecular hydrogels. In this minireview, we would like to highlight the interest, high potential, applications and perspectives of these innovative and emerging low-molecular weight nucleopeptide-based hydrogels.
Collapse
Affiliation(s)
- Tristan Giraud
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France.
| | | | | | | | - Loic Stefan
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France.
| |
Collapse
|
34
|
Lachowski D, Matellan C, Gopal S, Cortes E, Robinson BK, Saiani A, Miller AF, Stevens MM, del Río Hernández AE. Substrate Stiffness-Driven Membrane Tension Modulates Vesicular Trafficking via Caveolin-1. ACS NANO 2022; 16:4322-4337. [PMID: 35255206 PMCID: PMC9007531 DOI: 10.1021/acsnano.1c10534] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Liver fibrosis, a condition characterized by extensive deposition and cross-linking of extracellular matrix (ECM) proteins, is idiosyncratic in cases of chronic liver injury. The dysregulation of ECM remodeling by hepatic stellate cells (HSCs), the main mediators of fibrosis, results in an elevated ECM stiffness that drives the development of chronic liver disease such as cirrhosis and hepatocellular carcinoma. Tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) is a key element in the regulation of ECM remodeling, which modulates the degradation and turnover of ECM components. We have previously reported that a rigid, fibrotic-like substrate can impact TIMP-1 expression at the protein level in HSCs without altering its mRNA expression. While HSCs are known to be highly susceptible to mechanical stimuli, the mechanisms through which mechanical cues regulate TIMP-1 at the post-translational level remain unclear. Here, we show a mechanism of regulation of plasma membrane tension by matrix stiffness. We found that this effect is orchestrated by the β1 integrin/RhoA axis and results in elevated exocytosis and secretion of TIMP-1 in a caveolin-1- and dynamin-2-dependent manner. We then show that TIMP-1 and caveolin-1 expression increases in cirrhosis and hepatocellular carcinoma. These conditions are associated with fibrosis, and this effect can be recapitulated in 3D fibrosis models consisting of hepatic stellate cells encapsulated in a self-assembling polypeptide hydrogel. This work positions stiffness-dependent membrane tension as a key regulator of enzyme secretion and function and a potential target for therapeutic strategies that aim at modulating ECM remodeling in chronic liver disease.
Collapse
Affiliation(s)
- Dariusz Lachowski
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Carlos Matellan
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Sahana Gopal
- Department
of Materials, Department of Bioengineering and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Ernesto Cortes
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Benjamin K. Robinson
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alberto Saiani
- Department
of Materials and Manchester Institute of Biotechnology, Faculty of
Science and Engineering, The University
of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Aline F. Miller
- Department
of Chemical Engineering and Manchester Institute of Biotechnology,
Faculty of Science and Engineering, The
University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
BIOGEL, Mereside, Alderley Park, Alderley Edge, Cheshire SK10 4TG, United Kingdom
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering and Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E. del Río Hernández
- Cellular
and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
35
|
Hu XX, Zhang M, Xu F. Benzylic bromide induced peptide cross-linking for nanofiber assembly. NEW J CHEM 2022. [DOI: 10.1039/d2nj04355f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Herein, we report a cross-linking method for the assembly of coiled-coil peptides into long fibers under mild conditions.
Collapse
Affiliation(s)
- Xiao-Xiang Hu
- Ministry of Education Key Laboratory of Industrial Biotechnology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Meng Zhang
- Ministry of Education Key Laboratory of Industrial Biotechnology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Fei Xu
- Ministry of Education Key Laboratory of Industrial Biotechnology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| |
Collapse
|
36
|
Potjewyd G, Kellett K, Hooper N. 3D hydrogel models of the neurovascular unit to investigate blood-brain barrier dysfunction. Neuronal Signal 2021; 5:NS20210027. [PMID: 34804595 PMCID: PMC8579151 DOI: 10.1042/ns20210027] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022] Open
Abstract
The neurovascular unit (NVU), consisting of neurons, glial cells, vascular cells (endothelial cells, pericytes and vascular smooth muscle cells (VSMCs)) together with the surrounding extracellular matrix (ECM), is an important interface between the peripheral blood and the brain parenchyma. Disruption of the NVU impacts on blood-brain barrier (BBB) regulation and underlies the development and pathology of multiple neurological disorders, including stroke and Alzheimer's disease (AD). The ability to differentiate induced pluripotent stem cells (iPSCs) into the different cell types of the NVU and incorporate them into physical models provides a reverse engineering approach to generate human NVU models to study BBB function. To recapitulate the in vivo situation such NVU models must also incorporate the ECM to provide a 3D environment with appropriate mechanical and biochemical cues for the cells of the NVU. In this review, we provide an overview of the cells of the NVU and the surrounding ECM, before discussing the characteristics (stiffness, functionality and porosity) required of hydrogels to mimic the ECM when incorporated into in vitro NVU models. We summarise the approaches available to measure BBB functionality and present the techniques in use to develop robust and translatable models of the NVU, including transwell models, hydrogel models, 3D-bioprinting, microfluidic models and organoids. The incorporation of iPSCs either without or with disease-specific genetic mutations into these NVU models provides a platform in which to study normal and disease mechanisms, test BBB permeability to drugs, screen for new therapeutic targets and drugs or to design cell-based therapies.
Collapse
Affiliation(s)
- Geoffrey Potjewyd
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Katherine A.B. Kellett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, U.K
| |
Collapse
|
37
|
Functionalized organic nanotubes with highly tunable crosslinking site density for mechanical enhancement and pH-controlled drug release of nanocomposite hydrogels. Polym J 2021. [DOI: 10.1038/s41428-021-00556-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Sheehan F, Sementa D, Jain A, Kumar M, Tayarani-Najjaran M, Kroiss D, Ulijn RV. Peptide-Based Supramolecular Systems Chemistry. Chem Rev 2021; 121:13869-13914. [PMID: 34519481 DOI: 10.1021/acs.chemrev.1c00089] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peptide-based supramolecular systems chemistry seeks to mimic the ability of life forms to use conserved sets of building blocks and chemical reactions to achieve a bewildering array of functions. Building on the design principles for short peptide-based nanomaterials with properties, such as self-assembly, recognition, catalysis, and actuation, are increasingly available. Peptide-based supramolecular systems chemistry is starting to address the far greater challenge of systems-level design to access complex functions that emerge when multiple reactions and interactions are coordinated and integrated. We discuss key features relevant to systems-level design, including regulating supramolecular order and disorder, development of active and adaptive systems by considering kinetic and thermodynamic design aspects and combinatorial dynamic covalent and noncovalent interactions. Finally, we discuss how structural and dynamic design concepts, including preorganization and induced fit, are critical to the ability to develop adaptive materials with adaptive and tunable photonic, electronic, and catalytic properties. Finally, we highlight examples where multiple features are combined, resulting in chemical systems and materials that display adaptive properties that cannot be achieved without this level of integration.
Collapse
Affiliation(s)
- Fahmeed Sheehan
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States.,Department of Chemistry, Hunter College City University of New York 695 Park Avenue, New York, New York 10065, United States.,Ph.D. Program in Chemistry The Graduate Center of the City University of New York 365 fifth Avenue, New York, New York 10016, United States
| | - Deborah Sementa
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States
| | - Ankit Jain
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States
| | - Mohit Kumar
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States.,Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona 08028, Spain
| | - Mona Tayarani-Najjaran
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States.,Department of Chemistry, Hunter College City University of New York 695 Park Avenue, New York, New York 10065, United States.,Ph.D. Program in Chemistry The Graduate Center of the City University of New York 365 fifth Avenue, New York, New York 10016, United States
| | - Daniela Kroiss
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States.,Department of Chemistry, Hunter College City University of New York 695 Park Avenue, New York, New York 10065, United States.,Ph.D. Program in Biochemistry The Graduate Center of the City University of New York 365 5th Avenue, New York, New York 10016, United States
| | - Rein V Ulijn
- Advanced Science Research Center (ASRC) at the Graduate Center City University of New York 85 St. Nicholas Terrace New York, New York 10031, United States.,Department of Chemistry, Hunter College City University of New York 695 Park Avenue, New York, New York 10065, United States.,Ph.D. Program in Chemistry The Graduate Center of the City University of New York 365 fifth Avenue, New York, New York 10016, United States.,Ph.D. Program in Biochemistry The Graduate Center of the City University of New York 365 5th Avenue, New York, New York 10016, United States
| |
Collapse
|
39
|
Quantitative nanomechanical properties evaluation of a family of β-sheet peptide fibres using rapid bimodal AFM. J Mech Behav Biomed Mater 2021; 124:104776. [PMID: 34479107 DOI: 10.1016/j.jmbbm.2021.104776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 11/20/2022]
Abstract
Self-assembling peptides have become important building blocks for materials design (e.g. hydrogels) and play a crucial role in a range of diseases including Alzheimer and Parkinson. In this context, accessing the nanomechanical properties of ubiquitous β-sheet rich nanofibres (e.g.: amyloids) is key to the formulation of materials and design of therapies. Although the bulk mechanical properties of hydrogels can easily be accessed using common techniques and equipment, the mechanical properties of their constituent fibres, in particular if with radii in the nanometre scale, are more challenging to measure and estimate. In this work we show for the first time how the rapid nanomechanical mapping technique: amplitude modulation-frequency modulation (AM-FM), can be used to determine the heights, Young's moduli and viscosity coefficients of a series of β-sheet peptide nanofibres with high statistical confidence. Our results show how peptide sequence and in particular length, charge and interaction with the substrate affect the viscoelastic properties of the peptide fibres.
Collapse
|
40
|
Zhu H, Gu D, Rao Z, Li Y, Liu Y, Hao J. Design of gel-to-sol UCST transition peptides by controlling polypeptide β-sheet nanostructures. Polym J 2021. [DOI: 10.1038/s41428-021-00490-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
41
|
Janković P, Šantek I, Pina AS, Kalafatovic D. Exploiting Peptide Self-Assembly for the Development of Minimalistic Viral Mimetics. Front Chem 2021; 9:723473. [PMID: 34395387 PMCID: PMC8355586 DOI: 10.3389/fchem.2021.723473] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/15/2021] [Indexed: 12/03/2022] Open
Abstract
Viruses are natural supramolecular nanostructures that form spontaneously by molecular self-assembly of complex biomolecules. Peptide self-assembly is a versatile tool that allows mimicking viruses by creating their simplified versions through the design of functional, supramolecular materials with modularity, tunability, and responsiveness to chemical and physical stimuli. The main challenge in the design and fabrication of peptide materials is related to the precise control between the peptide sequence and its resulting supramolecular morphology. We provide an overview of existing sequence patterns employed for the development of spherical and fibrillar peptide assemblies that can act as viral mimetics, offering the opportunity to tackle the challenges of viral infections.
Collapse
Affiliation(s)
| | - Iva Šantek
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ana Sofia Pina
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
- UCIBIO – Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | | |
Collapse
|
42
|
Hughes MD, Hanson BS, Cussons S, Mahmoudi N, Brockwell DJ, Dougan L. Control of Nanoscale In Situ Protein Unfolding Defines Network Architecture and Mechanics of Protein Hydrogels. ACS NANO 2021; 15:11296-11308. [PMID: 34214394 PMCID: PMC8320229 DOI: 10.1021/acsnano.1c00353] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/15/2021] [Indexed: 05/10/2023]
Abstract
Hierarchical assemblies of proteins exhibit a wide-range of material properties that are exploited both in nature and by artificially by humankind. However, little is understood about the importance of protein unfolding on the network assembly, severely limiting opportunities to utilize this nanoscale transition in the development of biomimetic and bioinspired materials. Here we control the force lability of a single protein building block, bovine serum albumin (BSA), and demonstrate that protein unfolding plays a critical role in defining the architecture and mechanics of a photochemically cross-linked native protein network. The internal nanoscale structure of BSA contains "molecular reinforcement" in the form of 17 covalent disulphide "nanostaples", preventing force-induced unfolding. Upon addition of reducing agents, these nanostaples are broken rendering the protein force labile. Employing a combination of circular dichroism (CD) spectroscopy, small-angle scattering (SAS), rheology, and modeling, we show that stapled protein forms reasonably homogeneous networks of cross-linked fractal-like clusters connected by an intercluster region of folded protein. Conversely, in situ protein unfolding results in more heterogeneous networks of denser fractal-like clusters connected by an intercluster region populated by unfolded protein. In addition, gelation-induced protein unfolding and cross-linking in the intercluster region changes the hydrogel mechanics, as measured by a 3-fold enhancement of the storage modulus, an increase in both the loss ratio and energy dissipation, and markedly different relaxation behavior. By controlling the protein's ability to unfold through nanoscale (un)stapling, we demonstrate the importance of in situ unfolding in defining both network architecture and mechanics, providing insight into fundamental hierarchical mechanics and a route to tune biomaterials for future applications.
Collapse
Affiliation(s)
- Matt D.
G. Hughes
- School of
Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Benjamin S. Hanson
- School of
Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- Astbury Centre
for Structural Molecular Biology, University
of Leeds, Leeds LS2 9JT, U.K.
| | - Sophie Cussons
- Astbury Centre
for Structural Molecular Biology, University
of Leeds, Leeds LS2 9JT, U.K.
- School of
Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Najet Mahmoudi
- ISIS Neutron
and Muon Spallation Source, STFC Rutherford
Appleton Laboratory, Oxfordshire OX11 0QX, U.K.
| | - David J. Brockwell
- Astbury Centre
for Structural Molecular Biology, University
of Leeds, Leeds LS2 9JT, U.K.
- School of
Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Lorna Dougan
- School of
Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, U.K.
- Astbury Centre
for Structural Molecular Biology, University
of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
43
|
Self-Assembling Polypeptide Hydrogels as a Platform to Recapitulate the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13133286. [PMID: 34209094 PMCID: PMC8267709 DOI: 10.3390/cancers13133286] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The tumor microenvironment is characterized by increased tissue stiffness, low (acidic) pH, and elevated temperature, all of which contribute to the development of cancer. Improving our in vitro models of cancer, therefore, requires the development of cell culture platforms that can mimic these microenvironmental properties. Here, we study a new biomaterial composed of short amino acid chains that self-assemble into a fibrous hydrogel network. This material enables simultaneous and independent tuning of substrate rigidity, extracellular pH, and temperature, allowing us to mimic both healthy tissues and the tumor microenvironment. We used this platform to study the effect of these conditions on pancreatic cancer cells and found that high substrate rigidity and low pH promote proliferation and survival of cancer cells and activate important signaling pathways associated with cancer progression. Abstract The tumor microenvironment plays a critical role in modulating cancer cell migration, metabolism, and malignancy, thus, highlighting the need to develop in vitro culture systems that can recapitulate its abnormal properties. While a variety of stiffness-tunable biomaterials, reviewed here, have been developed to mimic the rigidity of the tumor extracellular matrix, culture systems that can recapitulate the broader extracellular context of the tumor microenvironment (including pH and temperature) remain comparably unexplored, partially due to the difficulty in independently tuning these parameters. Here, we investigate a self-assembled polypeptide network hydrogel as a cell culture platform and demonstrate that the culture parameters, including the substrate stiffness, extracellular pH and temperature, can be independently controlled. We then use this biomaterial as a cell culture substrate to assess the effect of stiffness, pH and temperature on Suit2 cells, a pancreatic cancer cell line, and demonstrate that these microenvironmental factors can regulate two critical transcription factors in cancer: yes-associated protein 1 (YAP) and hypoxia inducible factor (HIF-1A).
Collapse
|
44
|
Giraud T, Bouguet-Bonnet S, Stébé MJ, Richaudeau L, Pickaert G, Averlant-Petit MC, Stefan L. Co-assembly and multicomponent hydrogel formation upon mixing nucleobase-containing peptides. NANOSCALE 2021; 13:10566-10578. [PMID: 34100504 DOI: 10.1039/d1nr02417e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Peptide-based hydrogels are physical gels formed through specific supramolecular self-assembling processes, leading to ordered nanostructures which constitute the water entrapping scaffold of the soft material. Thanks to the inherent properties of peptides, these hydrogels are highly considered in the biomedical domain and open new horizons in terms of application in advanced therapies and biotechnologies. The use of one, and only one, native peptide to formulate a gel is by far the most reported approach to design such materials, but suffers from several limitations, including in terms of mechanical properties. To improve peptide-based hydrogels interest and give rise to innovative properties, several strategies have been proposed in the recent years, and the development of multicomponent peptide-based hydrogels appears as a promising and relevant strategy. Indeed, mixing two or more compounds to develop new materials is a much-used approach that has proven its effectiveness in a wide variety of domains, including polymers, composites and alloys. While still limited to a handful of examples, we would like to report herein on the formulation and the comprehensive study of multicomponent hybrid DNA-nucleobase/peptide-based hydrogels using a multiscale approach based on a large panel of analytical techniques (i.e., rheometry, proton relaxometry, SAXS, electronic microscopy, infrared, circular dichroism, fluorescence, Thioflavin T assays). Among the six multicomponent systems studied, the results highlight the synergistic role of the presence of the two complementary DNA-nucleobases (i.e., adenine/thymine and guanine/cytosine) on the co-assembling process from structural (e.g., morphology of the nanoobjects) to physicochemical (e.g., kinetics of formation, fluorescence properties) and mechanical (e.g., stiffness, resistance to external stress) properties. All the data confirm the relevance of the multicomponent peptide-based approach in the design of innovative hydrogels and bring another brick in the wall of the understanding of these complex and promising systems.
Collapse
Affiliation(s)
- Tristan Giraud
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France.
| | | | | | | | | | | | - Loic Stefan
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France.
| |
Collapse
|
45
|
Dual thermoresponsive mPEG-b-poly(O-benzyl-l-threonine acid) hydrogel based on β-sheet nano-structural disassembly and PEG dehydration. POLYMER 2021. [DOI: 10.1016/j.polymer.2021.123841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Fu K, Wu H, Su Z. Self-assembling peptide-based hydrogels: Fabrication, properties, and applications. Biotechnol Adv 2021; 49:107752. [PMID: 33838284 DOI: 10.1016/j.biotechadv.2021.107752] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/02/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
The hierarchical formation of self-assembling peptide-based hydrogels (SAPHs) starts from peptide to nanofibers, following with the entanglement into hydrogels with nanofibrous network. Such characteristic structure and extraordinary biocompatibility, and the peptide components endow the SAPHs with diverse applications in biotechnological field. Therefore, the thorough comprehension of SAPHs is significant to broadening their application. In this review, fabrication, properties, and biological applications of the SAPHs are introduced, and the factors influencing the synthesis process as well as the properties of the SAPHs products are also systematically explained. Meanwhile, we conclude the problems to be solved and provide our perspective to the future development of SAPHs in the biotechnology.
Collapse
Affiliation(s)
- Kun Fu
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hanguang Wu
- Beijing Key Laboratory of Clothing Materials R & D and Assessment, Beijing Institute of Fashion Technology, 100029 Beijing, China.
| | - Zhiqiang Su
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
47
|
Ding J, Zhang H, Wang W, Zhu Y, Wang Q, Wang A. Synergistic effect of palygorskite nanorods and ion crosslinking to enhance sodium alginate-based hydrogels. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
48
|
Saydé T, El Hamoui O, Alies B, Gaudin K, Lespes G, Battu S. Biomaterials for Three-Dimensional Cell Culture: From Applications in Oncology to Nanotechnology. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:481. [PMID: 33668665 PMCID: PMC7917665 DOI: 10.3390/nano11020481] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Three-dimensional cell culture has revolutionized cellular biology research and opened the door to novel discoveries in terms of cellular behavior and response to microenvironment stimuli. Different types of 3D culture exist today, including hydrogel scaffold-based models, which possess a complex structure mimicking the extracellular matrix. These hydrogels can be made of polymers (natural or synthetic) or low-molecular weight gelators that, via the supramolecular assembly of molecules, allow the production of a reproducible hydrogel with tunable mechanical properties. When cancer cells are grown in this type of hydrogel, they develop into multicellular tumor spheroids (MCTS). Three-dimensional (3D) cancer culture combined with a complex microenvironment that consists of a platform to study tumor development and also to assess the toxicity of physico-chemical entities such as ions, molecules or particles. With the emergence of nanoparticles of different origins and natures, implementing a reproducible in vitro model that consists of a bio-indicator for nano-toxicity assays is inevitable. However, the maneuver process of such a bio-indicator requires the implementation of a repeatable system that undergoes an exhaustive follow-up. Hence, the biggest challenge in this matter is the reproducibility of the MCTS and the associated full-scale characterization of this system's components.
Collapse
Affiliation(s)
- Tarek Saydé
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Limoges, France;
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Omar El Hamoui
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
- CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, Université de Pau et des Pays de l’Adour (E2S/UPPA), 2 Avenue Pierre Angot, 64053 Pau, France
| | - Bruno Alies
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Karen Gaudin
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Gaëtane Lespes
- CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, Université de Pau et des Pays de l’Adour (E2S/UPPA), 2 Avenue Pierre Angot, 64053 Pau, France
| | - Serge Battu
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Limoges, France;
| |
Collapse
|
49
|
El Hamoui O, Gaudin K, Battu S, Barthélémy P, Lespes G, Alies B. Self-Assembly of Nucleoside-Derived Low-Molecular-Weight Gelators: A Thermodynamics and Kinetics Study on Different Length Scales. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:297-310. [PMID: 33350837 DOI: 10.1021/acs.langmuir.0c02894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Biocompatible materials are of paramount importance in numerous fields. Unlike chemically bridge polymer-based hydrogels, low-molecular-weight gelators can form a reversible hydrogel as their structures rely on noncovalent interaction. Although many applications with this type of hydrogel can be envisioned, we still lack their understanding due to the complexity of their self-assembly process and the difficulty in predicting their behaviors (transition temperature, gelation kinetics, the impact of solvent, etc.). In this study, we extend the investigations of a series of nucleoside-derived gelators, which only differ by subtle chemical modifications. Using a multitechnique approach, we determined their thermodynamic and kinetic features on various scale (molecular to macro) in different conditions. Monitored at the supramolecular level by circular dichroism as well as macroscopic scales by rheology and turbidimetry, we found out that the sol-gel and gel-sol transitions are greatly dependent on the concentration and on the mechanisms that are probed. Self-assembly kinetics depends on hydrogel molecules and is modulated by temperature and solvent. This fundamental study provides insight on the impact of some parameters on the gelation process, such as concentration, cooling rate, and the nature of the solvent.
Collapse
Affiliation(s)
- Omar El Hamoui
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
- Université de Pau et des Pays de l'Adour (E2S/UPPA) CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), UMR 5254, 2 Avenue Pierre Angot, 64053 Pau Cedex, France
| | - Karen Gaudin
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Serge Battu
- EA3842- CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Limoges Cedex, France
| | - Philippe Barthélémy
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| | - Gaëtane Lespes
- Université de Pau et des Pays de l'Adour (E2S/UPPA) CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux (IPREM), UMR 5254, 2 Avenue Pierre Angot, 64053 Pau Cedex, France
| | - Bruno Alies
- Université de Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France
| |
Collapse
|
50
|
Abstract
Supramolecular biopolymers (SBPs) are those polymeric units derived from macromolecules that can assemble with each other by noncovalent interactions. Macromolecular structures are commonly found in living systems such as proteins, DNA/RNA, and polysaccharides. Bioorganic chemistry allows the generation of sequence-specific supramolecular units like SBPs that can be tailored for novel applications in tissue engineering (TE). SBPs hold advantages over other conventional polymers previously used for TE; these materials can be easily functionalized; they are self-healing, biodegradable, stimuli-responsive, and nonimmunogenic. These characteristics are vital for the further development of current trends in TE, such as the use of pluripotent cells for organoid generation, cell-free scaffolds for tissue regeneration, patient-derived organ models, and controlled delivery systems of small molecules. In this review, we will analyse the 3 subtypes of SBPs: peptide-, nucleic acid-, and oligosaccharide-derived. Then, we will discuss the role that SBPs will be playing in TE as dynamic scaffolds, therapeutic scaffolds, and bioinks. Finally, we will describe possible outlooks of SBPs for TE.
Collapse
|