1
|
Quan MC, Mai DJ. Biomolecular Actuators for Soft Robots. Chem Rev 2025; 125:4974-5002. [PMID: 40331746 DOI: 10.1021/acs.chemrev.4c00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Biomolecules present promising stimuli-responsive mechanisms to revolutionize soft actuators. Proteins, peptides, and nucleic acids foster specific intermolecular interactions, and their boundless sequence design spaces encode precise actuation capabilities. Drawing inspiration from nature, biomolecular actuators harness existing stimuli-responsive properties to meet the needs of diverse applications. This review features biomolecular actuators that respond to a wide variety of stimuli to drive both user-directed and autonomous actuation. We discuss how advances in biomaterial fabrication accelerate prototyping of precise, custom actuators, and we identify biomolecules with untapped actuation potential. Finally, we highlight opportunities for multifunctional and reconfigurable biomolecules to improve the versatility and sustainability of next-generation soft actuators.
Collapse
Affiliation(s)
- Michelle C Quan
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Danielle J Mai
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
2
|
Li S, Liu H, Fang Y, Li Y, Zhou L, Chen D, Liang J, Wang H. Programming two-component peptide self-assembly by tuning the hydrophobic linker. Faraday Discuss 2025. [PMID: 40366063 DOI: 10.1039/d4fd00209a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Molecular self-assembly enables the formation of intricate networks through non-covalent interactions, serving as a key strategy for constructing structures ranging from molecules to macroscopic forms. While zero-dimensional and one-dimensional nanostructures have been widely achieved, two-dimensional nanostrip structures present unique advantages in biomedical and other applications due to their high surface area and potential for functionalization. However, their efficient design and precise regulation remain challenging. This study systematically explores how different hydrophobic amino acid linkers impact the microscopic morphology in two-component co-assembly systems with strong electrostatic interactions. The introduction of the AA linker resulted in distinctive 2D nanostrips, which stacked to form bilayer sheets, whereas VV, LL, and NleNle linkers formed one-dimensional fibers. In contrast, GG and PP linkers did not produce stable aggregates. Our findings highlight the role of intermolecular interactions in the development of 2D assemblies, providing new insights into the design and application of 2D materials.
Collapse
Affiliation(s)
- Sangshuang Li
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Huayang Liu
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Yu Fang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Yaoting Li
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Laicheng Zhou
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Dinghao Chen
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Juan Liang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Huaimin Wang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| |
Collapse
|
3
|
Jafary Aryan N, Mehralitabar H, Kazemi Noureini S, Kazemi Beydokhti A. A new design and computational survey on RGD biofunctionalized RADA16-I self-assembling peptide for tissue engineering applications. Int J Biol Macromol 2025; 307:142071. [PMID: 40090651 DOI: 10.1016/j.ijbiomac.2025.142071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
Tissue engineering constantly needs innovative and biocompatible materials, and peptide-based materials seem very inspiring. Here we developed two new self-assembling peptides based on RADA16-I and RGD peptides and studied their potential in forming nanofibers under various conditions using all-atom and coarse-grained molecular dynamics simulation methods. First, a double-tailed RGD (dtRGD) peptide was designed by attaching two RADA16-I tails to an RGD-containing loop in which two disulfide bonds stabilized the loop integrity. In the second design, we bonded one side of the loop to the DA16-I tail (otRGD). The dtRGD peptides exhibited a remarkable propensity to form beta-sheet structures during all-atom MD simulations, starting from the initial random coil structure. The most promising outcomes in nanofiber formation were observed when simulating these peptides in a salt concentration that mimics the extracellular matrix. The representation of the RGD epitope was also significantly evident under these conditions. In the otRGD design, the final structure displayed a globular-like morphology, predominantly possessing coils and alpha-helices secondary structures, while maintaining effective RGD peptide exposure. This investigation signified the possibility of a new RGD representing biomaterial for tissue engineering purposes, however, further theoretical and experimental investigations are imperative to unlock their capabilities and applications.
Collapse
Affiliation(s)
- Nasrin Jafary Aryan
- Department of Chemical Engineering, School of Petroleum and Petrochemical Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Havva Mehralitabar
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Sakineh Kazemi Noureini
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Sabzevar 9617976487, Iran.
| | - Amin Kazemi Beydokhti
- Department of Chemical Engineering, School of Petroleum and Petrochemical Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| |
Collapse
|
4
|
Verbraeken B, Plazier M, Put E, Meeuws S, Roosen G, Ughratdar I, Zisakis A, Vergani F, Ganau M, Plaha P, Apostolopoulos V, Feyen B, Raymaekers V, Aboukais R, Menovsky T. Self-Assembling Peptide IEIK13 for Intraoperative Hemostasis in Cranial Neurosurgery: Clinical Trial on Efficacy and Safety. World Neurosurg 2025; 197:123856. [PMID: 40054845 DOI: 10.1016/j.wneu.2025.123856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVE This article describes the results of the first-in-human prospective clinical trial of a hemostatic hydrogel of the self-assembling peptide IEIK13 for hemostasis in intracranial surgery. Both safety and efficacy of IEIK13 for intraoperative hemostasis of oozing bleeding were evaluated in this multicenter trial. METHODS A total of 80 participants underwent a surgical intervention, during which 203 intradural bleeding sites were treated with IEIK13. Hemostasis was evaluated at 30 seconds and again at 1, 2, 3, and 6 minutes after application. A performance goal of >70% of bleeding sites reaching hemostasis within 3 minutes after application of IEIK13 was used to demonstrate noninferiority in statistical analysis. Clinical safety evaluation was performed postoperatively, at hospital discharge, and at 1 and 3 months after surgery. This included follow-up radiological imaging within the first 72 hours postoperatively and at 3 months. RESULTS Hemostasis was achieved within 3 minutes in 94.1% of bleeding sites, and within 6 minutes in 95.6% of bleeding sites. Subanalysis revealed that hemostasis occurred within the first minute in 89.2% of cases. There were no intraoperative device deficiencies. Results of the safety assessment did not raise any specific concerns. The nature and rate of adverse events did not significantly differ from what is typically expected in neurosurgical practice. CONCLUSIONS IEIK13 is effective and safe for hemostasis of oozing bleeding during intracranial neurosurgery. Based on this trial, the transparent IEIK13 hydrogel is a useful addition to the neurosurgical hemostasis toolbox.
Collapse
Affiliation(s)
- Barbara Verbraeken
- Department of Neurosurgery, Antwerp University Hospital (UZA), Antwerp, Belgium; Department of Pathology, Antwerp University Hospital (UZA), Antwerp, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp (UA), Antwerp, Belgium.
| | - Mark Plazier
- Department of Neurosurgery, Jessa Hospital, Limburg, Belgium
| | - Eric Put
- Department of Neurosurgery, Jessa Hospital, Limburg, Belgium
| | - Sacha Meeuws
- Department of Neurosurgery, Jessa Hospital, Limburg, Belgium
| | - Gert Roosen
- Department of Neurosurgery, Jessa Hospital, Limburg, Belgium
| | - Ismail Ughratdar
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Athanasios Zisakis
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Francesco Vergani
- Department of Neurosurgery, King's College London Hospital, London, United Kingdom
| | - Mario Ganau
- John Radcliffe Hospital, Oxford University, Oxford, United Kingdom
| | - Puneet Plaha
- John Radcliffe Hospital, Oxford University, Oxford, United Kingdom
| | | | - Bart Feyen
- Department of Neurosurgery, Antwerp University Hospital (UZA), Antwerp, Belgium
| | - Vincent Raymaekers
- Department of Neurosurgery, Antwerp University Hospital (UZA), Antwerp, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Rabih Aboukais
- Department of Neurosurgery, Lille University Hospital, Lille, France
| | - Tomas Menovsky
- Department of Neurosurgery, Antwerp University Hospital (UZA), Antwerp, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp (UA), Antwerp, Belgium
| |
Collapse
|
5
|
Souza PCT, Borges-Araújo L, Brasnett C, Moreira RA, Grünewald F, Park P, Wang L, Razmazma H, Borges-Araújo AC, Cofas-Vargas LF, Monticelli L, Mera-Adasme R, Melo MN, Wu S, Marrink SJ, Poma AB, Thallmair S. GōMartini 3: From large conformational changes in proteins to environmental bias corrections. Nat Commun 2025; 16:4051. [PMID: 40307210 PMCID: PMC12043922 DOI: 10.1038/s41467-025-58719-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Coarse-grained modeling has become an important tool to supplement experimental measurements, allowing access to spatio-temporal scales beyond all-atom based approaches. The GōMartini model combines structure- and physics-based coarse-grained approaches, balancing computational efficiency and accurate representation of protein dynamics with the capabilities of studying proteins in different biological environments. This paper introduces an enhanced GōMartini model, which combines a virtual-site implementation of Gō models with Martini 3. The implementation has been extensively tested by the community since the release of the reparametrized version of Martini. This work demonstrates the capabilities of the model in diverse case studies, ranging from protein-membrane binding to protein-ligand interactions and AFM force profile calculations. The model is also versatile, as it can address recent inaccuracies reported in the Martini protein model. Lastly, the paper discusses the advantages, limitations, and future perspectives of the Martini 3 protein model and its combination with Gō models.
Collapse
Affiliation(s)
- Paulo C T Souza
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon, France.
- Centre Blaise Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon, France.
| | - Luís Borges-Araújo
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon, France
- Centre Blaise Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon, France
| | - Christopher Brasnett
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands
| | - Rodrigo A Moreira
- NEIKER, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Bizkaia, P812, Derio, Spain
| | - Fabian Grünewald
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, Heidelberg, Germany
| | - Peter Park
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Liguo Wang
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands
| | - Hafez Razmazma
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, Lyon, France
- Institut des Biomolecules Max Mousseron, UMR5247, CNRS, Université De Montpellier, ENSCM, 1919 Route de Mende, Montpellier, Cedex, France
| | - Ana C Borges-Araújo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Luis Fernando Cofas-Vargas
- Biosystems and Soft Matter Division, Institute of Fundamental Technological Research, Polish Academy of Sciences, ul. Pawińskiego 5B, 02-106, Warsaw, Poland
| | - Luca Monticelli
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, Lyon, France
| | - Raúl Mera-Adasme
- Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica, Chile
| | - Manuel N Melo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Sangwook Wu
- PharmCADD, Busan, Republic of Korea
- Department of Physics, Pukyong National University, Busan, Republic of Korea
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands.
| | - Adolfo B Poma
- Biosystems and Soft Matter Division, Institute of Fundamental Technological Research, Polish Academy of Sciences, ul. Pawińskiego 5B, 02-106, Warsaw, Poland.
| | - Sebastian Thallmair
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Straße 1, Frankfurt am Main, Germany.
| |
Collapse
|
6
|
Du Z, Dai J, Wang Z, Ye Z, Lei H, Guo QY, Yan XY, Li M, Li M, Zhao W, Zhang B, Huang Y, Yang Z, Huang Z, Yin P, Jiang L, Zhang R, Tang W. Engineering the Self-Assembly Pathways of POSS-Peptide Amphiphiles to Form Diverse Cross-β Structures. Angew Chem Int Ed Engl 2025; 64:e202420043. [PMID: 39715729 DOI: 10.1002/anie.202420043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Indexed: 12/25/2024]
Abstract
Cross-β structures are crucial in driving protein folding and aggregation. However, due to their strong aggregating tendency, the precise control of the self-assembly of β-sheet-forming peptides remains a challenge. We propose a molecular geometry strategy to study and control the self-assembly of cross-β structures. We conjugate the peptide with shape-persistent polyhedral oligomeric silsesquioxane (POSS), which acts as a hydrophilic head and senses the solvent environment. The POSS-peptide amphiphiles display two distinct self-assembly pathways: twisted nanoribbons transforming into either nanotubes at low water content or flat nanoribbons at high water content. The peptide packing in flat nanoribbons is predominantly modulated by POSS, diverting the system away from crystal formation, which is the absolute lowest energy state of pure peptide self-assemblies. For the first time, we have demonstrated that POSS can serve as a useful tool to adjust the interactions between cross-β strands, achieving fine-tuning of the pathway complexity (i.e., the kinetic and thermodynamic aspects of peptide self-assembly). With this versatile molecular platform incorporating multiple functionalities of POSS and programable peptide sequences, this study provides a platform to exploit cross-β-based nanomaterials with functional and pathological significance.
Collapse
Affiliation(s)
- Zhen Du
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Junhao Dai
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Zhibo Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Zushan Ye
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Huanyu Lei
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Qing-Yun Guo
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Xiao-Yun Yan
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Mu Li
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Minzhao Li
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Wangshen Zhao
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Beijing Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Yupeng Huang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Ziqiong Yang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Zongwu Huang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Panchao Yin
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Lingxiang Jiang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Rui Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Wen Tang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| |
Collapse
|
7
|
Chen T, Liu Y, Gao Z, Gao Y, Chen H, Ye H, Luo Q, Wang K, Wu D. Template-assisted Flexible-to-rigid Transition of Peptides in Head-to-tail Self-polymerization Enables Sequence-controllable and Post-modifiable Peptide Nanofibers. Angew Chem Int Ed Engl 2025; 64:e202415809. [PMID: 39266463 DOI: 10.1002/anie.202415809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/14/2024]
Abstract
Peptide-based nanofibers are promising materials for many essential applications and can be generalized into two categories, self-assembling peptide nanofibers (SAPNs) and poly(amino acid) nanofibers (PAANs). Non-covalent SAPNs are sequence-controllable, but poorly stable and not suitable for post-modification. While covalent PAANs are post-modifiable, however, their sequences are either monotonic or undefined. The nanofibers obtained by head-to-tail covalent coupling polymerization of sequence-known peptides, which we call series-connected peptide nanofibers (SCPNs), promise to have the advantages of both SAPNs and PAANs, but they are barely reported. The undesired backbiting effect during the head-to-tail polymerization is one of the possible challenges. Here, we present a template-assisted strategy to trigger the flexible-to-rigid transition of peptide units, which can avoid the backbiting effect and enable consecutive intermolecular polymerization of peptides to produce desired sequence-controlled covalent SCPNs. SCPNs are highly stable and can function as excellent parent materials for various post-processing to create diverse hierarchical materials independent of the peptide sequence. Moreover, SCPNs allow for the display of predetermined functional groups at regular intervals along the nanofibers by pre-modification of the initial peptide sequence. SCPNs represent a new category of peptide-based nanofibers with outstanding performances and vast potential.
Collapse
Affiliation(s)
- Tianzi Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yin Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Zhanshan Gao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yue Gao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Haijin Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Haonan Ye
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Qiuhao Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Dongdong Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, Sichuan, China
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
8
|
Asadi Sarabi P, Shabanpouremam M, Eghtedari AR, Barat M, Moshiri B, Zarrabi A, Vosough M. AI-Based solutions for current challenges in regenerative medicine. Eur J Pharmacol 2024; 984:177067. [PMID: 39454850 DOI: 10.1016/j.ejphar.2024.177067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
The emergence of Artificial Intelligence (AI) and its usage in regenerative medicine represents a significant opportunity that holds the promise of tackling critical challenges and improving therapeutic outcomes. This article examines the ways in which AI, including machine learning and data fusion techniques, can contribute to regenerative medicine, particularly in gene therapy, stem cell therapy, and tissue engineering. In gene therapy, AI tools can boost the accuracy and safety of treatments by analyzing extensive genomic datasets to target and modify genetic material in a precise manner. In cell therapy, AI improves the characterization and optimization of cell products like mesenchymal stem cells (MSCs) by predicting their function and potency. Additionally, AI enhances advanced microscopy techniques, enabling accurate, non-invasive and quantitative analyses of live cell cultures. AI enhances tissue engineering by optimizing biomaterial and scaffold designs, predicting interactions with tissues, and streamlining development. This leads to faster and more cost-effective innovations by decreasing trial and error. The convergence of AI and regenerative medicine holds great transformative potential, promising effective treatments and innovative therapeutic strategies. This review highlights the importance of interdisciplinary collaboration and the continued integration of AI-based technologies, such as data fusion methods, to overcome current challenges and advance regenerative medicine.
Collapse
Affiliation(s)
- Pedram Asadi Sarabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahshid Shabanpouremam
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Amir Reza Eghtedari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Mahsa Barat
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Behzad Moshiri
- School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
9
|
Pogostin BH, Godbe K, Dubackic M, Angstman I, Fox W, Giovino N, Lagator M, Payson A, LaBarca M, Frohm B, Bernfur K, Linse S, Londergan CH, Olsson U, Gentile L, Åkerfeldt KS. Insights into the Hierarchical Assembly of a Chemically Diverse Peptide Hydrogel Derived from Human Semenogelin I. ACS NANO 2024; 18:31109-31122. [PMID: 39487039 PMCID: PMC11562788 DOI: 10.1021/acsnano.4c08672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
A peptide corresponding to a 13-residue segment of the human protein semenogelin I has been shown to generate a hydrogel consisting of amyloid-like fibrils. The relative chemical diversity (compared to synthetic de novo sequences) with 11 distinct amino acids makes this peptide (P0) an ideal candidate for investigating the role of individual residues in gelation. Herein, the N-terminal residues have been sequentially removed to furnish a series of truncated peptides, P1-P10, ranging from 12 to 3 residues in length. FTIR spectroscopy investigations reveal that P0-P6 forms a β-sheet secondary structure while shorter sequences do not self-assemble. Site-specific isotope labeling of the amide backbone of P0-P2 with the IR-sensitive vibrational probe 13C═O yields FTIR spectra indicative of the initial formation of a kinetic product that slowly transforms into a structurally different thermodynamic product. The effects of the isotopic labels on the IR spectra facilitate the assignment of parallel and antiparallel structures, which are sometimes coexistent. Additional IR studies of three PheCN-labeled P0 sequences are consistent with an H-bonded β-sheet amide core, spanning the 7 central residues. The macromolecular assembly of peptides that form β-sheets was assessed by cryo-TEM, SAXS/WAXS, and rheology. Cryo-TEM images of peptides P1-P6 display μm-long nanofibrils. Peptides P0-P3 generate homogeneous hydrogels composed of colloidally stable nanofibrils, and P4-P6 undergo phase separation due to the accumulation of attractive interfibrillar interactions. Three amino acid residues, Ser39, Phe40, and Gln43, were identified to be of particular interest in the truncated peptide series as the removal of any one of them, as the sequence shortens, leads to a major change in material properties.
Collapse
Affiliation(s)
- Brett H. Pogostin
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
- Department
of Physical Chemistry, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Kerilyn Godbe
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Marija Dubackic
- Department
of Physical Chemistry, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Isabelle Angstman
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - William Fox
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Natalie Giovino
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Matija Lagator
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Abigail Payson
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Marisa LaBarca
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Birgitta Frohm
- Biochemistry
and Structural Biology, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Katja Bernfur
- Biochemistry
and Structural Biology, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Sara Linse
- Biochemistry
and Structural Biology, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Casey H. Londergan
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| | - Ulf Olsson
- Department
of Physical Chemistry, Lund University, PO Box 124, Lund SE-221 00, Sweden
| | - Luigi Gentile
- Department
of Physical Chemistry, Lund University, PO Box 124, Lund SE-221 00, Sweden
- Department
of Chemistry, University of Bari Aldo Moro, Via Orabona 4, Bari 70126, Italy
| | - Karin S. Åkerfeldt
- Department
of Chemistry, Haverford College, Haverford, Pennsylvania 19041, United States
| |
Collapse
|
10
|
Miki T, Hashimoto M, Takahashi H, Shimizu M, Nakayama S, Furuta T, Mihara H. De novo designed YK peptides forming reversible amyloid for synthetic protein condensates in mammalian cells. Nat Commun 2024; 15:8503. [PMID: 39424799 PMCID: PMC11489810 DOI: 10.1038/s41467-024-52708-5] [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: 03/01/2024] [Accepted: 09/18/2024] [Indexed: 10/21/2024] Open
Abstract
In mammalian cells, protein condensates underlie diverse cell functions. Intensive synthetic biological research has been devoted to fabricating liquid droplets using de novo peptides/proteins designed from scratch in test tubes or bacterial cells. However, the development of de novo sequences for synthetic droplets forming in eukaryotes is challenging. Here, we report YK peptides, comprising 9-15 residues of alternating repeats of tyrosine and lysine, which form reversible amyloid-like fibrils accompanied by binding with poly-anion species such as ATP. By genetically tagging the YK peptide, superfolder GFPs assemble into artificial liquid-like droplets in living cells. Rational design of the YK system allows fine-tuning of the fluidity and construction of multi-component droplets. The YK system not only facilitates intracellular reconstitution of simplified models for natural protein condensates, but it also provides a toolbox for the systematic creation of droplets with different dynamics and composition for in situ evaluation.
Collapse
Affiliation(s)
- Takayuki Miki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Masahiro Hashimoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Hiroki Takahashi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Masatoshi Shimizu
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sae Nakayama
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Tadaomi Furuta
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Hisakazu Mihara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| |
Collapse
|
11
|
Prince E. Designing Biomimetic Strain-Stiffening into Synthetic Hydrogels. Biomacromolecules 2024; 25:6283-6295. [PMID: 39356204 DOI: 10.1021/acs.biomac.4c00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Biological tissues are mechanoresponsive; that is, their properties dynamically change in response to mechanical stimuli. For example, in response to shear or elongational strain, collagen, fibrin, actin, and other filamentous biomaterials undergo dramatic strain-stiffening. Above a critical strain, their stiffness increases over orders of magnitude. While it is widely accepted that the stiffness of biological tissues impacts cell phenotype and several diseases, the biological impact of strain-stiffening remains understudied. Synthetic hydrogels that mimic the mechanoresponsive nature of biological tissues could serve as an in vitro platform for these studies. This review highlights recent efforts to mimic the strain-stiffening behavior of biological materials in synthetic hydrogels. We discuss the design principles for imparting synthetic hydrogels with biomimetic strain-stiffening, critically compare designs of strain-stiffening hydrogels that have been reported thus far, and discuss their use as in vitro platforms to probe how strain-stiffening impacts cell behavior, diseases, and other biological processes.
Collapse
Affiliation(s)
- Elisabeth Prince
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. West, N2L 3G1 Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Ave. West, N2L 3G1 Waterloo, ON, Canada
| |
Collapse
|
12
|
Soliman BG, Nguyen AK, Gooding JJ, Kilian KA. Advancing Synthetic Hydrogels through Nature-Inspired Materials Chemistry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404235. [PMID: 38896849 PMCID: PMC11486603 DOI: 10.1002/adma.202404235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/25/2024] [Indexed: 06/21/2024]
Abstract
Synthetic extracellular matrix (ECM) mimics that can recapitulate the complex biochemical and mechanical nature of native tissues are needed for advanced models of development and disease. Biomedical research has heavily relied on the use of animal-derived biomaterials, which is now impeding their translational potential and convoluting the biological insights gleaned from in vitro tissue models. Natural hydrogels have long served as a convenient and effective cell culture tool, but advances in materials chemistry and fabrication techniques now present promising new avenues for creating xenogenic-free ECM substitutes appropriate for organotypic models and microphysiological systems. However, significant challenges remain in creating synthetic matrices that can approximate the structural sophistication, biochemical complexity, and dynamic functionality of native tissues. This review summarizes key properties of the native ECM, and discusses recent approaches used to systematically decouple and tune these properties in synthetic matrices. The importance of dynamic ECM mechanics, such as viscoelasticity and matrix plasticity, is also discussed, particularly within the context of organoid and engineered tissue matrices. Emerging design strategies to mimic these dynamic mechanical properties are reviewed, such as multi-network hydrogels, supramolecular chemistry, and hydrogels assembled from biological monomers.
Collapse
Affiliation(s)
- Bram G Soliman
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Ashley K Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - J Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Kristopher A Kilian
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
13
|
Fasciano S, Wheba A, Ddamulira C, Wang S. Recent advances in scaffolding biomaterials for cultivated meat. BIOMATERIALS ADVANCES 2024; 162:213897. [PMID: 38810509 DOI: 10.1016/j.bioadv.2024.213897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/07/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
The emergence of cultivated meat provides a sustainable and ethical alternative to traditional animal agriculture, highlighting its increasing importance in the food industry. Biomaterial scaffolds are critical components in cultivated meat production for enabling cell adhesion, proliferation, differentiation, and orientation. While there's extensive research on scaffolding biomaterials, applying them to cultivated meat production poses distinct challenges, with each material offering its own set of advantages and disadvantages. This review summarizes the most recent scaffolding biomaterials used in the last five years for cell-cultured meat, detailing their respective advantages and disadvantages. We suggest future research directions and provide recommendations for scaffolds that support scalable, cost-effective, and safe high-quality meat production. Additionally, we highlight commercial challenges cultivated meat faces, encompassing bioreactor design, cell culture mediums, and regulatory and food safety issues. In summary, this review provides a comprehensive guide and valuable insights for researchers and companies in the field of cultivated meat production.
Collapse
Affiliation(s)
- Samantha Fasciano
- Department of Cellular and Molecular Biology, University of New Haven, West Haven, CT, 06516, USA
| | - Anas Wheba
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA
| | - Christopher Ddamulira
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA
| | - Shue Wang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA.
| |
Collapse
|
14
|
He W, Wang Y, Li X, Ji Y, Yuan J, Yang W, Yan S, Yan J. Sealing the Pandora's vase of pancreatic fistula through entrapping the digestive enzymes within a dextrorotary (D)-peptide hydrogel. Nat Commun 2024; 15:7235. [PMID: 39174548 PMCID: PMC11341566 DOI: 10.1038/s41467-024-51734-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 08/16/2024] [Indexed: 08/24/2024] Open
Abstract
A variety of therapeutic possibilities have emerged for skillfully regulating protein function or conformation through intermolecular interaction modulation to rectify abnormal biochemical reactions in diseases. Herein, a devised strategy of enzyme coordinators has been employed to alleviate postoperative pancreatic fistula (POPF), which is characterized by the leakage of digestive enzymes including trypsin, chymotrypsin, and lipase. The development of a dextrorotary (D)-peptide supramolecular gel (CP-CNDS) under this notion showcases its propensity for forming gels driven by intermolecular interaction. Upon POPF, CP-CNDS not only captures enzymes from solution into hydrogel, but also effectively entraps them within the internal gel, preventing their exchange with counterparts in the external milieu. As a result, CP-CNDS completely suppresses the activity of digestive enzymes, effectively alleviating POPF. Remarkably, rats with POPF treated with CP-CNDS not only survived but also made a recovery within a mere 3-day period, while mock-treated POPF rats had a survival rate of less than 5 days when experiencing postoperative pancreatic fistula, leak or abscess. Collectively, the reported CP-CNDS provides promising avenues for preventing and treating POPF, while exemplifying precision medicine-guided regulation of protein activity that effectively targets specific pathogenic molecules across multiple diseases.
Collapse
Affiliation(s)
- Wangxiao He
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China.
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| | - Yang Wang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiao Li
- Department of General Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yanlin Ji
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Juzheng Yuan
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
- Department of General Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wenguang Yang
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Siqi Yan
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jin Yan
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China.
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
15
|
Guo HD, Wu JH, Wang HJ, Tan YZ. Delivery of Stem Cells and BMP-2 With Functionalized Self-Assembling Peptide Enhances Regeneration of Infarcted Myocardium. Stem Cell Rev Rep 2024; 20:1540-1554. [PMID: 38656478 DOI: 10.1007/s12015-024-10721-7] [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] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
Stem cell transplantation is a promising therapeutic strategy for myocardial infarction (MI). However, engraftment, survival and differentiation of the transplanted stem cells in ischemic and inflammatory microenvironment are poor. We designed a novel self-assembly peptide (SAP) by modifying the peptide RADA16 with cell-adhesive motif and BMP-2 (bone morphogenetic protein-2)-binding motif. Effects of the functionalized SAP on adhesion, survival and differentiation of c-kit+ MSCs (mesenchymal stem cells) were examined. Myocardial regeneration, neovascularization and cardiac function were assessed after transplantation of the SAP loading c-kit+ MSCs and BMP-2 in rat MI models. The SAP could spontaneously assemble into well-ordered nanofibrous scaffolds. The cells adhered to the SAP scaffolds and spread well. The SAP protected the cells in the condition of hypoxia and serum deprivation. Following degradation of the SAP, BMP-2 was released sustainedly and induced c-kit+ MSCs to differentiate into cardiomyocytes. At four weeks after transplantation of the SAP loading c-kit+ MSCs and BMP-2, myocardial regeneration and angiogenesis were enhanced, and cardiac function was improved significantly. The cardiomyocytes differentiated from the engrafted c-kit+ MSCs were increased markedly. The differentiated cells connected with recipient cardiomyocytes to form gap junctions. Collagen volume was decreased dramatically. These results suggest that the functionalized SAP promotes engraftment, survival and differentiation of stem cells effectively. Local sustained release of BMP-2 with SAP is a viable strategy to enhance differentiation of the engrafted stem cells and repair of the infarcted myocardium.
Collapse
Affiliation(s)
- Hai-Dong Guo
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Jin-Hong Wu
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
- Department of Anesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China
| | - Hai-Jie Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
- Rehabilitation Therapy Department, School of Health Sciences, West Yunnan University of Applied Sciences, Dali, Yunnan Province, 671000, People's Republic of China.
| | - Yu-Zhen Tan
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
16
|
Zhao S, Xue C, Burns DC, Shoichet MS. Viscoelastic Supramolecular Hyaluronan-Peptide Cross-Linked Hydrogels. Biomacromolecules 2024; 25:3946-3958. [PMID: 38913947 DOI: 10.1021/acs.biomac.4c00095] [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: 06/26/2024]
Abstract
Viscoelasticity plays a key role in hydrogel design. We designed a physically cross-linked hydrogel with tunable viscoelasticity, comprising supramolecular-assembled peptides coupled to hyaluronan (HA), a native extracellular matrix component. We then explored the structural and molecular mechanisms underlying the mechanical properties of a series of these HA-peptide hydrogels. By modifying the peptide sequence, we modulated both long- and short-time stress relaxation rates as a way to target viscoelasticity with limited impact on stiffness, leading to gels that relax up to 60% of stress in 10 min. Gels with the highest viscoelasticity exhibited large mesh sizes and β-sheet secondary structures. The stiffness of the gel correlated with hydrogen bonding between the peptide chains. These gels are cytocompatible: highly viscoelastic gels that mimic the native skin microenvironment promote dermal fibroblast cell spreading. Moreover, HA-peptide gels enabled cell encapsulation, as shown with primary human T cells. Overall, these physically-cross-linked hydrogels enable tunable viscoelasticity that can be used to modulate cell morphology.
Collapse
Affiliation(s)
- Spencer Zhao
- Division of Engineering Science, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Department of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Chang Xue
- Department of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Darcy C Burns
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Molly S Shoichet
- Department of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| |
Collapse
|
17
|
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
|
18
|
Andretto V, Rosso A, Zilio S, Sidi-Boumedine J, Boschetti G, Sankar S, Buffier M, Miele AE, Denis M, Choffour PA, Briançon S, Nancey S, Kryza D, Lollo G. Peptide-Based Hydrogel for Nanosystems Encapsulation: the Next Generation of Localized Delivery Systems for the Treatment of Intestinal Inflammations. Adv Healthc Mater 2024; 13:e2303280. [PMID: 38445812 DOI: 10.1002/adhm.202303280] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/10/2024] [Indexed: 03/07/2024]
Abstract
Conventional therapies for inflammatory bowel diseases are mainly based on systemic treatments which cause side effects and toxicity over long-term administration. Nanoparticles appear as a valid alternative to allow a preferential accumulation in inflamed tissues following oral administration while reducing systemic drug exposure. To increase their residence time in the inflamed intestine, the nanoparticles are here associated with a hydrogel matrix. A bioadhesive peptide-based hydrogel is mixed with nanoemulsions, creating a hybrid lipid-polymer nanocomposite. Mucopenetrating nanoemulsions of 100 nm are embedded in a scaffold constituted of the self-assembling peptide hydrogel product PuraStat. The nanocomposite is fully characterized to study the impact of lipid particles in the hydrogel structure. Rheological measurements and circular dichroism analyses are performed to investigate the system's microstructure and physical properties. Biodistribution studies demonstrate that the nanocomposite acts as a depot in the stomach and facilitates the slow release of the nanoemulsions in the intestine. Efficacy studies upon oral administration of the drug-loaded system show the improvement of the disease score in a mouse model of intestinal inflammation.
Collapse
Affiliation(s)
- Valentina Andretto
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Annalisa Rosso
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
- SATT, Ouest Valorisation, 14C Rue du Patis Tatelin, Renne, 35708, France
| | - Jacqueline Sidi-Boumedine
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Gilles Boschetti
- Department of Gastroenterology, Lyon Sud Hospital, Hospices Civil de Lyon and CIRI, Lyon, 69495, France
| | - Sharanya Sankar
- 3-D Matrix Europe SAS, Medical Technology, Caluire-et-Cuire, 69300, France
| | - Marie Buffier
- 3-D Matrix Europe SAS, Medical Technology, Caluire-et-Cuire, 69300, France
| | - Adriana Erica Miele
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, ISA UMR 5280, 5 rue de la Doua, Villeurbanne, F-69100, France
- Dept Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome, I-00185, Italy
| | - Morgane Denis
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
- Antineo, R&D Department, Lyon, 69008, France
| | | | - Stéphanie Briançon
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Stéphane Nancey
- Department of Gastroenterology, Lyon Sud Hospital, Hospices Civil de Lyon and CIRI, Lyon, 69495, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
- Hospices Civils de Lyon, Lyon, 69437, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| |
Collapse
|
19
|
Liu H, Wang H. From cells to subcellular organelles: Next-generation cancer therapy based on peptide self-assembly. Adv Drug Deliv Rev 2024; 209:115327. [PMID: 38703895 DOI: 10.1016/j.addr.2024.115327] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
Due to the editability, functionality, and excellent biocompatibility of peptides, in situ self-assembly of peptides in cells is a powerful strategy for biomedical applications. Subcellular organelle targeting of peptides assemblies enables more precise drug delivery, enhances selectivity to disease cells, and mitigates drug resistance, providing an effective strategy for disease diagnosis and therapy. This reviewer first introduces the triggering conditions, morphological changes, and intracellular locations of self-assembling peptides. Then, the functions of peptide assemblies are summarized, followed by a comprehensive understanding of the interactions between peptide assemblies and subcellular organelles. Finally, we provide a brief outlook and the remaining challenges in this field.
Collapse
Affiliation(s)
- Huayang Liu
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| |
Collapse
|
20
|
Nilsson BL, Celebi Torabfam G, Dias CL. Peptide Self-Assembly into Amyloid Fibrils: Unbiased All-Atom Simulations. J Phys Chem B 2024; 128:3320-3328. [PMID: 38447080 PMCID: PMC11466223 DOI: 10.1021/acs.jpcb.3c07861] [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] [Indexed: 03/08/2024]
Abstract
Protein self-assembly plays an important role in biological systems, accounting for the formation of mesoscopic structures that can be highly symmetric as in the capsid of viruses or disordered as in molecular condensates or exhibit a one-dimensional fibrillar morphology as in amyloid fibrils. Deposits of the latter in tissues of individuals with degenerative diseases like Alzheimer's and Parkinson's has motivated extensive efforts to understand the sequence of molecular events accounting for their formation. These studies aim to identify on-pathway intermediates that may be the targets for therapeutic intervention. This detailed knowledge of fibril formation remains obscure, in part due to challenges with experimental analyses of these processes. However, important progress is being achieved for short amyloid peptides due to advances in our ability to perform completely unbiased all-atom simulations of the self-assembly process. This perspective discusses recent developments, their implications, and the hurdles that still need to be overcome to further advance the field.
Collapse
Affiliation(s)
- Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
- Materials Science Program, University of Rochester, Rochester, New York 14627-0216, United States
| | - Gizem Celebi Torabfam
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| | - Cristiano L Dias
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| |
Collapse
|
21
|
Yang Q, Miki T. Characterization of peptide-fused protein assemblies in living cells. Methods Enzymol 2024; 697:293-319. [PMID: 38816127 DOI: 10.1016/bs.mie.2024.01.022] [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: 06/01/2024]
Abstract
Assembly of de novo peptides designed from scratch is in a semi-rational manner and creates artificial supramolecular structures with unique properties. Considering that the functions of various proteins in living cells are highly regulated by their assemblies, building artificial assemblies within cells holds the potential to simulate the functions of natural protein assemblies and engineer cellular activities for controlled manipulation. How can we evaluate the self-assembly of designed peptides in cells? The most effective approach involves the genetic fusion of fluorescent proteins (FPs). Expressing a self-assembling peptide fused with an FP within cells allows for evaluating assemblies through fluorescence signal. When µm-scale assemblies such as condensates are formed, the peptide assemblies can be directly observed by imaging. For sub-µm-scale assemblies, fluorescence correlation spectroscopy analysis is more practical. Additionally, the fluorescence resonance energy transfer (FRET) signal between FPs is valuable evidence of proximity. The decrease in fluorescence anisotropy associated with homo-FRET reveals the properties of self-assembly. Furthermore, by combining two FPs, one acting as a donor and the other as an acceptor, the heteromeric interaction between two different components can be studied through the FRET signal. In this chapter, we provide detailed protocols, from designing and constructing plasmid DNA expressing the peptide-fused protein to analysis of self-assembly in living cells.
Collapse
Affiliation(s)
- Qinxuan Yang
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takayuki Miki
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan; School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
22
|
Hao Z, Feng Q, Wang Y, Wang Y, Li H, Hu Y, Chen T, Wang J, Chen R, Lv X, Yang Z, Chen J, Guo X, Li J. A parathyroid hormone related supramolecular peptide for multi-functionalized osteoregeneration. Bioact Mater 2024; 34:181-203. [PMID: 38235308 PMCID: PMC10792172 DOI: 10.1016/j.bioactmat.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/30/2023] [Accepted: 12/17/2023] [Indexed: 01/19/2024] Open
Abstract
Supramolecular peptide nanofiber hydrogels are emerging biomaterials for tissue engineering, but it is difficult to fabricate multi-functional systems by simply mixing several short-motif-modified supramolecular peptides because relatively abundant motifs generally hinder nanofiber cross-linking or the formation of long nanofiber. Coupling bioactive factors to the assembling backbone is an ideal strategy to design multi-functional supramolecular peptides in spite of challenging synthesis and purification. Herein, a multi-functional supramolecular peptide, P1R16, is developed by coupling a bioactive factor, parathyroid hormone related peptide 1 (PTHrP-1), to the basic supramolecular peptide RADA16-Ⅰ via solid-phase synthesis. It is found that P1R16 self-assembles into long nanofibers and co-assembles with RADA16-Ⅰ to form nanofiber hydrogels, thus coupling PTHrP-1 to hydrogel matrix. P1R16 nanofiber retains osteoinductive activity in a dose-dependent manner, and P1R16/RADA16-Ⅰ nanofiber hydrogels promote osteogenesis, angiogenesis and osteoclastogenesis in vitro and induce multi-functionalized osteoregeneration by intramembranous ossification and bone remodeling in vivo when loaded to collagen (Col) scaffolds. Abundant red blood marrow formation, ideal osteointegration and adapted degradation are observed in the 50% P1R16/Col scaffold group. Therefore, this study provides a promising strategy to develop multi-functional supramolecular peptides and a new method to topically administrate parathyroid hormone or parathyroid hormone related peptides for non-healing bone defects.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qinyu Feng
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xuan Lv
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhiqiang Yang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiayao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaodong Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
23
|
Daniel G, Hilan G, Ploeg L, Sabatino D. Self-assembly of amphiphilic helical-coiled peptide nanofibers and inhibition of fibril formation with curcumin. Bioorg Med Chem Lett 2024; 102:129682. [PMID: 38432287 DOI: 10.1016/j.bmcl.2024.129682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
Amphiphilic peptide sequences are conducive to secondary structures that self-assemble into higher-ordered peptide nanostructures. A select set of amphiphilic polycationic peptides displayed stable helical-coiled structures that self-assembled into peptide nanofibers. The progression of peptide fibril formation revealed short protofibrils that extended into thin filaments and into an entangled network of nanofibers over an extended (5 days) incubation period. Ligand binding with 8-anilinonaphthalene-1-sulfonic acid (ANS) and Congo Red (CR) confirmed the amphiphilic helical-coiled peptide structure assembly into nanofibers, whereas curcumin treatment led to inhibition of fibril formation. Considering the vast repertoire of fibrous biomaterials and peptide or protein (mis)folding contingent on fibril formation, this work relates the molecular interplay in between sequence composition, structural folding and the ligand binding events impacting peptide self-assembly into nanofibers.
Collapse
Affiliation(s)
- Grace Daniel
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada; Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - George Hilan
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Lisa Ploeg
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - David Sabatino
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada; Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
24
|
Ciulla MG, Marchini A, Gazzola J, Forouharshad M, Pugliese R, Gelain F. In Situ Transglutaminase Cross-Linking Improves Mechanical Properties of Self-Assembling Peptides for Biomedical Applications. ACS APPLIED BIO MATERIALS 2024; 7:1723-1734. [PMID: 38346174 DOI: 10.1021/acsabm.3c01148] [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: 03/19/2024]
Abstract
The development of three-dimensional (3D) biomaterials that mimic natural tissues is required for efficiently restoring physiological functions of injured tissues and organs. In the field of soft hydrogels, self-assembled peptides (SAPs) stand out as distinctive biomimetic scaffolds, offering tunable properties. They have garnered significant attention in nanomedicine due to their innate ability to self-assemble, resulting in the creation of fibrous nanostructures that closely mimic the microenvironment of the extracellular matrix (ECM). This unique feature ensures their biocompatibility and bioactivity, making them a compelling area of study over the past few decades. As they are soft hydrogels, approaches are necessary to enhance the stiffness and resilience of the SAP materials. This work shows an enzymatic strategy to selectively increase the stiffness and resiliency of functionalized SAPs using transglutaminase (TGase) type 2, an enzyme capable of triggering the formation of isopeptide bonds. To this aim, we synthesized a set of SAP sequences and characterized their cross-linking via rheological experiments, atomic force microscopy (AFM), thioflavin-T binding assay, and infrared spectroscopy (ATR-FTIR) tests. The results showed an improvement of the storage modulus of cross-linked SAPs at no cost of the maximum stress-at-failure. Further, in in vitro tests, we examined and validated the TGase capability to cross-link SAPs without hampering seeded neural stem cells (hNSCs) viability and differentiation, potentially leaving the door open for safe in situ cross-linking reactions in vivo.
Collapse
Affiliation(s)
- Maria Gessica Ciulla
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Amanda Marchini
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Jacopo Gazzola
- Department of Biotechnology and Biosciences, University of Milan - Bicocca, 20125 Milan, Italy
| | - Mahdi Forouharshad
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Raffaele Pugliese
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| |
Collapse
|
25
|
Yang T, Xue T, Mao J, Chen Y, Tian H, Bartolome A, Xia H, Yao X, Kumar CV, Cheng J, Lin Y. Tailoring Synthetic Polypeptide Design for Directed Fibril Superstructure Formation and Enhanced Hydrogel Properties. J Am Chem Soc 2024; 146:5823-5833. [PMID: 38174701 DOI: 10.1021/jacs.3c10762] [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] [Indexed: 01/05/2024]
Abstract
The biological significance of self-assembled protein filament networks and their unique mechanical properties have sparked interest in the development of synthetic filament networks that mimic these attributes. Building on the recent advancement of autoaccelerated ring-opening polymerization of amino acid N-carboxyanhydrides (NCAs), this study strategically explores a series of random copolymers comprising multiple amino acids, aiming to elucidate the core principles governing gelation pathways of these purpose-designed copolypeptides. Utilizing glutamate (Glu) as the primary component of copolypeptides, two targeted pathways were pursued: first, achieving a fast fibrillation rate with lower interaction potential using serine (Ser) as a comonomer, facilitating the creation of homogeneous fibril networks; and second, creating more rigid networks of fibril clusters by incorporating alanine (Ala) and valine (Val) as comonomers. The selection of amino acids played a pivotal role in steering both the morphology of fibril superstructures and their assembly kinetics, subsequently determining their potential to form sample-spanning networks. Importantly, the viscoelastic properties of the resulting supramolecular hydrogels can be tailored according to the specific copolypeptide composition through modulations in filament densities and lengths. The findings enhance our understanding of directed self-assembly in high molecular weight synthetic copolypeptides, offering valuable insights for the development of synthetic fibrous networks and biomimetic supramolecular materials with custom-designed properties.
Collapse
Affiliation(s)
- Tianjian Yang
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Tianrui Xue
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jianan Mao
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Yingying Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Huidi Tian
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Arlene Bartolome
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Hongwei Xia
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Xudong Yao
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Challa V Kumar
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jianjun Cheng
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yao Lin
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
26
|
Lee ALZ, Balakrishnan N, Ng JY, Liu S, Ong ZY, Wang Y, Gao S, Yang YY. Injectable Hydrogels Prepared Using Novel Synthetic Short Peptides with Defined Structure and Gelatin as Scaffolds to Support Cell and Tissue Growth. Adv Healthc Mater 2024; 13:e2302786. [PMID: 37837308 DOI: 10.1002/adhm.202302786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Indexed: 10/15/2023]
Abstract
Animal-derived basement-membrane matrices such as Geltrex are used to grow cells and tissues. Particularly, these are commonly applied to support tumor growth in animals for cancer research. However, a material derived from an animal source has an undefined composition, and may thus have unavoidable batch-to-batch variation in properties. To overcome these issues, a series of synthetic short peptides to form hydrogels is designed in combination with gelatin to promote cell adhesion and growth. The peptides have sequences of (X1Y1X2Y2)2 , where X1 and X2 are hydrophobic residues, while Y1 and Y2 are hydrophilic residues. The peptides spontaneously fold and self-assemble into a β-sheet secondary structure upon contact with salts, and then aggregate to form hydrophilic networks of hydrogels. Hybrid hydrogels formed by mixing the peptide IEVEIRVK (IVK8) with gelatin are injectable and enzymatically degradable. The hybrid hydrogels at optimal compositions support SW480 and HepG2 tumor spheroid growth in vitro as effectively as Geltrex. More importantly, the peptide/gelatin hydrogels support tumor growth in a SW480 human colorectal adenocarcinoma xenograft mouse model. Altogether, the results illustrate that the synthetic peptide/gelatin hybrid hydrogel is a promising scaffold that can be used to support cell and tissue growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Ashlynn Ling Zhi Lee
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Nithiyaa Balakrishnan
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Jian Yao Ng
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Shaoqiong Liu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Zhan Yuin Ong
- School of Physics and Astronomy, St. James's University of Leeds, Leeds, LS2 9JT, UK
- Leeds Institute of Medical Research, St. James's University of Leeds, Leeds, LS2 9JT, UK
| | - Yanming Wang
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology, and Research (A*STAR), 31 Biopolis Way, Singapore, 138669, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| |
Collapse
|
27
|
Zeng L, Luo G, Yue Z, Tang Y, Wang Z, Chang Y. Experimental Study on Rapid Hemostasis Using Peptide Hydrogels. ACS OMEGA 2024; 9:9247-9255. [PMID: 38434851 PMCID: PMC10905740 DOI: 10.1021/acsomega.3c08310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 03/05/2024]
Abstract
Uncontrolled hemorrhaging resulting from trauma, surgery, and disease-associated or drug-induced blood disorders can cause significant morbidities and mortalities in civilian and military populations. Self-assembling peptide nanofibers are particularly attractive due to their rapid and efficient hemostasis, biocompatibility, and wound-healing properties. In this study, we designed two types of 12-residue peptides by using a strong fishnet-like peptide sequence and a pro-cell adhesion sequence (Arg-Gly-Asp, RGD). The peptides are HN2-X-Ser-Phe-Cys-Phe-Lys-Phe-Glu-X-Arg-Gly-Asp-OH (where X is Pro or Tyr), which dissolve in deionized (DI) water and form stable and transparent functional hydrogels. Transmission electron microscopy and scanning electron microscopy demonstrated that the two peptides self-assemble into nanowebs and nanofibers, forming a fishnet-like and three-dimensional network structure. Circular dichroism and Fourier transform infrared spectroscopy analysis demonstrated that the self-assembled peptides mainly adopt a β-sheet structure with β-turn and α-helix as auxiliary assembly growth. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and SEM analysis showed that the cell survival rates were very good, delivering an obvious promotion of cell proliferation of fibroblasts and hepatocytes. Importantly, in vivo hemostasis delivered that the self-assembled peptide nanowebs and nanofibers had a good hemostatic effect on rat saphenous vein and liver bleeding, achieving 38 s faster hemostasis, which was better than commercial "Instantaneous" hemostatic powder. Accoupling the fast hemostasis and effective promotion of liver defect rapid repair, the peptide self-assembly strategy offers a clinically promising treatment option for life-threatening liver bleeding and serves as a renewed impetus for the development of peptide hydrogels as effective hemostatic agents.
Collapse
Affiliation(s)
- Linru Zeng
- Department
of Orthopedics, Hangzhou Xiaoshan Traditional
Chinese Medical Hospital, Hangzhou 311201, P. R. China
| | - Gan Luo
- Department
of Orthopedics, Hangzhou Xiaoshan Traditional
Chinese Medical Hospital, Hangzhou 311201, P. R. China
| | - Zhenshuang Yue
- Department
of Orthopedics, Hangzhou Xiaoshan Traditional
Chinese Medical Hospital, Hangzhou 311201, P. R. China
| | - Yanghua Tang
- Department
of Orthopedics, Hangzhou Xiaoshan Traditional
Chinese Medical Hospital, Hangzhou 311201, P. R. China
| | - Zhetian Wang
- The
Third Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, P. R. China
| | - Yitie Chang
- The
Third Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, P. R. China
| |
Collapse
|
28
|
Abstract
Peptide epitopes mediate as many as 40% of protein-protein interactions and fulfill signaling, inhibition, and activation roles within the cell. Beyond protein recognition, some peptides can self- or coassemble into stable hydrogels, making them a readily available source of biomaterials. While these 3D assemblies are routinely characterized at the fiber level, there are missing atomistic details about the assembly scaffold. Such atomistic detail can be useful in the rational design of more stable scaffold structures and with improved accessibility to functional motifs. Computational approaches can in principle reduce the experimental cost of such an endeavor by predicting the assembly scaffold and identifying novel sequences that adopt said structure. Yet, inaccuracies in physical models and inefficient sampling have limited atomistic studies to short (two or three amino acid) peptides. Given recent developments in machine learning and advances in sampling strategies, we revisit the suitability of physical models for this task. We use the MELD (Modeling Employing Limited Data) approach to drive self-assembly in combination with generic data in cases where conventional MD is unsuccessful. Finally, despite recent developments in machine learning algorithms for protein structure and sequence predictions, we find the algorithms are not yet suited for studying the assembly of short peptides.
Collapse
Affiliation(s)
- Stephen J Jones
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| | - Alberto Perez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
29
|
Li T, Kambanis J, Sorenson TL, Sunde M, Shen Y. From Fundamental Amyloid Protein Self-Assembly to Development of Bioplastics. Biomacromolecules 2024; 25:5-23. [PMID: 38147506 PMCID: PMC10777412 DOI: 10.1021/acs.biomac.3c01129] [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: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/28/2023]
Abstract
Proteins can self-assemble into a range of nanostructures as a result of molecular interactions. Amyloid nanofibrils, as one of them, were first discovered with regard to the relevance of neurodegenerative diseases but now have been exploited as building blocks to generate multiscale materials with designed functions for versatile applications. This review interconnects the mechanism of amyloid fibrillation, the current approaches to synthesizing amyloid protein-based materials, and the application in bioplastic development. We focus on the fundamental structures of self-assembled amyloid fibrils and how external factors can affect protein aggregation to optimize the process. Protein self-assembly is essentially the autonomous congregation of smaller protein units into larger, organized structures. Since the properties of the self-assembly can be manipulated by changing intrinsic factors and external conditions, protein self-assembly serves as an excellent building block for bioplastic development. Building on these principles, general processing methods and pathways from raw protein sources to mature state materials are proposed, providing a guide for the development of large-scale production. Additionally, this review discusses the diverse properties of protein-based amyloid nanofibrils and how they can be utilized as bioplastics. The economic feasibility of the protein bioplastics is also compared to conventional plastics in large-scale production scenarios, supporting their potential as sustainable bioplastics for future applications.
Collapse
Affiliation(s)
- Tianchen Li
- School
of Chemical and Biomolecular Engineering and Sydney Nano, The University of Sydney, PNR Building, Darlington NSW 2008, Australia
| | - Jordan Kambanis
- School
of Chemical and Biomolecular Engineering and Sydney Nano, The University of Sydney, PNR Building, Darlington NSW 2008, Australia
| | - Timothy L. Sorenson
- School
of Chemical and Biomolecular Engineering and Sydney Nano, The University of Sydney, PNR Building, Darlington NSW 2008, Australia
| | - Margaret Sunde
- School
of Medical Sciences and Sydney Nano, The
University of Sydney, Sydney NSW 2006, Australia
| | - Yi Shen
- School
of Chemical and Biomolecular Engineering and Sydney Nano, The University of Sydney, PNR Building, Darlington NSW 2008, Australia
| |
Collapse
|
30
|
Zhang Q, Wang W, Zhou S, Zhang R, Bischofberger I. Flow-induced periodic chiral structures in an achiral nematic liquid crystal. Nat Commun 2024; 15:7. [PMID: 38191525 PMCID: PMC10774319 DOI: 10.1038/s41467-023-43978-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 11/26/2023] [Indexed: 01/10/2024] Open
Abstract
Supramolecular chirality typically originates from either chiral molecular building blocks or external chiral stimuli. Generating chirality in achiral systems in the absence of a chiral input, however, is non-trivial and necessitates spontaneous mirror symmetry breaking. Achiral nematic lyotropic chromonic liquid crystals have been reported to break mirror symmetry under strong surface or geometric constraints. Here we describe a previously unrecognised mechanism for creating chiral structures by subjecting the material to a pressure-driven flow in a microfluidic cell. The chirality arises from a periodic double-twist configuration of the liquid crystal and manifests as a striking stripe pattern. We show that the mirror symmetry breaking is triggered at regions of flow-induced biaxial-splay configurations of the director field, which are unstable to small perturbations and evolve into lower energy structures. The simplicity of this unique pathway to mirror symmetry breaking can shed light on the requirements for forming macroscopic chiral structures.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Weiqiang Wang
- Department of Physics, Hong Kong University of Science and Technology, Hong Kong, China
| | - Shuang Zhou
- Department of Physics, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Rui Zhang
- Department of Physics, Hong Kong University of Science and Technology, Hong Kong, China.
| | - Irmgard Bischofberger
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
31
|
Pandey G, Phatale V, Khairnar P, Kolipaka T, Shah S, Famta P, Jain N, Srinivasarao DA, Rajinikanth PS, Raghuvanshi RS, Srivastava S. Supramolecular self-assembled peptide-engineered nanofibers: A propitious proposition for cancer therapy. Int J Biol Macromol 2024; 256:128452. [PMID: 38042321 DOI: 10.1016/j.ijbiomac.2023.128452] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Cancer is a devastating disease that causes a substantial number of deaths worldwide. Current therapeutic interventions for cancer include chemotherapy, radiation therapy, or surgery. These conventional therapeutic approaches are associated with disadvantages such as multidrug resistance, destruction of healthy tissues, and tissue toxicity. Therefore, there is a paradigm shift in cancer management wherein nanomedicine-based novel therapeutic interventions are being explored to overcome the aforementioned disadvantages. Supramolecular self-assembled peptide nanofibers are emerging drug delivery vehicles that have gained much attention in cancer management owing to their biocompatibility, biodegradability, biomimetic property, stimuli-responsiveness, transformability, and inherent therapeutic property. Supramolecules form well-organized structures via non-covalent linkages, the intricate molecular arrangement helps to improve tissue permeation, pharmacokinetic profile and chemical stability of therapeutic agents while enabling targeted delivery and allowing efficient tumor imaging. In this review, we present fundamental aspects of peptide-based self-assembled nanofiber fabrication their applications in monotherapy/combinatorial chemo- and/or immuno-therapy to overcome multi-drug resistance. The role of self-assembled structures in targeted/stimuli-responsive (pH, enzyme and photo-responsive) drug delivery has been discussed along with the case studies. Further, recent advancements in peptide nanofibers in cancer diagnosis, imaging, gene therapy, and immune therapy along with regulatory obstacles towards clinical translation have been deliberated.
Collapse
Affiliation(s)
- Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Naitik Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Rajeev Singh Raghuvanshi
- Central Drugs Standard Control Organization (CDSCO), Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
32
|
Wu J, Zhou L, Peng H, Wang Z, Wang Z, Keasling JD, Liu S, Zhou G, Ding S, Wang Q, Wang X, Chen X, Lang Y, Xia M, Guan X, Dong M, Zhou J, Chen J. A General and Convenient Peptide Self-Assembling Mechanism for Developing Supramolecular Versatile Nanomaterials Based on The Biosynthetic Hybrid Amyloid-Resilin Protein. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304364. [PMID: 37885340 DOI: 10.1002/adma.202304364] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Self-assembling peptides are valuable building blocks to fabricate supramolecular biomaterials, which have broad applications from biomedicine to biotechnology. However, limited choices to induce different globular proteins into hydrogels hinder these designs. Here, an easy-to-implement and tunable self-assembling strategy, which employs Ure2 amyloidogenic peptide, are described to induce any target proteins to assemble into supramolecular hydrogels alone or in combination with notable compositional control. Furthermore, the collective effect of nanoscale interactions among amyloid nanofibrils and partially disordered elastomeric polypeptides are investigated. This led to many useful macroscopic material properties simultaneously emerging from one pure protein material, i.e. strong adhesion to any substrates under wet conditions, rapidly self--assembling into robust and porous hydrogels, adaptation to remodeling processes, strongly promoting cell adhesion, proliferation and differentiation. Moreover, he demonstrated this supramolecular material's robust performance in vitro and vivo for tissue engineering, cosmetic and hemostasis applications and exhibited superior performance compared to corresponding commercial counterparts. To the best of his knowledge, few pure protein-based materials could meet such seemingly mutually exclusive properties simultaneously. Such versatility renders this novel supramolecular nanomaterial as next-generation functional protein-based materials, and he demonstrated the sequence level modulation of structural order and disorder as an untapped principle to design new proteins.
Collapse
Affiliation(s)
- Junjun Wu
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China
| | - Lin Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Hu Peng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Zhaojun Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Zhaoshi Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Jay D Keasling
- Departments of Chemical & Biomolecular Engineering and of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- Center for Biosustainability, Technical University of Denmark, Lyngby, 2800, Denmark
| | - Shike Liu
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Guanghong Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Shijie Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Qiong Wang
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China
| | - Xuejian Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xinxiu Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yifei Lang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Mo Xia
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xin Guan
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Mingsheng Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Jingwen Zhou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
33
|
Ishikawa S, Sakai T. One-Pot Approach to Synthesize Tough and Cell Adhesive Double-Network Hydrogels Consisting of Fully Synthetic Materials of Self-Assembling Peptide and Poly(ethylene glycol). ACS APPLIED BIO MATERIALS 2023; 6:5282-5289. [PMID: 37862142 DOI: 10.1021/acsabm.3c00562] [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: 10/22/2023]
Abstract
Hydrogels with a double network (DN) structure are compelling biomaterials, holding potential for use as artificial extracellular matrices. Generally, the DN approach imparts hydrogels with high mechanical strength and cell-adhesive properties. However, achieving this often demands a complex multistep process involving potentially hazardous free-radical polymerization, which can result in toxicity. This limits their broad biological applications. In this work, we introduce a straightforward yet biocompatible method to fabricate tough and cell-adhesive DN hydrogels using entirely synthetic materials: the self-assembling peptide (RADA16) and poly(ethylene glycol) (PEG). An in situ mixing of these components leads to the sequential formation of DN hydrogels─first through the self-assembly of the RADA16 peptide and then via chemical cross-linking between PEG molecules. Hydrogels produced this way exhibited up to a 10-fold increase in fracture energy, and cells seeded on their surfaces showcased good attachment. Our design underscores the efficacy of the DN approach and the promising applications of peptides in tissue engineering.
Collapse
Affiliation(s)
- Shohei Ishikawa
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Takamasa Sakai
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
34
|
Wu Y, Li H, Liu T, Xu M. Versatile Protein and Its Subunit Biomolecules for Advanced Rechargeable Batteries. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305063. [PMID: 37474115 DOI: 10.1002/adma.202305063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Rechargeable batteries are of great significance for alleviating the growing energy crisis by providing efficient and sustainable energy storage solutions. However, the multiple issues associated with the diverse components in a battery system as well as the interphase problems greatly hinder their applications. Proteins and their subunits, peptides, and amino acids, are versatile biomolecules. Functional groups in different amino acids endow these biomolecules with unique properties including self-assembly, ion-conducting, antioxidation, great affinity to exterior species, etc. Besides, protein and its subunit materials can not only work in solid forms but also in liquid forms when dissolved in solutions, making them more versatile to realize materials engineering via diverse approaches. In this review, it is aimed to offer a comprehensive understanding of the properties of proteins and their subunits, and research progress of using these versatile biomolecules to address the engineering issues of various rechargeable batteries, including alkali-ion batteries, lithium-sulfur batteries, metal-air batteries, and flow batteries. The state-of-the-art advances in electrode, electrolyte, separator, binder, catalyst, interphase modification, as well as recycling of rechargeable batteries are involved, and the impacts of biomolecules on electrochemical properties are particularly emphasized. Finally, perspectives on this interesting field are also provided.
Collapse
Affiliation(s)
- Yulun Wu
- School of Metallurgy and Environment, Central South University, Changsha, 410083, P.R. China
| | - Huangxu Li
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, P.R. China
| | - Tiancheng Liu
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, P.R. China
| | - Ming Xu
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
35
|
Zeng G, Zheng Y, Xiang Y, Liu R, Yang X, Lin Z. A novel protein purification scheme based on salt inducible self-assembling peptides. Microb Cell Fact 2023; 22:224. [PMID: 37899435 PMCID: PMC10614350 DOI: 10.1186/s12934-023-02229-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND Protein purification remains a critical need for biosciences and biotechnology. It frequently requires multiple rounds of chromatographic steps that are expensive and time-consuming. Our lab previously reported a cleavable self-aggregating tag (cSAT) scheme for streamlined protein expression and purification. The tag consists of a self-assembling peptide (SAP) and a controllable self-cleaving intein. The SAP drives the target protein into an active aggregate, then by intein-mediated cleavage, the target protein is released. Here we report a novel cSAT scheme in which the self-assembling peptide is replaced with a salt inducible self-assembling peptide. This allows a target protein to be expressed first in the soluble form, and the addition of salt then drives the target protein into the aggregated form, followed by cleavage and release. RESULTS In this study, we used MpA (MKQLEDKIEELLSKAAMKQLEDKIEELLSK) as a second class of self-assembling peptide in the cSAT scheme. This scheme utilizes low salt concentration to keep the fusion protein soluble, while eliminating insoluble cellular matters by centrifugation. Salt then triggers MpA-mediated self-aggregation of the fusion, removing soluble background host cell proteins. Finally, intein-mediated cleavage releases the target protein into solution. As a proof-of-concept, we successfully purified four proteins and peptides (human growth hormone, 22.1 kDa; LCB3, 7.7 kDa; SpyCatcherΔN-ELP-SpyCatcherΔN, 26.2 kDa; and xylanase, 45.3 kDa) with yields ranging from 12 to 87 mg/L. This was comparable to the classical His-tag method both in yield and purity (72-97%), but without the His-tag. By using a further two-step column purification process that included ion-exchange chromatography and size-exclusion chromatography, the purity was increased to over 99%. CONCLUSION Our results demonstrate that a salt-inducible self-assembling peptide can serve as a controllable aggregating tag, which might be advantageous in applications where soluble expression of the target protein is preferred. This work also demonstrates the potential and advantages of utilizing salt inducible self-assembling peptides for protein separation.
Collapse
Affiliation(s)
- Guang Zeng
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China
| | - Yinzhen Zheng
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China
| | - Ya Xiang
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China
| | - Run Liu
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China
| | - Xiaofeng Yang
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China.
| | - Zhanglin Lin
- School of Biology and Biological Engineering, South China University of Technology, 382 East Outer Loop Road, University Park, Guangzhou, 510006, China.
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
36
|
Ciulla MG, Marchini A, Gazzola J, Sambrotta M, Gelain F. Low-power microwaves: a cell-compatible physical treatment to enhance the mechanical properties of self-assembling peptides. NANOSCALE 2023; 15:15840-15854. [PMID: 37747054 DOI: 10.1039/d3nr02738d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Biomaterials designed for tissue engineering applications should, among other requirements, mimic the native extracellular matrix (ECM) of the tissues to be regenerated, both in terms of biomimetic and mechanical properties. Ideally, the scaffold stiffness and stress resistance should be tuned for each specific implantation therapy. Self-assembling peptides (SAPs) are promising synthetic bionanomaterials prone to easy multi-functionalization, bestowing biomimetic properties. However, they usually yield soft and fragile hydrogels unsuited for the regeneration of medium-to-hard tissues. For this purpose, chemical cross-linking of SAPs is an option, but it often requires a moderately toxic and expensive chemical compound and/or the presence of specific residues/reactive sites, posing issues for its feasibility and translational potential. In this work, we introduced, characterized by rheology, atomic force microscopy (AFM), Thioflavin-T assay (ThT), and Fourier transform infrared (FT-IR) tests, and optimized (by tuning the power, temperature and treatment time) a novel fast, green and affordable methodology using mild microwave (MW) irradiation to increase the mechanical properties of diverse classes of SAPs. Low-power MWs increase stiffness, resilience, and β-structuration, while high-power MW treatments partially denature the tested SAPs. Our pure-physical methodology does not alter the SAP biomimetic properties (verified via in vitro tests of viability and differentiation of human neural stem cells), is compatible with already seeded cells, and is also synergic with genipin-based cross-linking of SAPs; therefore, it may become the next standard for SAP preparation in tissue engineering applications at hand of all research labs and in clinics.
Collapse
Affiliation(s)
- Maria Gessica Ciulla
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
| | - Amanda Marchini
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Jacopo Gazzola
- Department of Biotechnology and Bioscience, University of Milan - Bicocca, 20125 Milan, Italy
| | - Manuel Sambrotta
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| |
Collapse
|
37
|
Yao X, Hu Y, Lin M, Peng K, Wang P, Gao Y, Gao X, Guo T, Zhang X, Zhou H. Self-assembling peptide RADA16: a promising scaffold for tissue engineering and regenerative medicine. Nanomedicine (Lond) 2023. [PMID: 37750388 DOI: 10.2217/nnm-2023-0161] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
RADA16 is a peptide-based biomaterial whose acidic aqueous solution spontaneously forms an extracellular matrix-like 3D structure within seconds upon contact with physiological pH body fluids. Meanwhile, its good biocompatibility, low immunogenicity, nontoxic degradation products and ease of modification make it an ideal scaffold for tissue engineering. RADA16 is a good delivery vehicle for cells, drugs and factors. Its shear thinning and thixotropic properties allow it to fill tissue voids by injection and not to swell. However, the weaker mechanical properties and poor hydrophilicity are troubling limitations of RADA16. To compensate for this limitation, various functional groups and polymers have been designed to modify RADA16, thus contributing to its scope and progress in the field of tissue engineering.
Collapse
Affiliation(s)
- Xin Yao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Yicun Hu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Maoqiang Lin
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Kaichen Peng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Peng Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Yanbing Gao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| | - Taowen Guo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| | - Haiyu Zhou
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| |
Collapse
|
38
|
Nie C, Zou Y, Liao S, Gao Q, Li Q. Peptides as carriers of active ingredients: A review. Curr Res Food Sci 2023; 7:100592. [PMID: 37766891 PMCID: PMC10519830 DOI: 10.1016/j.crfs.2023.100592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/20/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Bioactive compounds are highly valuable in the fields of food and medicine, but their application is limited due to easy deterioration after oral or skin administration. In recent years, the use of peptides as delivery systems for bioactive compounds has been intensively researched because of their special physicochemical characteristics. Peptides can be assembled using various preparation methods and can form several composite materials such as hydrogels, micelles, emulsions and particles. The composite material properties are determined by peptides, bioactive compounds and the construction methods employed. Herein, this paper provides a comprehensive review of the peptides used for active ingredients delivery, fabrication methods for creating delivery systems, structures, targeting characteristics, functional activities and mechanism of delivery systems, as well as their absorption and metabolism, which provided theoretical basis and reference for further research and development of functional composites.
Collapse
Affiliation(s)
- Congyi Nie
- Guangdong Academy of Agricultural Sciences, Sericultural & Agri-Food Research Institute/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Yuxiao Zou
- Guangdong Academy of Agricultural Sciences, Sericultural & Agri-Food Research Institute/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Sentai Liao
- Guangdong Academy of Agricultural Sciences, Sericultural & Agri-Food Research Institute/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Qunyu Gao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Qian Li
- Guangdong Academy of Agricultural Sciences, Sericultural & Agri-Food Research Institute/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| |
Collapse
|
39
|
Xu YE, Ao DS, Sun X, Chen W, Luo X, Zhao C, Wang SY, Song H. A Novel Airway-Organoid Model Based on a Nano-Self-Assembling Peptide: Construction and Application in Adenovirus Infection Studies. Int J Nanomedicine 2023; 18:5225-5241. [PMID: 37727651 PMCID: PMC10505585 DOI: 10.2147/ijn.s413743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Purpose Hydrogels containing the nano-self-assembling peptide RADA16-I (Nanogels) were utilized as scaffolds to establish airway organoids and an adenovirus-infected model. The results support in vitro adenovirus studies, including isolation and culture, pathogenesis research, and antiviral drug screening. Methods HSAEC1-KT, HuLEC-5a and HELF cells were cocultured in RADA16-I hydrogel scaffolds to construct an airway organoid model. Adenovirus was used to infect this model for adenovirus-related studies. The morphological characteristics and the proliferation and activity of airway organoids before and after adenovirus infection were evaluated. The expression of the airway organoid marker proteins CC10, KRT8, AQP5, SPC, VIM and CD31 was detected. TEM and qPCR were used to detect adenovirus proliferation in airway organoids. Results HSAEC1-KT, HuLEC-5a and HELF cells cocultured at 10:7:2 self-assembled into airway organoids and maintained long-term proliferation in a RADA16-I hydrogel 3D culture system. The organoids stably expressed the lumen-forming protein KRT8 and the terminal airway markers AQP5 and SPC. Adenoviruses maintained long-term proliferation in this model. Conclusion An airway-organoid model of adenovirus infection was constructed in vitro from three human lung-derived cell lines on RADA16-I hydrogels. The model has potential as a novel research tool for adenovirus isolation and culture, pathogenesis research, and antiviral drug screening.
Collapse
Affiliation(s)
- Yun-E Xu
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Di-Shu Ao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Xin Sun
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Wei Chen
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, 563000, People’s Republic of China
| | - Xue Luo
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Can Zhao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Sheng-Yu Wang
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Hong Song
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| |
Collapse
|
40
|
Yang Z, Chen L, Liu J, Zhuang H, Lin W, Li C, Zhao X. Short Peptide Nanofiber Biomaterials Ameliorate Local Hemostatic Capacity of Surgical Materials and Intraoperative Hemostatic Applications in Clinics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301849. [PMID: 36942893 DOI: 10.1002/adma.202301849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/12/2023] [Indexed: 06/18/2023]
Abstract
Short designer self-assembling peptide (dSAP) biomaterials are a new addition to the hemostat group. It may provide a diverse and robust toolbox for surgeons to integrate wound microenvironment with much safer and stronger hemostatic capacity than conventional materials and hemostatic agents. Especially in noncompressible torso hemorrhage (NCTH), diffuse mucosal surface bleeding, and internal medical bleeding (IMB), with respect to the optimal hemostatic formulation, dSAP biomaterials are the ingenious nanofiber alternatives to make bioactive neural scaffold, nasal packing, large mucosal surface coverage in gastrointestinal surgery (esophagus, gastric lesion, duodenum, and lower digestive tract), epicardiac cell-delivery carrier, transparent matrix barrier, and so on. Herein, in multiple surgical specialties, dSAP-biomaterial-based nano-hemostats achieve safe, effective, and immediate hemostasis, facile wound healing, and potentially reduce the risks in delayed bleeding, rebleeding, post-operative bleeding, or related complications. The biosafety in vivo, bleeding indications, tissue-sealing quality, surgical feasibility, and local usability are addressed comprehensively and sequentially and pursued to develop useful surgical techniques with better hemostatic performance. Here, the state of the art and all-round advancements of nano-hemostatic approaches in surgery are provided. Relevant critical insights will inspire exciting investigations on peptide nanotechnology, next-generation biomaterials, and better promising prospects in clinics.
Collapse
Affiliation(s)
- Zehong Yang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lihong Chen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ji Liu
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hua Zhuang
- Department of Ultrasonography, West China Hospital of Sichuan University, No. 37 Guoxue Road, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Wei Lin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Women and Children Diseases of the Ministry of Education, Sichuan University, No. 17 People's South Road, Chengdu, Sichuan, 610041, China
| | - Changlong Li
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
41
|
Uchida N, Muraoka T. Self-assembling materials functionalizing bio-interfaces of phospholipid membranes and extracellular matrices. Chem Commun (Camb) 2023; 59:9687-9697. [PMID: 37440181 DOI: 10.1039/d3cc01875j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
This Feature Article focuses on recent studies on the development of self-assembling materials that mimic and control dynamic bio-interfaces. Extracellular matrix (ECM) is a fundamental tissue at the cellular interface constructed by networks of fibrous proteins, which regulates a variety of cellular activities. Reconstruction of ECM has been demonstrated by self-assembling peptides. By combining the dynamic properties of the self-assembling peptides conjugated with full-length proteins, peptide-based supramolecular materials enable neuronal migration and regeneration of injured neural tissue. The phospholipid bilayer is the main component of the cell membrane. The morphology and deformation of the phospholipid bilayer relate directly to dynamic interfacial functions. Stabilization of the phospholipid nanosheet structure has been demonstrated by self-assembling peptides, and the stabilized bicelle is functional for extended blood circulation. By using a photo-responsive synthetic surfactant showing a mechanical opening/closing motion, endocytosis-like outside-in membrane deformation is triggered. The outside-in deformation allows for efficient encapsulation of micrometer-size substances such as phage viruses into the liposomes, and the encapsulated viruses can be delivered to multiple organs in a living body via blood administration. These supramolecular approaches to mimicking and controlling bio-interfaces present powerful ways to develop unprecedented regenerative medicines and drug delivery systems.
Collapse
Affiliation(s)
- Noriyuki Uchida
- Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Takahiro Muraoka
- Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-Shi, Tokyo 183-8538, Japan.
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 705-1 Shimoimaizumi, Ebina, Kanagawa 243-0435, Japan
| |
Collapse
|
42
|
Yang R, Chen J, Wang D, Xu Y, Ou G. Self-Assembling Peptide RADA16 Nanofiber Scaffold Hydrogel-Wrapped Concentrated Growth Factors in Osteogenesis of MC3T3. J Funct Biomater 2023; 14:jfb14050260. [PMID: 37233370 DOI: 10.3390/jfb14050260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/27/2023] Open
Abstract
Concentrated growth factors (CGFs) are widely used in surgery with bone grafting, but the release of growth factors from CGFs is rapid. RADA16, a self-assembling peptide, can form a scaffold that is similar to the extracellular matrix. Based on the properties of RADA16 and CGF, we hypothesized that the RADA16 nanofiber scaffold hydrogel could enhance the function of CGFs and that the RADA16 nanofiber scaffold hydrogel-wrapped CGFs (RADA16-CGFs) would perform a good osteoinductive function. This study aimed to investigate the osteoinductive function of RADA16-CGFs. Scanning electron microscopy, rheometry, and ELISA were performed, and MC3T3-E1 cells were used to test cell adhesion, cytotoxicity, and mineralization after administration with RADA16-CGFs. We found that RADA16 endowed with the sustained release of growth factors from CGFs, which can help maximize the function of CGFs in osteoinduction. The application of the atoxic RADA16 nanofiber scaffold hydrogel with CGFs can be a new therapeutic strategy for the treatment of alveolar bone loss and other problems that require bone regeneration.
Collapse
Affiliation(s)
- Renjie Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Eastern Clinic, West China Hospital of Stomatology, Sichuan University, Chengdu 610051, China
| | - Jiali Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dingjie Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yichen Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Guomin Ou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
43
|
Dzierżyńska M, Sawicka J, Deptuła M, Sosnowski P, Sass P, Peplińska B, Pietralik-Molińska Z, Fularczyk M, Kasprzykowski F, Zieliński J, Kozak M, Sachadyn P, Pikuła M, Rodziewicz-Motowidło S. Release systems based on self-assembling RADA16-I hydrogels with a signal sequence which improves wound healing processes. Sci Rep 2023; 13:6273. [PMID: 37072464 PMCID: PMC10113214 DOI: 10.1038/s41598-023-33464-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
Self-assembling peptides can be used for the regeneration of severely damaged skin. They can act as scaffolds for skin cells and as a reservoir of active compounds, to accelerate scarless wound healing. To overcome repeated administration of peptides which accelerate healing, we report development of three new peptide biomaterials based on the RADA16-I hydrogel functionalized with a sequence (AAPV) cleaved by human neutrophil elastase and short biologically active peptide motifs, namely GHK, KGHK and RDKVYR. The peptide hybrids were investigated for their structural aspects using circular dichroism, thioflavin T assay, transmission electron microscopy, and atomic force microscopy, as well as their rheological properties and stability in different fluids such as water or plasma, and their susceptibility to digestion by enzymes present in the wound environment. In addition, the morphology of the RADA-peptide hydrogels was examined with a unique technique called scanning electron cryomicroscopy. These experiments enabled us to verify if the designed peptides increased the bioactivity of the gel without disturbing its gelling processes. We demonstrate that the physicochemical properties of the designed hybrids were similar to those of the original RADA16-I. The materials behaved as expected, leaving the active motif free when treated with elastase. XTT and LDH tests on fibroblasts and keratinocytes were performed to assess the cytotoxicity of the RADA16-I hybrids, while the viability of cells treated with RADA16-I hybrids was evaluated in a model of human dermal fibroblasts. The hybrid peptides revealed no cytotoxicity; the cells grew and proliferated better than after treatment with RADA16-I alone. Improved wound healing following topical delivery of RADA-GHK and RADA-KGHK was demonstrated using a model of dorsal skin injury in mice and histological analyses. The presented results indicate further research is warranted into the engineered peptides as scaffolds for wound healing and tissue engineering.
Collapse
Affiliation(s)
- Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł Sosnowski
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Piotr Sass
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | | | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | | | - Jacek Zieliński
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
44
|
Ohno Y, Nakajima C, Ajioka I, Muraoka T, Yaguchi A, Fujioka T, Akimoto S, Matsuo M, Lotfy A, Nakamura S, Herranz-Pérez V, García-Verdugo JM, Matsukawa N, Kaneko N, Sawamoto K. Amphiphilic peptide-tagged N-cadherin forms radial glial-like fibers that enhance neuronal migration in injured brain and promote sensorimotor recovery. Biomaterials 2023; 294:122003. [PMID: 36736095 DOI: 10.1016/j.biomaterials.2023.122003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/05/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
The mammalian brain has very limited ability to regenerate lost neurons and recover function after injury. Promoting the migration of young neurons (neuroblasts) derived from endogenous neural stem cells using biomaterials is a new and promising approach to aid recovery of the brain after injury. However, the delivery of sufficient neuroblasts to distant injured sites is a major challenge because of the limited number of scaffold cells that are available to guide neuroblast migration. To address this issue, we have developed an amphiphilic peptide [(RADA)3-(RADG)] (mRADA)-tagged N-cadherin extracellular domain (Ncad-mRADA), which can remain in mRADA hydrogels and be injected into deep brain tissue to facilitate neuroblast migration. Migrating neuroblasts directly contacted the fiber-like Ncad-mRADA hydrogel and efficiently migrated toward an injured site in the striatum, a deep brain area. Furthermore, application of Ncad-mRADA to neonatal cortical brain injury efficiently promoted neuronal regeneration and functional recovery. These results demonstrate that self-assembling Ncad-mRADA peptides mimic both the function and structure of endogenous scaffold cells and provide a novel strategy for regenerative therapy.
Collapse
Affiliation(s)
- Yuya Ohno
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan; Department of Neurology and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Chikako Nakajima
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Itsuki Ajioka
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kanagawa, 243-0435, Japan
| | - Takahiro Muraoka
- Kanagawa Institute of Industrial Science and Technology (KISTEC), Kanagawa, 243-0435, Japan; Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo, 184-8588, Japan
| | - Atsuya Yaguchi
- Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo, 184-8588, Japan
| | - Teppei Fujioka
- Department of Neurology and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Saori Akimoto
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kanagawa, 243-0435, Japan
| | - Misaki Matsuo
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Sayuri Nakamura
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED, Valencia, 46980, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED, Valencia, 46980, Spain
| | - Noriyuki Matsukawa
- Department of Neurology and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan; Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan; Division of Neural Development and Regeneration, National Institute of Physiological Sciences, Okazaki, Aichi, 444-8585, Japan.
| |
Collapse
|
45
|
Sun W, Gregory DA, Zhao X. Designed peptide amphiphiles as scaffolds for tissue engineering. Adv Colloid Interface Sci 2023; 314:102866. [PMID: 36898186 DOI: 10.1016/j.cis.2023.102866] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David Alexander Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; Department of Material Science and Engineering, University of Sheffield, Sheffield S3 7HQ, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
46
|
Advances in Self-Assembled Peptides as Drug Carriers. Pharmaceutics 2023; 15:pharmaceutics15020482. [PMID: 36839803 PMCID: PMC9964150 DOI: 10.3390/pharmaceutics15020482] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
In recent years, self-assembled peptide nanotechnology has attracted a great deal of attention for its ability to form various regular and ordered structures with diverse and practical functions. Self-assembled peptides can exist in different environments and are a kind of medical bio-regenerative material with unique structures. These materials have good biocompatibility and controllability and can form nanoparticles, nanofibers and hydrogels to perform specific morphological functions, which are widely used in biomedical and material science fields. In this paper, the properties of self-assembled peptides, their influencing factors and the nanostructures that they form are reviewed, and the applications of self-assembled peptides as drug carriers are highlighted. Finally, the prospects and challenges for developing self-assembled peptide nanomaterials are briefly discussed.
Collapse
|
47
|
Natarajan A, Rangan K, Vadrevu R. Self-assembly of a peptide sequence, EKKE, composed of exclusively charged amino acids: Role of charge in morphology and lead binding. J Pept Sci 2023; 29:e3451. [PMID: 36098076 DOI: 10.1002/psc.3451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/14/2022] [Accepted: 09/08/2022] [Indexed: 01/12/2023]
Abstract
The self-assembly of peptides is influenced by their amino acid sequence and other factors including pH, charge, temperature, and solvent. Herein, we explore whether a four-residue sequence, EKKE, consisting of exclusively charged amino acids shows the propensity to form self-assembled ordered nanostructures and whether the overall charge plays any role in morphological and functional properties. From a combination of experimental data provided by Thioflavin T fluorescence, Congo red absorbance, circular dichroism spectroscopy, dynamic light scattering, field emission-scanning electron microscopy, atomic force microscopy, and confocal microscopy, it is clear that the all-polar peptide and charged EKKE sequence shows a pH-dependent tendency to form amyloid-like structures, and the self-assembled entities under acidic, basic and neutral conditions exhibit morphological variation. Additionally, the ability of the self-assembled amyloid nanostructures to bind to the toxic metal, lead (Pb2+ ), was demonstrated from the analysis of the ultraviolet absorbance and X-ray photoelectron spectroscopy data. The modulation at the sequence level for the amyloid-forming EKKE scaffold can further extend its potential role not only in the remediation of other toxic metals but also towards biomedical applications.
Collapse
Affiliation(s)
- Aishwarya Natarajan
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Krishnan Rangan
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Ramakrishna Vadrevu
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| |
Collapse
|
48
|
Cruz-Chamorro I, Santos-Sánchez G, Álvarez-López AI, Pedroche J, Lardone PJ, Arnoldi A, Lammi C, Carrillo-Vico A. Pleiotropic biological effects of Lupinus spp. protein hydrolysates. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
49
|
Thursch LJ, Lima TA, O'Neill N, Ferreira FF, Schweitzer-Stenner R, Alvarez NJ. Influence of central sidechain on self-assembly of glycine-x-glycine peptides. SOFT MATTER 2023; 19:394-409. [PMID: 36454226 DOI: 10.1039/d2sm01082h] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Low molecular weight gelators (LMWGs) are the subject of intense research for a range of biomedical and engineering applications. Peptides are a special class of LMWG, which offer infinite sequence possibilities and, therefore, engineered properties. This work examines the propensity of the GxG peptide family, where x denotes a guest residue, to self-assemble into fibril networks via changes in pH and ethanol concentration. These triggers for gelation are motivated by recent work on GHG and GAG, which unexpectedly self-assemble into centimeter long fibril networks with unique rheological properties. The propensity of GxG peptides to self-assemble, and the physical and chemical properties of the self-assembled structures are characterized by microscopy, spectroscopy, rheology, and X-ray diffraction. Interestingly, we show that the number, length, size, and morphology of the crystalline self-assembled aggregates depend significantly on the x-residue chemistry and the solution conditions, i.e. pH, temperature, peptide concentration, etc. The different x-residues allow us to probe the importance of different peptide interactions, e.g. π-π stacking, hydrogen bonding, and hydrophobicity, on the formation of fibrils. We conclude that fibril formation requires π-π stacking interactions in pure water, while hydrogen bonding can form fibrils in the presence of ethanol-water solutions. These results validate and support theoretical arguments on the propensity for self-assembly and leads to a better understanding of the relationship between peptide chemistry and fibril self-assembly. Overall, GxG peptides constitute a unique family of peptides, whose characterization will aid in advancing our understanding of self-assembly driving forces for fibril formation in peptide systems.
Collapse
Affiliation(s)
- Lavenia J Thursch
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | - Thamires A Lima
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | - Nichole O'Neill
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA.
| | - Fabio F Ferreira
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo, Brazil
| | | | - Nicolas J Alvarez
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Li XF, Lu P, Jia HR, Li G, Zhu B, Wang X, Wu FG. Emerging materials for hemostasis. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|