1
|
Hao R, Niu X, Jiang X, Liu K, Ma X, Chen C. Transglutaminase-triggered dual gradients of mechanical and biochemical cues self-assembling peptide hydrogel for guiding MC3T3-E1 cell behaviors. Int J Biol Macromol 2024; 285:138281. [PMID: 39631574 DOI: 10.1016/j.ijbiomac.2024.138281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/18/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
The mechanical properties and bioactive motif densities of extracellular matrix materials play crucial roles in regulating cell behaviors, such as cell adhesion, migration, proliferation, and differentiation. However, current studies on cellular responses to ECM predominantly concentrated on polymer hydrogels featuring a single factor, such as the mechanical strength, the types of bioactive motifs, and the morphology of the polymers. This limited focus may overlook the complex interplay of multiple factors. Here, we developed dual gradient peptide Q3GT-I3K hydrogels with tunable mechanical strength (0.3-4.0 kPa) and different density of bioactive motif (0.45-3.67 mM) by enzymatic crosslinking. These hydrogels can mimic the viscoelasticity of natural soft tissues. The properties of mechanical strength and cell responsive motif density could be controlled by modulating the proportion of the substrates in the enzymatic reaction. MC3T3 cells significantly differentiated into osteoblasts after seeded on the Q3GT-I3K hydrogel (2.8 kPa, 1.83 mM Q3GT) for 21 days, identifying from the elevated expression of alkaline phosphatase and substantial calcium nodule formation. Importantly, the engineered hydrogels exert a synergistic effect on the cell behaviors such as early adhesion, late proliferation, and differentiation of MC3T3-E1 cells. This paper introduces a new strategy for designing tissue engineering scaffold materials with specific functions.
Collapse
Affiliation(s)
- Ruirui Hao
- Heze Branch, Qilu University of Technology (Shandong Academy of Sciences), Biological Engineering Technology Innovation Center of Shandong Province, 1999 Taishan Road, Heze, 274000, China; State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xiwen Niu
- Heze Branch, Qilu University of Technology (Shandong Academy of Sciences), Biological Engineering Technology Innovation Center of Shandong Province, 1999 Taishan Road, Heze, 274000, China
| | - Xinrui Jiang
- Heze Branch, Qilu University of Technology (Shandong Academy of Sciences), Biological Engineering Technology Innovation Center of Shandong Province, 1999 Taishan Road, Heze, 274000, China
| | - Kang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xiaoyue Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Cuixia Chen
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China.
| |
Collapse
|
2
|
Benita BA, Koss KM. Peptide discovery across the spectrum of neuroinflammation; microglia and astrocyte phenotypical targeting, mediation, and mechanistic understanding. Front Mol Neurosci 2024; 17:1443985. [PMID: 39634607 PMCID: PMC11616451 DOI: 10.3389/fnmol.2024.1443985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Uncontrolled and chronic inflammatory states in the Central Nervous System (CNS) are the hallmark of neurodegenerative pathology and every injury or stroke-related insult. The key mediators of these neuroinflammatory states are glial cells known as microglia, the resident immune cell at the core of the inflammatory event, and astroglia, which encapsulate inflammatory insults in proteoglycan-rich scar tissue. Since the majority of neuroinflammation is exclusively based on the responses of said glia, their phenotypes have been identified to be on an inflammatory spectrum encompassing developmental, homeostatic, and reparative behaviors as opposed to their ability to affect devastating cell death cascades and scar tissue formation. Recently, research groups have focused on peptide discovery to identify these phenotypes, find novel mechanisms, and mediate or re-engineer their actions. Peptides retain the diverse function of proteins but significantly reduce the activity dependence on delicate 3D structures. Several peptides targeting unique phenotypes of microglia and astroglia have been identified, along with several capable of mediating deleterious behaviors or promoting beneficial outcomes in the context of neuroinflammation. A comprehensive review of the peptides unique to microglia and astroglia will be provided along with their primary discovery methodologies, including top-down approaches using known biomolecules and naïve strategies using peptide and phage libraries.
Collapse
Affiliation(s)
| | - Kyle M. Koss
- Department of Surgery, University of Arizona, Tucson, AZ, United States
- Department of Neurobiology, University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
- Sealy Institute for Drug Discovery (SIDD), University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
| |
Collapse
|
3
|
Wei FL, Zhai Y, Wang TF, Zhao JW, Wang CL, Tang Z, Shen K, Wu H, Zheng R, Du MR, Heng W, Li XX, Yan XD, Gao QY, Guo Z, Qian JX, Zhou CP. Stem cell-homing biomimetic hydrogel promotes the repair of osteoporotic bone defects through osteogenic and angiogenic coupling. SCIENCE ADVANCES 2024; 10:eadq6700. [PMID: 39485837 PMCID: PMC11529719 DOI: 10.1126/sciadv.adq6700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
Osteoporotic bone defects refer to the disruption of bone structural integrity in patients with osteoporosis and pose a substantial challenge to orthopedic surgeons. In this study, we developed a biomimetic hydrogel to improve the osteogenic microenvironment and promote stem cell homing. This hydrogel served as a container for S-nitrosoglutathione and Ca2+, promoting the release of bioactive nitric oxide (NO) from bone marrow mesenchymal stem cells (BMSCs) and human vascular endothelial cells and activating the NO/cyclic guanosine monophosphate signaling pathway. These changes promote osteogenic and angiogenic couplings. The hydrogel simultaneously recruited BMSCs by conjugating the stem cell homing peptide SKPPGTSS. Using a rat distal femoral defect model, it was demonstrated that this hydrogel can effectively increase the formation of bone tissue and new blood vessels and has immune-regulating functions. We envision that this hydrogel may be a minimally invasive yet highly effective strategy for expediting the healing of osteoporotic bone defects.
Collapse
Affiliation(s)
- Fei-Long Wei
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
- Department of Orthopaedics, General Hospital of Central Theater Command (Wuhan General Hospital of Guangzhou Command, previously), Wuhan 430030, China
| | - Yuan Zhai
- Basic Medical College, Fourth Military Medical University, Xi'an 710032, China
| | - Tian-Fu Wang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Jing-Wei Zhao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Chao-Li Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen Tang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, China
| | - Hao Wu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Rui Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming-Rui Du
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Wei Heng
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Xiao-Xiang Li
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Xiao-Dong Yan
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Quan-You Gao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Ji-Xian Qian
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Cheng-Pei Zhou
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| |
Collapse
|
4
|
Malinowska AL, Huynh HL, Bose S. Peptide-Oligonucleotide Conjugation: Chemistry and Therapeutic Applications. Curr Issues Mol Biol 2024; 46:11031-11047. [PMID: 39451535 PMCID: PMC11506717 DOI: 10.3390/cimb46100655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Oligonucleotides have been identified as powerful therapeutics for treating genetic disorders and diseases related to epigenetic factors such as metabolic and immunological dysfunctions. However, they face certain obstacles in terms of limited delivery to tissues and poor cellular uptake due to their large size and often highly charged nature. Peptide-oligonucleotide conjugation is an extensively utilized approach for addressing the challenges associated with oligonucleotide-based therapeutics by improving their delivery, cellular uptake and bioavailability, consequently enhancing their overall therapeutic efficiency. In this review, we present an overview of the conjugation of oligonucleotides to peptides, covering the different strategies associated with the synthesis of peptide-oligonucleotide conjugates (POC), the commonly used peptides employed to generate POCs, with the aim to develop oligonucleotides with favourable pharmacokinetic (PK) or pharmacodynamic (PD) properties for therapeutic applications. The advantages and drawbacks of the synthetic methods and applications of POCs are also described.
Collapse
Affiliation(s)
| | | | - Sritama Bose
- Medical Research Council, Nucleic Acid Therapy Accelerator (UKRI), Research Complex at Harwell (RCaH), Rutherford Appleton Laboratory, Harwell OX11 0FA, UK
| |
Collapse
|
5
|
Rasoulian B, Sheikholislam Z, Houshdar Tehrani MH, Chegeni S, Hoveizi E, Rezayat SM, Tavakol S. Unveiling the superior function of RADA in bone regeneration compared to KSL as two critical cores within self-assembling peptide nanofibers: Insights from in vitro and in vivo studies. Regen Ther 2024; 26:999-1009. [PMID: 39553539 PMCID: PMC11564076 DOI: 10.1016/j.reth.2024.09.010] [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: 08/09/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Self-assembling peptide nanofibers have emerged as promising biomaterials in the realm of bone tissue engineering due to their biocompatibility, biodegradability, and ability to mimic the native extracellular matrix. This study delved into the comparative efficacy of two distinct self-assembling peptide nanofibers, RADA-BMHP1 and KSL-BMHP1, both incorporating the biological motif of BMHP1, but differing in their core peptide sequences. Methods Cell viability and osteogenic differentiation in rat mesenchymal stem cells (rMSCs), and bone regeneration in rat were compared. Results In vitro assays revealed that KSL-BMHP1 promoted enhanced cell viability, and nitric oxide production than RADA-BMHP1, an effect potentially attributable to its lower hydrophobicity and higher net charge at physiological pH. Conversely, RADA-BMHP1 induced superior osteogenic differentiation, evidenced by upregulation of key osteogenic genes, increased alkaline phosphatase activity (ALP), and enhanced matrix mineralization which may be attributed to its higher protein-binding potential and grand hydropathy, facilitating interactions between the peptide nanofibers and proteins involved in osteogenesis. In vivo experiments utilizing a rat bone defect model demonstrated that both peptide nanofibers improved bone regeneration at the genes level and ALP activity, with RADA-BMHP1 exhibiting a more pronounced increase in bone formation compared to KSL-BMHP1. Histological evaluation using H&E, Masson's trichrome and Wright-Giemsa staining confirmed the biocompatibility of both nanofibers. Conclusion These findings underscore the pivotal role of the core structure of self-assembling peptide nanofibers, beyond their biological motif, in the fate of tissue regeneration. Further research is warranted to optimize the physicochemical properties and functionalization of these nanofibers to enhance their efficacy in bone regeneration applications.
Collapse
Affiliation(s)
- Bita Rasoulian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- School of Biomedical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Zahra Sheikholislam
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Solmaz Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Tavakol BioMimetic Technologies Company, Tehran, Iran
| |
Collapse
|
6
|
Asar M, Newton-Northup J, Soendergaard M. Improving Pharmacokinetics of Peptides Using Phage Display. Viruses 2024; 16:570. [PMID: 38675913 PMCID: PMC11055145 DOI: 10.3390/v16040570] [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: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Phage display is a versatile method often used in the discovery of peptides that targets disease-related biomarkers. A major advantage of this technology is the ease and cost efficiency of affinity selection, also known as biopanning, to identify novel peptides. While it is relatively straightforward to identify peptides with optimal binding affinity, the pharmacokinetics of the selected peptides often prove to be suboptimal. Therefore, careful consideration of the experimental conditions, including the choice of using in vitro, in situ, or in vivo affinity selections, is essential in generating peptides with high affinity and specificity that also demonstrate desirable pharmacokinetics. Specifically, in vivo biopanning, or the combination of in vitro, in situ, and in vivo affinity selections, has been proven to influence the biodistribution and clearance of peptides and peptide-conjugated nanoparticles. Additionally, the marked difference in properties between peptides and nanoparticles must be considered. While peptide biodistribution depends primarily on physiochemical properties and can be modified by amino acid modifications, the size and shape of nanoparticles also affect both absorption and distribution. Thus, optimization of the desired pharmacokinetic properties should be an important consideration in biopanning strategies to enable the selection of peptides and peptide-conjugated nanoparticles that effectively target biomarkers in vivo.
Collapse
Affiliation(s)
- Mallika Asar
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Mette Soendergaard
- Cell Origins LLC, 1601 South Providence Road Columbia, Columbia, MO 65203, USA;
- Department of Chemistry, Western Illinois University, Macomb, IL 61455, USA
| |
Collapse
|
7
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
8
|
Binaymotlagh R, Chronopoulou L, Palocci C. Peptide-Based Hydrogels: Template Materials for Tissue Engineering. J Funct Biomater 2023; 14:jfb14040233. [PMID: 37103323 PMCID: PMC10145623 DOI: 10.3390/jfb14040233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Tissue and organ regeneration are challenging issues, yet they represent the frontier of current research in the biomedical field. Currently, a major problem is the lack of ideal scaffold materials' definition. As well known, peptide hydrogels have attracted increasing attention in recent years thanks to significant properties such as biocompatibility, biodegradability, good mechanical stability, and tissue-like elasticity. Such properties make them excellent candidates for 3D scaffold materials. In this review, the first aim is to describe the main features of a peptide hydrogel in order to be considered as a 3D scaffold, focusing in particular on mechanical properties, as well as on biodegradability and bioactivity. Then, some recent applications of peptide hydrogels in tissue engineering, including soft and hard tissues, will be discussed to analyze the most relevant research trends in this field.
Collapse
Affiliation(s)
- Roya Binaymotlagh
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Laura Chronopoulou
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Cleofe Palocci
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
9
|
Ho C, Wang C, Wu T, Kuan C, Liu Y, Wang T. Peptide-functionalized double network hydrogel with compressible shape memory effect for intervertebral disc regeneration. Bioeng Transl Med 2023; 8:e10447. [PMID: 36925718 PMCID: PMC10013763 DOI: 10.1002/btm2.10447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/23/2022] [Accepted: 10/30/2022] [Indexed: 11/19/2022] Open
Abstract
As a prominent approach to treat intervertebral disc (IVD) degeneration, disc transplantation still falls short to fully reconstruct and restore the function of native IVD. Here, we introduce an IVD scaffold consists of a cellulose-alginate double network hydrogel-based annulus fibrosus (AF) and a cellulose hydrogel-based nucleus pulposus (NP). This scaffold mimics native IVD structure and controls the delivery of Growth Differentiation Factor-5 (GDF-5), which induces differentiation of endogenous mesenchymal stem cells (MSCs). In addition, this IVD scaffold has modifications on MSC homing peptide and RGD peptide which facilitate the recruitment of MSCs to injured area and enhances their cell adhesion property. The benefits of this double network hydrogel are high compressibility, shape memory effect, and mechanical strength comparable to native IVD. In vivo animal study demonstrates successful reconstruction of injured IVD including both AF and NP. These findings suggest that this double network hydrogel can serve as a promising approach to IVD regeneration with other potential biomedical applications.
Collapse
Affiliation(s)
- Chia‐Yu Ho
- Department of Materials Science and EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Chen‐Chie Wang
- Department of Orthopedic SurgeryTaipei Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
- Department of Orthopedics, School of MedicineTzu Chi UniversityHualienTaiwan
| | - Tsung‐Chiao Wu
- Department of Orthopedic SurgeryTaipei Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationNew Taipei CityTaiwan
| | - Chen‐Hsiang Kuan
- Division of Plastic Surgery, Department of SurgeryNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Research Center for Developmental Biology and Regenerative MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Yu‐Chung Liu
- Department of Materials Science and EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Tzu‐Wei Wang
- Department of Materials Science and EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
10
|
Qin S, Zhu J, Zhang G, Sui Q, Niu Y, Ye W, Ma G, Liu H. Research progress of functional motifs based on growth factors in cartilage tissue engineering: A review. Front Bioeng Biotechnol 2023; 11:1127949. [PMID: 36824354 PMCID: PMC9941568 DOI: 10.3389/fbioe.2023.1127949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis is a chronic degenerative joint disease that exerts significant impacts on personal life quality, and cartilage tissue engineering is a practical treatment in clinical. Various growth factors are involved in cartilage regeneration and play important roles therein, which is the focus of current cartilage repair strategy. To compensate for the purification difficulty, high cost, poor metabolic stability, and circulating dilution of natural growth factors, the concept of functional motifs (also known as mimetic peptides) from original growth factor was introduced in recent studies. Here, we reviewed the selection mechanisms, biological functions, carrier scaffolds, and modification methods of growth factor-related functional motifs, and evaluated the repair performance in cartilage tissue engineering. Finally, the prospects of functional motifs in researches and clinical application were discussed.
Collapse
Affiliation(s)
- Shengao Qin
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Jiaman Zhu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guangyong Zhang
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Qijia Sui
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Yimeng Niu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Weilong Ye
- School of Stomatology, Dalian Medical University, Dalian, China,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Guowu Ma
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Huiying Liu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| |
Collapse
|
11
|
Targeting Agents in Biomaterial-Mediated Bone Regeneration. Int J Mol Sci 2023; 24:ijms24032007. [PMID: 36768328 PMCID: PMC9916506 DOI: 10.3390/ijms24032007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Bone diseases are a global public concern that affect millions of people. Even though current treatments present high efficacy, they also show several side effects. In this sense, the development of biocompatible nanoparticles and macroscopic scaffolds has been shown to improve bone regeneration while diminishing side effects. In this review, we present a new trend in these materials, reporting several examples of materials that specifically recognize several agents of the bone microenvironment. Briefly, we provide a subtle introduction to the bone microenvironment. Then, the different targeting agents are exposed. Afterward, several examples of nanoparticles and scaffolds modified with these agents are shown. Finally, we provide some future perspectives and conclusions. Overall, this topic presents high potential to create promising translational strategies for the treatment of bone-related diseases. We expect this review to provide a comprehensive description of the incipient state-of-the-art of bone-targeting agents in bone regeneration.
Collapse
|
12
|
Kanda M, Nagai T, Kondo N, Matsuura K, Akazawa H, Komuro I, Kobayashi Y. Pericardial Grafting of Cardiac Progenitor Cells in Self-Assembling Peptide Scaffold Improves Cardiac Function After Myocardial Infarction. Cell Transplant 2023; 32:9636897231174078. [PMID: 37191272 PMCID: PMC10192947 DOI: 10.1177/09636897231174078] [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: 03/17/2022] [Revised: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Many studies have explored cardiac progenitor cell (CPC) therapy for heart disease. However, optimal scaffolds are needed to ensure the engraftment of transplanted cells. We produced a three-dimensional hydrogel scaffold (CPC-PRGmx) in which high-viability CPCs were cultured for up to 8 weeks. CPC-PRGmx contained an RGD peptide-conjugated self-assembling peptide with insulin-like growth factor-1 (IGF-1). Immediately after creating myocardial infarction (MI), we transplanted CPC-PRGmx into the pericardial space on to the surface of the MI area. Four weeks after transplantation, red fluorescent protein-expressing CPCs and in situ hybridization analysis in sex-mismatched transplantations revealed the engraftment of CPCs in the transplanted scaffold (which was cellularized with host cells). The average scar area of the CPC-PRGmx-treated group was significantly smaller than that of the non-treated group (CPC-PRGmx-treated group = 46 ± 5.1%, non-treated MI group = 59 ± 4.5%; p < 0.05). Echocardiography showed that the transplantation of CPC-PRGmx improved cardiac function and attenuated cardiac remodeling after MI. The transplantation of CPCs-PRGmx promoted angiogenesis and inhibited apoptosis, compared to the untreated MI group. CPCs-PRGmx secreted more vascular endothelial growth factor than CPCs cultured on two-dimensional dishes. Genetic fate mapping revealed that CPC-PRGmx-treated mice had more regenerated cardiomyocytes than non-treated mice in the MI area (CPC-PRGmx-treated group = 0.98 ± 0.25%, non-treated MI group = 0.25 ± 0.04%; p < 0.05). Our findings reveal the therapeutic potential of epicardial-transplanted CPC-PRGmx. Its beneficial effects may be mediated by sustainable cell viability, paracrine function, and the enhancement of de novo cardiomyogenesis.
Collapse
Affiliation(s)
- Masato Kanda
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Nagai
- Department of Cardiology, Chemotherapy
Research Institute, KAKEN Hospital, International University of Health and Welfare,
Ichikawa-shi, Japan
| | - Naomichi Kondo
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical
Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women’s
Medical University, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
13
|
An Acid-Sensitive Bone Targeting Delivery System Carrying Acacetin Prevents Osteoporosis in Ovariectomized Mice. Pharmaceuticals (Basel) 2022; 16:ph16010002. [PMID: 36678499 PMCID: PMC9867347 DOI: 10.3390/ph16010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
One effective treatment for postmenopausal osteoporosis is to inhibit osteoclasts and subsequent bone resorption. In our study, we demonstrated that acacetin, a flavone with potential therapeutic effects in infections, cancers, and several metabolic disorders, inhibited osteoclast differentiation and bone resorption in vitro. For improving the efficacy of acacetin in vivo, we developed an acid-sensitive bone-targeting delivery system composed of an acid-sensitive linker (N-ε-maleimidocaproic acid hydrazide, EMCH) for ensuring an effective release of acacetin at the site of action and a hydrophilic aspartic acid hexapeptide ((Asp)6, D6) as the effective bone targeting agent. Our results revealed that Acacetin-EMCH-D6 specifically bound to the bone surface once administrated in vivo, prolonged the retention time in bone and released acacetin at the osteoclastic bone resorption sites where the acidity is higher. We further demonstrated that, in ovariectomy-induced osteoporosis mice, treatment with Acacetin-EMCH-D6 inhibited osteoclast formation and increased trabecular bone mass. On the contrary, neither acacetin nor EMCH-D6 with the same dosage alone showed significant anti-osteoporosis effects in vivo. Mechanistically, targeted delivery of acacetin to the bone resorption sites by Acacetin-EMCH-D6 inhibited autophagy through activating PI3K/AKT/mTOR pathway in osteoclasts, while the activation of autophagy by rapamycin partially reversed the inhibitory effects of acacetin in vitro and in vivo. In summary, our study, for the first time, showed that the acid-sensitive bone-targeting delivery system carrying acacetin was effective for the treatment of postmenopausal osteoporosis. Thus, targeted delivery of acacetin using Acacetin-EMCH-D6 to bone resorption sites is a promising therapy for osteoporosis.
Collapse
|
14
|
Zhang R, He Y, Tao B, Wu J, Hu X, Li X, Xia Z, Cai K. Multifunctional silicon calcium phosphate composite scaffolds promote stem cell recruitment and bone regeneration. J Mater Chem B 2022; 10:5218-5230. [PMID: 35737023 DOI: 10.1039/d2tb00687a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A scaffold is one of the most significant implants for treating bone injury, while the precise control of stem cell proliferation and differentiation within a scaffold is still challenging. In this work, a composite scaffold was designed to be capable of recruiting endogenous stem cells, stimulating osteogenic differentiation and achieving significant bone repair function. The designed SiCP + SF@PFS silica-calcium phosphate composite scaffold was obtained by mixing the peptide PFS containing silk fibroin solution with the SiCP scaffold, and treating with horseradish peroxidase and H2O2. The results showed that the composite scaffold was able to release the PFS peptide continuously to induce the migration of mesenchymal stem cells. Meanwhile, cell proliferation and osteogenic differentiation were also improved after being seeded on the scaffold. In the cranial defect rat model, the composite scaffold was able to recruit CD29+ and CD90+ cells one week after implantation around the injury sites. The results of Micro-CT, H&E staining, Masson's staining and immunohistochemical staining indicated that the composite scaffold was able to promote new bone formation significantly.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Ye He
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jing Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xinqiang Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xuan Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Zengzilu Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
15
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
16
|
Huang B, Li P, Chen M, Peng L, Luo X, Tian G, Wang H, Wu L, Tian Q, Li H, Yang Y, Jiang S, Yang Z, Zha K, Sui X, Liu S, Guo Q. Hydrogel composite scaffolds achieve recruitment and chondrogenesis in cartilage tissue engineering applications. J Nanobiotechnology 2022; 20:25. [PMID: 34991615 PMCID: PMC8740469 DOI: 10.1186/s12951-021-01230-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The regeneration and repair of articular cartilage remains a major challenge for clinicians and scientists due to the poor intrinsic healing of this tissue. Since cartilage injuries are often clinically irregular, tissue-engineered scaffolds that can be easily molded to fill cartilage defects of any shape that fit tightly into the host cartilage are needed. METHOD In this study, bone marrow mesenchymal stem cell (BMSC) affinity peptide sequence PFSSTKT (PFS)-modified chondrocyte extracellular matrix (ECM) particles combined with GelMA hydrogel were constructed. RESULTS In vitro experiments showed that the pore size and porosity of the solid-supported composite scaffolds were appropriate and that the scaffolds provided a three-dimensional microenvironment supporting cell adhesion, proliferation and chondrogenic differentiation. In vitro experiments also showed that GelMA/ECM-PFS could regulate the migration of rabbit BMSCs. Two weeks after implantation in vivo, the GelMA/ECM-PFS functional scaffold system promoted the recruitment of endogenous mesenchymal stem cells from the defect site. GelMA/ECM-PFS achieved successful hyaline cartilage repair in rabbits in vivo, while the control treatment mostly resulted in fibrous tissue repair. CONCLUSION This combination of endogenous cell recruitment and chondrogenesis is an ideal strategy for repairing irregular cartilage defects.
Collapse
Affiliation(s)
- Bo Huang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.,Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Pinxue Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Mingxue Chen
- Department of Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Liqing Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.,Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xujiang Luo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Guangzhao Tian
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Hao Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.,Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Liping Wu
- Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Qinyu Tian
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.,Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Huo Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.,Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yu Yang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Shuangpeng Jiang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Zhen Yang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Kangkang Zha
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Xiang Sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
| |
Collapse
|
17
|
Halloy F, Brönnimann P, Hall J, Schümperli D. Analysis of Oligonucleotide Biodistribution and Metabolization in Experimental Animals. Methods Mol Biol 2022; 2537:335-350. [PMID: 35895273 DOI: 10.1007/978-1-0716-2521-7_19] [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: 06/15/2023]
Abstract
We describe methods to follow the fate of oligonucleotides after their injection into experimental animals. The quantitation in various tissues, blood or bone marrow cells is possible by chemical ligation PCR. This method works independently of chemical modifications of the oligonucleotide and/or its conjugations to lipid or peptide moieties. Moreover, metabolization intermediates can be detected by mass spectrometry. Together with a readout assay for the biochemical or physiological effects, which will differ, depending on the particular purpose of the oligonucleotide, these methods allow for a comprehensive understanding of oligonucleotide behavior in a living organism.
Collapse
Affiliation(s)
- François Halloy
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Paulina Brönnimann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
- Translational Research Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Jonathan Hall
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Daniel Schümperli
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
18
|
Su J, Liu C, Bai H, Cong W, Tang H, Hu H, Su L, He S, Wang Y. Development of novel bone targeting peptide-drug conjugate of 13-aminomethyl-15-thiomatrine for osteoporosis therapy. RSC Adv 2021; 12:221-227. [PMID: 35424502 PMCID: PMC8978659 DOI: 10.1039/d1ra08136e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/13/2021] [Indexed: 01/01/2023] Open
Abstract
13-Aminomethyl-15-thiomatrine (M19) previously developed by our research group was a promising candidate for novel anti-osteoporosis drug development. However, the application of M19 was limited by its unsatisfactory druggability including poor chemical stability, excessively broad pharmacological activity and some degree of cytotoxicity. To solve these problems, M19-based bone targeting and cathepsin K sensitive peptide–drug conjugates (BTM19-1, BTM19-2 and BTM19-3) were developed to realize precise drug release in the bone tissue. Subsequent studies showed a rapid drug release process via cathepsin K digestion but sufficient stability over several hours in chymotrypsin. Besides, greatly improved chemical stability and strong hydroxyapatite binding affinity were also demonstrated. In biological evaluation studies, these PDCs showed less cytotoxicity and similar osteoclast inhibitory activity compared with the prototype drug. The optimal BTM19-2 could serve as a suitable candidate for further osteoporosis therapy research. 13-Aminomethyl-15-thiomatrine (M19) previously developed by our research group was a promising candidate for novel anti-osteoporosis drug development.![]()
Collapse
Affiliation(s)
- Jia Su
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine Zhejiang China
| | - Chao Liu
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Haohao Bai
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Wei Cong
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Hua Tang
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Li Su
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University Shanghai China
| | - Yong Wang
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine Zhejiang China
| |
Collapse
|
19
|
Halloy F, Hill AC, Hall J. Efficient Synthesis of 2'-O-Methoxyethyl Oligonucleotide-Cationic Peptide Conjugates. ChemMedChem 2021; 16:3391-3395. [PMID: 34358416 PMCID: PMC9291120 DOI: 10.1002/cmdc.202100388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/02/2021] [Indexed: 11/06/2022]
Abstract
Single-stranded phosphorothioate (PS) oligonucleotide drugs have shown potential for the treatment of several rare diseases. However, a barrier to their widespread use is that they exhibit activity in only a narrow range of tissues. One way to circumvent this constraint is to conjugate them to cationic cell-penetrating peptides (CPPs). Although there are several examples of morpholino and peptide nucleic acids conjugated with CPPs, there are noticeably few examples of PS oligonucleotide-CPP conjugates. This is surprising given that PS oligonucleotides presently represent the largest class of approved RNA-based drugs, including Nusinersen, that bears the 2'-O-methoxyethyl (MOE)-chemistry. In this work, we report a method for in-solution conjugation of cationic, hydrophobic peptides or human serum albumin to a 22-nucleotide MOE-PS oligonucleotide. Conjugates were obtained in high yields and purities. Our findings pave the way for their large-scale synthesis and testing in vivo.
Collapse
Affiliation(s)
- François Halloy
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir Prelog Weg 48093ZurichSwitzerland
- Current address: Department of PaediatricsMedical Sciences DivisionUniversity of OxfordOX1 3QXOxfordUK
| | - Alyssa C. Hill
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir Prelog Weg 48093ZurichSwitzerland
| | - Jonathan Hall
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir Prelog Weg 48093ZurichSwitzerland
| |
Collapse
|
20
|
Xia B, Deng Y, Lv Y, Chen G. Stem cell recruitment based on scaffold features for bone tissue engineering. Biomater Sci 2020; 9:1189-1203. [PMID: 33355545 DOI: 10.1039/d0bm01591a] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem-cell based therapy strategies are promising approaches for the treatment of bone defects. However, extensive cell expansion steps, the low rate of cell survival and uncontrolled differentiation of stem cells transplanted into the body currently remain key challenges in advancing stem cell therapeutics. An alternative strategy is to use specifically designed bone scaffolds to recruit endogenous stem cells upon implantation and to stimulate new bone formation and remodeling. Stem cell recruitment based on scaffold features for bone tissue engineering relies on the development of scaffolds that can effectively mobilize and recruit endogenous stem cells to the implantation site. This article addresses the recent advances in the recruitment of endogenous stem cells in applications of bone scaffolds, particularly focusing on chemical modification and physical characteristic modification of the scaffold for endogenous stem cell homing and recruitment. Finally, the continuing challenges and future directions of scaffold-based stem cell recruitment are discussed.
Collapse
Affiliation(s)
- Bin Xia
- Chongqing Technology and Business University, Chongqing 400067, P. R. China
| | | | | | | |
Collapse
|
21
|
Gelain F, Luo Z, Zhang S. Self-Assembling Peptide EAK16 and RADA16 Nanofiber Scaffold Hydrogel. Chem Rev 2020; 120:13434-13460. [DOI: 10.1021/acs.chemrev.0c00690] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Fabrizio Gelain
- Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013, Italy
- Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore, 3, Milan 20162, Italy
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, United States
| |
Collapse
|
22
|
Chu W, Prodromou R, Day KN, Schneible JD, Bacon KB, Bowen JD, Kilgore RE, Catella CM, Moore BD, Mabe MD, Alashoor K, Xu Y, Xiao Y, Menegatti S. Peptides and pseudopeptide ligands: a powerful toolbox for the affinity purification of current and next-generation biotherapeutics. J Chromatogr A 2020; 1635:461632. [PMID: 33333349 DOI: 10.1016/j.chroma.2020.461632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Following the consolidation of therapeutic proteins in the fight against cancer, autoimmune, and neurodegenerative diseases, recent advancements in biochemistry and biotechnology have introduced a host of next-generation biotherapeutics, such as CRISPR-Cas nucleases, stem and car-T cells, and viral vectors for gene therapy. With these drugs entering the clinical pipeline, a new challenge lies ahead: how to manufacture large quantities of high-purity biotherapeutics that meet the growing demand by clinics and biotech companies worldwide. The protein ligands employed by the industry are inadequate to confront this challenge: while featuring high binding affinity and selectivity, these ligands require laborious engineering and expensive manufacturing, are prone to biochemical degradation, and pose safety concerns related to their bacterial origin. Peptides and pseudopeptides make excellent candidates to form a new cohort of ligands for the purification of next-generation biotherapeutics. Peptide-based ligands feature excellent target biorecognition, low or no toxicity and immunogenicity, and can be manufactured affordably at large scale. This work presents a comprehensive and systematic review of the literature on peptide-based ligands and their use in the affinity purification of established and upcoming biological drugs. A comparative analysis is first presented on peptide engineering principles, the development of ligands targeting different biomolecular targets, and the promises and challenges connected to the industrial implementation of peptide ligands. The reviewed literature is organized in (i) conventional (α-)peptides targeting antibodies and other therapeutic proteins, gene therapy products, and therapeutic cells; (ii) cyclic peptides and pseudo-peptides for protein purification and capture of viral and bacterial pathogens; and (iii) the forefront of peptide mimetics, such as β-/γ-peptides, peptoids, foldamers, and stimuli-responsive peptides for advanced processing of biologics.
Collapse
Affiliation(s)
- Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kevin N Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kaitlyn B Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Carly M Catella
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Matthew D Mabe
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kawthar Alashoor
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14642
| | - Yiman Xu
- College of Material Science and Engineering, Donghua University, 201620 Shanghai, People's Republic of China
| | - Yuanxin Xiao
- College of Textile, Donghua University, Songjiang District, Shanghai, 201620, People's Republic of China
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606.
| |
Collapse
|
23
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
24
|
Halloy F, Iyer P, Ćwiek P, Ghidini A, Barman-Aksözen J, Wildner-Verhey van Wijk N, Theocharides APA, Minder E, Schneider-Yin X, Schümperli D, Hall J. Delivery of oligonucleotides to bone marrow to modulate ferrochelatase splicing in a mouse model of erythropoietic protoporphyria. Nucleic Acids Res 2020; 48:4658-4671. [PMID: 32313951 PMCID: PMC7229840 DOI: 10.1093/nar/gkaa229] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/25/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022] Open
Abstract
Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.
Collapse
Affiliation(s)
- François Halloy
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Pavithra S Iyer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Paulina Ćwiek
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Alice Ghidini
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | | | | | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | | | - Daniel Schümperli
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Jonathan Hall
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| |
Collapse
|
25
|
Xu H, Cao B, Li Y, Mao C. Phage nanofibers in nanomedicine: Biopanning for early diagnosis, targeted therapy, and proteomics analysis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1623. [PMID: 32147974 DOI: 10.1002/wnan.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Display of a peptide or protein of interest on the filamentous phage (also known as bacteriophage), a biological nanofiber, has opened a new route for disease diagnosis and therapy as well as proteomics. Earlier phage display was widely used in protein-protein or antigen-antibody studies. In recent years, its application in nanomedicine is becoming increasingly popular and encouraging. We aim to review the current status in this research direction. For better understanding, we start with a brief introduction of basic biology and structure of the filamentous phage. We present the principle of phage display and library construction method on the basis of the filamentous phage. We summarize the use of the phage displayed peptide library for selecting peptides with high affinity against cells or tissues. We then review the recent applications of the selected cell or tissue targeting peptides in developing new targeting probes and therapeutics to advance the early diagnosis and targeted therapy of different diseases in nanomedicine. We also discuss the integration of antibody phage display and modern proteomics in discovering new biomarkers or target proteins for disease diagnosis and therapy. Finally, we propose an outlook for further advancing the potential impact of phage display on future nanomedicine. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Hong Xu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
26
|
Chander V, Gangenahalli G. Pluronic-F127/Platelet Microvesicles nanocomplex delivers stem cells in high doses to the bone marrow and confers post-irradiation survival. Sci Rep 2020; 10:156. [PMID: 31932650 PMCID: PMC6957521 DOI: 10.1038/s41598-019-57057-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/21/2019] [Indexed: 01/06/2023] Open
Abstract
Platelet microvesicles (pMVs) are submicron-sized heterogeneous vesicles released upon activation and contain several membrane receptors and proteins (CD41, CD61, CD62, CXCR4, PAR-1, etc.). We have revealed their ability to adhere to the triblock copolymer pluronic-F127 (PF127) and form a platelet microvesicular nanocloud which has the potential to enhance the transvascular migration of hematopoietic stem cells across the sinusoidal endothelium to the bone marrow. Besides, the pMVs nanoclouds bestow survival benefits when present on the cells used for infusion, particularly with PF127-stabilized with chitosan-alginate (PF127-CA HSCs). The vesicles were found to be firmly associated with PF127 in the nanocloud, which was detected by confocal laser scanning microscopy. The abrogation of CXCR4/SDF-1 axis regulating the transmigration of the cells by antagonist AMD3100 revealed that the enriched CXCR4 receptors on pMVs robustize the transmigration of the infused cells. The homing of the cells led to effective engraftment and faster regeneration of the critical blood lineages, which elicited 100% survival of the mice receiving lethal doses of radiation. The Human Long-Term Culture Initiating Cells (LTC-ICs), Severe Combined Immunodeficient (SCID) - Repopulating Cells (SRCs) and Colony Forming Cells (CFCs) responsible for the regeneration, but present in extremely low numbers in the infused cell dose, have enabled the cells to reach the bone marrow in high numbers. This potential of the PF127 to sequester the pMVs and its application to achieve over 10-fold delivery of HSCs across the trans-endothelial checkpoint has so far not been reported. Thus, this mechanistic innovation is a potential post-exposure life-saving regimen capable of circumventing the irreparable damage to the bone marrow caused by lethal doses of radiation.
Collapse
Affiliation(s)
- Vikas Chander
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Delhi, 110054, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Delhi, 110054, India.
| |
Collapse
|
27
|
Cao B, Li Y, Yang T, Bao Q, Yang M, Mao C. Bacteriophage-based biomaterials for tissue regeneration. Adv Drug Deliv Rev 2019; 145:73-95. [PMID: 30452949 PMCID: PMC6522342 DOI: 10.1016/j.addr.2018.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 07/24/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Bacteriophage, also called phage, is a human-safe bacteria-specific virus. It is a monodisperse biological nanostructure made of proteins (forming the outside surface) and nucleic acids (encased in the protein capsid). Among different types of phages, filamentous phages have received great attention in tissue regeneration research due to their unique nanofiber-like morphology. They can be produced in an error-free format, self-assemble into ordered scaffolds, display multiple signaling peptides site-specifically, and serve as a platform for identifying novel signaling or homing peptides. They can direct stem cell differentiation into specific cell types when they are organized into proper patterns or display suitable peptides. These unusual features have allowed scientists to employ them to regenerate a variety of tissues, including bone, nerves, cartilage, skin, and heart. This review will summarize the progress in the field of phage-based tissue regeneration and the future directions in this field.
Collapse
Affiliation(s)
- Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Zhejiang, Hangzhou 310058, China.
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States; School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| |
Collapse
|
28
|
Tavakol S, Rasoulian B, Ramezani F, Hoveizi E, Tavakol B, Rezayat SM. Core and biological motif of self-assembling peptide nanofiber induce a stronger electrostatic interaction than BMP2 with BMP2 receptor 1A. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:148-158. [PMID: 31029307 DOI: 10.1016/j.msec.2019.03.097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 02/03/2019] [Accepted: 03/25/2019] [Indexed: 12/27/2022]
Abstract
Recent studies suggest that nanotopography can trigger colocalization of integrins and bone morphogenetic protein 2 (BMP2) receptors (e.g., BMPR1A), thereby leading to osteogenesis. In this study, the bone marrow homing peptide 1 (BMHP1) motif was bound to a self-assembling peptide core to form a hydrogel-based nanofiber (R-BMHP1). The docking and molecular dynamic study revealed that the R-BMHP1 sequence induced a stronger electrostatic interaction than BMP2 through arginines in the RADA core sequence and through lysine24 in the BMHP1 motif with BMPR1A. Notably, decrease of polar solvation binding energy will enhance the total binding energy and increases bone regeneration even more than BMP2 The enhanced osteogenesis and bone repair potential of R-BMHP1 nanofiber might be related to its chemical interaction with BMPR1A, which triggered downstream signal transduction through osteogenic genes overexpression in osteo-differentiated mesenchymal stem cells (MSCs), as well as implanted critical-sized bone defects in rats. Following that, calcium deposition occurred by osteoblast-like cells, ALP activity increased in osteodifferentiation MSCs and rat serum, and calcium density improved in bone defects (X-ray). The nanofiber was biocompatible and enhanced the cell viability of MSCs, without multinuclear cell infiltration into the defect site. Taking everything into account, not only does nanotopography induce osteogenesis through colocalization of BMPRs and integrins, but also R-BMHP1 nanofibers (considering their chemical structure) induce cell proliferation, osteogenesis, and bone repair through strong electrostatic interaction with BMPR1A and downstream signaling. The entire outcome of this study manifests the plausibility of R-BMHP1 for spine and spinal cord injury repair.
Collapse
Affiliation(s)
- Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Bita Rasoulian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Behnaz Tavakol
- School of Medicine, Kashan University of Medical Sciences, Isfahan, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Lu J, Shen X, Sun X, Yin H, Yang S, Lu C, Wang Y, Liu Y, Huang Y, Yang Z, Dong X, Wang C, Guo Q, Zhao L, Sun X, Lu S, Mikos AG, Peng J, Wang X. Increased recruitment of endogenous stem cells and chondrogenic differentiation by a composite scaffold containing bone marrow homing peptide for cartilage regeneration. Theranostics 2018; 8:5039-5058. [PMID: 30429885 PMCID: PMC6217070 DOI: 10.7150/thno.26981] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/27/2018] [Indexed: 01/17/2023] Open
Abstract
Even small cartilage defects could finally degenerate to osteoarthritis if left untreated, owing to the poor self-healing ability of articular cartilage. Stem cell transplantation has been well implemented as a common approach in cartilage tissue engineering but has technical complexity and safety concerns. The stem cell homing-based technique emerged as an alternative promising therapy for cartilage repair to overcome traditional limitations. In this study, we constructed a composite hydrogel scaffold by combining an oriented acellular cartilage matrix (ACM) with a bone marrow homing peptide (BMHP)-functionalized self-assembling peptide (SAP). We hypothesized that increased recruitment of endogenous stem cells by the composite scaffold could enhance cartilage regeneration. Methods: To test our hypothesis, in vitro proliferation, attachment and chondrogenic differentiation of rabbit mesenchymal stem cells (MSCs) were tested to confirm the bioactivities of the functionalized peptide hydrogel. The composite scaffold was then implanted into full-thickness cartilage defects on rabbit knee joints for cartilage repair, in comparison with microfracture or other sample groups. Stem cell recruitment was monitored by dual labeling with CD29 and CD90 under confocal microcopy at 1 week after implantation, followed by chondrogenic differentiation examined by qRT-PCR. Repaired tissue of the cartilage defects was evaluated by histological and immunohistochemistry staining, microcomputed tomography (micro-CT) and magnetic resonance imaging (MRI) at 3 and 6 months post-surgery. Macroscopic and histological scoring was done to evaluate the optimal in vivo repair outcomes of this composite scaffold. Results: The functionalized SAP hydrogels could stimulate rabbit MSC proliferation, attachment and chondrogenic differentiation during in vitro culture. At 7 days after implantation, increased recruitment of MSCs based on CD29+ /CD90+ double-positive cells was found in vivo in the composite hydrogel scaffold, as well as upregulation of cartilage-associated genes (aggrecan, Sox9 and type II collagen). After 3 and 6 months post-surgery, the articular cartilage defect in the composite scaffold-treated group was fully covered with cartilage-like tissue with a smooth surface, which was similar to the surrounding native cartilage, according to the results of histological and immunohistochemistry staining, micro-CT and MRI analysis. Macroscopic and histological scoring confirmed that the quality of cartilage repair was significantly improved with implantation of the composite scaffold at each timepoint, in comparison with microfracture or other sample groups. Conclusion: Our findings demonstrated that the composite scaffold could enhance endogenous stem cell homing and chondrogenic differentiation and significantly improve the therapeutic outcome of chondral defects. The present study provides a promising approach for in vivo cartilage repair without cell transplantation. Optimization of this strategy may offer great potential and benefits for clinical application in the future.
Collapse
Affiliation(s)
- Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xuezhen Shen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Heyong Yin
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Changfeng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yifan Liu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Yingqi Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zijin Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xianqi Dong
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Chenhao Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Shibi Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Antonios G. Mikos
- Department of Bioengineering, Bioscience Research Collaborative, Rice University, Texas 77030, USA
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
30
|
Mason TO, Shimanovich U. Fibrous Protein Self-Assembly in Biomimetic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706462. [PMID: 29883013 DOI: 10.1002/adma.201706462] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/28/2018] [Indexed: 05/22/2023]
Abstract
Protein self-assembly processes, by which polypeptides interact and independently form multimeric structures, lead to a wide array of different endpoints. Structures formed range from highly ordered molecular crystals to amorphous aggregates. Order arises in the system from a balance between many low-energy processes occurring due to a set of interactions between residues in a chain, between residues in different chains, and between solute and solvent. In Nature, self-assembling protein systems have evolved over millions of years to organize into supramolecular structures, optimized for specific functions, with this propensity determined by the sequence of their constituent amino acids, of which only 20 are encoded in DNA. The structural materials that arise from biological self-assembly can display remarkable mechanical properties, often as a result of hierarchical structure on the nano- and microscales, and much research has been devoted to mimicking and exploiting these properties for a variety of end uses. This work presents a review of a range of studies in which biological functions are effectively reproduced through the design of self-assembling fibrous protein systems.
Collapse
Affiliation(s)
- Thomas O Mason
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ulyana Shimanovich
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, 7610001, Israel
| |
Collapse
|
31
|
Martins IM, Reis RL, Azevedo HS. Phage Display Technology in Biomaterials Engineering: Progress and Opportunities for Applications in Regenerative Medicine. ACS Chem Biol 2016; 11:2962-2980. [PMID: 27661443 DOI: 10.1021/acschembio.5b00717] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of regenerative medicine has been gaining momentum steadily over the past few years. The emphasis in regenerative medicine is to use various in vitro and in vivo approaches that leverage the intrinsic healing mechanisms of the body to treat patients with disabling injuries and chronic diseases such as diabetes, osteoarthritis, and degenerative disorders of the cardiovascular and central nervous system. Phage display has been successfully employed to identify peptide ligands for a wide variety of targets, ranging from relatively small molecules (enzymes, cell receptors) to inorganic, organic, and biological (tissues) materials. Over the past two decades, phage display technology has advanced tremendously and has become a powerful tool in the most varied fields of research, including biotechnology, materials science, cell biology, pharmacology, and diagnostics. The growing interest in and success of phage display libraries is largely due to its incredible versatility and practical use. This review discusses the potential of phage display technology in biomaterials engineering for applications in regenerative medicine.
Collapse
Affiliation(s)
- Ivone M. Martins
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- CEB − Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
| | - Rui L. Reis
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helena S. Azevedo
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
- Institute
of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom
| |
Collapse
|
32
|
Koss K, Unsworth L. Neural tissue engineering: Bioresponsive nanoscaffolds using engineered self-assembling peptides. Acta Biomater 2016; 44:2-15. [PMID: 27544809 DOI: 10.1016/j.actbio.2016.08.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 12/25/2022]
Abstract
UNLABELLED Rescuing or repairing neural tissues is of utmost importance to the patient's quality of life after an injury. To remedy this, many novel biomaterials are being developed that are, ideally, non-invasive and directly facilitate neural wound healing. As such, this review surveys the recent approaches and applications of self-assembling peptides and peptide amphiphiles, for building multi-faceted nanoscaffolds for direct application to neural injury. Specifically, methods enabling cellular interactions with the nanoscaffold and controlling the release of bioactive molecules from the nanoscaffold for the express purpose of directing endogenous cells in damaged or diseased neural tissues is presented. An extensive overview of recently derived self-assembling peptide-based materials and their use as neural nanoscaffolds is presented. In addition, an overview of potential bioactive peptides and ligands that could be used to direct behaviour of endogenous cells are categorized with their biological effects. Finally, a number of neurotrophic and anti-inflammatory drugs are described and discussed. Smaller therapeutic molecules are emphasized, as they are thought to be able to have less potential effect on the overall peptide self-assembly mechanism. Options for potential nanoscaffolds and drug delivery systems are suggested. STATEMENT OF SIGNIFICANCE Self-assembling nanoscaffolds have many inherent properties making them amenable to tissue engineering applications: ease of synthesis, ease of customization with bioactive moieties, and amenable for in situ nanoscaffold formation. The combination of the existing knowledge on bioactive motifs for neural engineering and the self-assembling propensity of peptides is discussed in specific reference to neural tissue engineering.
Collapse
|
33
|
Silva VL, Ferreira D, Nobrega FL, Martins IM, Kluskens LD, Rodrigues LR. Selection of Novel Peptides Homing the 4T1 CELL Line: Exploring Alternative Targets for Triple Negative Breast Cancer. PLoS One 2016; 11:e0161290. [PMID: 27548261 PMCID: PMC4993384 DOI: 10.1371/journal.pone.0161290] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/27/2016] [Indexed: 11/30/2022] Open
Abstract
The use of bacteriophages to select novel ligands has been widely explored for cancer therapy. Their application is most warranted in cancer subtypes lacking knowledge on how to target the cancer cells in question, such as the triple negative breast cancer, eventually leading to the development of alternative nanomedicines for cancer therapeutics. Therefore, the following study aimed to select and characterize novel peptides for a triple negative breast cancer murine mammary carcinoma cell line– 4T1. Using phage display, 7 and 12 amino acid random peptide libraries were screened against the 4T1 cell line. A total of four rounds, plus a counter-selection round using the 3T3 murine fibroblast cell line, was performed. The enriched selective peptides were characterized and their binding capacity towards 4T1 tissue samples was confirmed by immunofluorescence and flow cytometry analysis. The selected peptides (4T1pep1 –CPTASNTSC and 4T1pep2—EVQSSKFPAHVS) were enriched over few rounds of selection and exhibited specific binding to the 4T1 cell line. Interestingly, affinity to the human MDA-MB-231 cell line was also observed for both peptides, promoting the translational application of these novel ligands between species. Additionally, bioinformatics analysis suggested that both peptides target human Mucin-16. This protein has been implicated in different types of cancer, as it is involved in many important cellular functions. This study strongly supports the need of finding alternative targeting systems for TNBC and the peptides herein selected exhibit promising future application as novel homing peptides for breast cancer therapy.
Collapse
Affiliation(s)
- Vera L. Silva
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
| | - Debora Ferreira
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
| | - Franklin L. Nobrega
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
| | - Ivone M. Martins
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
| | - Leon D. Kluskens
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
| | - Ligia R. Rodrigues
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, Braga, Portugal
- * E-mail:
| |
Collapse
|
34
|
Fan Z, Cui X, Wei D, Liu W, Li B, He H, Ye H, Zhu N, Wei X. eEF1A1 binds and enriches protoporphyrin IX in cancer cells in 5-aminolevulinic acid based photodynamic therapy. Sci Rep 2016; 6:25353. [PMID: 27150264 PMCID: PMC4858656 DOI: 10.1038/srep25353] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/08/2016] [Indexed: 12/11/2022] Open
Abstract
Photodynamic therapy (PDT) with protoporphyrin IX (PpIX), which is endogenously derived from 5-aminolevulinic acid (5-ALA) or its derivatives, is a promising modality for the treatment of both pre-malignant and malignant lesions. However, the mechanisms of how ALA-induced PpIX selectively accumulated in the tumors are not fully elucidated. Here we discovered that eukaryotic translation elongation factor 1 alpha 1 (eEF1A1) interacted with PpIX (with an affinity constant of 2.96 × 10(6) M(-1)). Microscopy imaging showed that ALA-induced PpIX was co-localized with eEF1A1 in cancer cells. eEF1A1 was found to enrich ALA-induced PpIX in cells by competitively blocking the downstream bioavailability of PpIX. Taken together, our study discovered eEF1A1 as a novel photosensitizer binding protein, which may play an essential role in the enrichment of ALA-induced PpIX in cancer cells during PDT. These suggested eEF1A1 as a molecular marker to predict the selectivity and efficiency of 5-ALA based PDT in cancer therapy.
Collapse
Affiliation(s)
- Zhichao Fan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, California, USA
| | - Xiaojun Cui
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Dan Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Liu
- Cell Death and Survival Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Buhong Li
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, China
| | - Hao He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Huamao Ye
- Department of Urology, Changhai Hospital, Second Military University, Shanghai, China
| | - Naishuo Zhu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Han K, Yin WN, Fan JX, Cao FY, Zhang XZ. Photo-Activatable Substrates for Site-Specific Differentiation of Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:23679-23684. [PMID: 26452046 DOI: 10.1021/acsami.5b07455] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this report, a UV sensitive, PEGylated PFSSTKTC (Pro-Phe-Ser-Ser-Thr-Lys-Thr-Cys) peptide was modified on quartz substrate to investigate the spatial controlled differentiation of stem cells. This substrate could restrict the cell adhesion due to the steric hindrance of PEG shell. With UV irradiation, PFSSTKTC became exposed owing to the breakage of o-nitrobenzyl group with the detachment of PEG shell. The irradiation boundary on substrate was stable in the long term. The in vitro osteogenic differentiation results revealed that under the site-specific irradiation, the mesenchymal stem cells (MSCs) could specifically differentiate into osteoblast under the induction of PFSSTKTC peptide. This photoactivatable biomaterial shows great potential for region controllable and precise MSCs differentiation.
Collapse
Affiliation(s)
- Kai Han
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
- College of Science, Huazhong Agricultural University , Wuhan 430070, China
| | - Wei-Na Yin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Feng-Yi Cao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University , Wuhan 430072, China
| |
Collapse
|
36
|
Wu M, Ye Z, Zhu H, Zhao X. Self-Assembling Peptide Nanofibrous Hydrogel on Immediate Hemostasis and Accelerative Osteosis. Biomacromolecules 2015; 16:3112-8. [PMID: 26348089 DOI: 10.1021/acs.biomac.5b00493] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The use of local agents to achieve hemostasis of bone that does not interfere with repair and recovery is a complex and emergency subject in surgery. In this study, the dual functional biodegradable self-assembling nanopeptide (SAP) RADA16-I was synthesized by solid phase synthesis and was shown to exhibit immediate hemostasis and accelerative osteosis. The RADA16-I showed good performance as a hemostatic agent, which was investigated by comparison with the effects of bone wax in the ilium bone defect model of New Zealand rabbits. The RADA16-I exhibited efficient function of bone regeneration in both radiographic analysis and histological examination, while the bone wax inhibited osteogenesis. Moreover, in in vivo experiment, the RADA16-I was shown to exhibit excellent biocompatibility, while the group with bone wax showed a severe inflammatory response at the interface with bone. Thus, the RADA16-I is proven to be an excellent biocompatible material with effective dual function of hemostasis and osteosis.
Collapse
Affiliation(s)
- Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University , Chengdu, 610041 Sichuan, China.,Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University , Chengdu, 610041 Sichuan, China
| | - Zhaoyang Ye
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University , Chengdu, 610041 Sichuan, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University , Chengdu, 610041 Sichuan, China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University , Chengdu, 610041 Sichuan, China.,Center for Biomedical Engineering, NE47-379, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139-4307, United States
| |
Collapse
|
37
|
Tavakol S, Saber R, Hoveizi E, Aligholi H, Ai J, Rezayat SM. Chimeric Self-assembling Nanofiber Containing Bone Marrow Homing Peptide’s Motif Induces Motor Neuron Recovery in Animal Model of Chronic Spinal Cord Injury; an In Vitro and In Vivo Investigation. Mol Neurobiol 2015; 53:3298-3308. [DOI: 10.1007/s12035-015-9266-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/28/2015] [Indexed: 02/04/2023]
|
38
|
Cao FY, Yin WN, Fan JX, Tao L, Qin SY, Zhuo RX, Zhang XZ. Evaluating the effects of charged oligopeptide motifs coupled with RGD on osteogenic differentiation of mesenchymal stem cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:6698-6705. [PMID: 25748883 DOI: 10.1021/acsami.5b00064] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Mesenchymal stem cells, due to their multilineage differentiation potential, have emerged as a promising cell candidate for cell-based therapy. In recent years, biomaterials were artificially synthesized to control the differentiation of mesenchymal stem cells. In this study, a series of charged or neutral oligopeptide motifs coupled with RGD were synthesized and used for surface modification using quartz substrates as model. Cell behaviors on the modified surfaces with different charged oligopeptide motifs were studied. It was found that these different charged oligopeptide motifs coupled with RGD were biocompatible for cell proliferation and adhesion. Moreover, it was demonstrated that the positively charged oligopeptide motif could inhibit osteogenic differentiation, while the negatively charged and neutral oligopeptide motifs could enhance osteogenic differentiation in the presence of RGD. This work may bring us enlightenment that different charged oligopeptide motifs coupled with RGD may be used for biomaterial surface modification for different stem cell-based therapies.
Collapse
|
39
|
Cao FY, Yin WN, Fan JX, Zhuo RX, Zhang XZ. A novel function of BMHP1 and cBMHP1 peptides to induce the osteogenic differentiation of mesenchymal stem cells. Biomater Sci 2015. [DOI: 10.1039/c4bm00300d] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BMHP1 or cBMHP1 peptide is found to induce MSCs towards the osteogenic lineage when tethered to modified quartz substrates.
Collapse
Affiliation(s)
- Feng-Yi Cao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Wei-Na Yin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| |
Collapse
|
40
|
Lei J, Li P, Zhang Q, Wang Y, Zhang Z, Ding X, Zang W. Anti-idiotypic nanobody-phage based real-time immuno-PCR for detection of hepatocarcinogen aflatoxin in grains and feedstuffs. Anal Chem 2014; 86:10841-6. [PMID: 25273352 DOI: 10.1021/ac5029424] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aflatoxins are a group of extremely toxic small molecules that have been involved in human hepatic and extrahepatic carcinogenesis as causative agents. Herein, we developed a real-time immuno polymerase chain reaction (IPCR) assay for the accurately quantitative detection of aflatoxins in agri-products base on a M13 phage containing aflatoxin anti-idiotypic nanobody and its encoding DNA which was used to design the specific primers. The limit of detection (LOD) of the assay is 0.02 ng/mL, which exhibits a 4-fold improvement over traditional phage ELISA. The developed method was successfully validated with the samples of corn, rice, peanut, and feedstuff, which are major aflatoxin-contaminated agri-products. And the recoveries were from 77.05 to 122.16%. For further validation, the developed assay was also compared with a reference HPLC method for the analysis of aflatoxins in corn and peanuts, and concordant results (R(2) = 0.991) were obtained. In this context, this study provides a novel opportunity to analyze aflatoxins in agri-products.
Collapse
Affiliation(s)
- Jiawen Lei
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences , Wuhan, 430062, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
41
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
42
|
Zhao W, Jin L, Yuan H, Tan Z, Zhou C, Li LS, Ma L. Targeting human embryonic stem cells with quantum dot-conjugated phages. Sci Rep 2013; 3:3134. [PMID: 24190250 PMCID: PMC3817438 DOI: 10.1038/srep03134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 10/15/2013] [Indexed: 12/11/2022] Open
Abstract
Targeting embryonic stem cells (ESCs) is important for ESC labeling, drug delivery and cell fate control. In this study, we identified twenty-two phage clones that bind specifically to the hESC cell line X-01, which was derived from human blastocysts of Chinese origin. One phage (H178), which displays the sequence VGGEAWSSPTDL, showed higher binding affinity to hESCs than to a monkey ES cell line (RS366.4) and two mouse ES cell lines (R1 and E14). Using quantum dots (QDs) conjugated to the H178 phage, we demonstrate that the phage can specifically bind to hESCs in vitro. Our results suggest a possible interaction between the selected peptide and the stem cell extracellular matrix (ECM). The selection method described here allows rapid and efficient screening of unique phage clones and targeting cells. The phages displaying peptides identified by this study have potential applications for cargo delivery and receptor studies.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Division of Life Science & Health, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Wu KC, Tseng CL, Wu CC, Kao FC, Tu YK, C So E, Wang YK. Nanotechnology in the regulation of stem cell behavior. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2013; 14:054401. [PMID: 27877605 PMCID: PMC5090368 DOI: 10.1088/1468-6996/14/5/054401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/16/2013] [Indexed: 05/19/2023]
Abstract
Stem cells are known for their potential to repair damaged tissues. The adhesion, growth and differentiation of stem cells are likely controlled by the surrounding microenvironment which contains both chemical and physical cues. Physical cues in the microenvironment, for example, nanotopography, were shown to play important roles in stem cell fate decisions. Thus, controlling stem cell behavior by nanoscale topography has become an important issue in stem cell biology. Nanotechnology has emerged as a new exciting field and research from this field has greatly advanced. Nanotechnology allows the manipulation of sophisticated surfaces/scaffolds which can mimic the cellular environment for regulating cellular behaviors. Thus, we summarize recent studies on nanotechnology with applications to stem cell biology, including the regulation of stem cell adhesion, growth, differentiation, tracking and imaging. Understanding the interactions of nanomaterials with stem cells may provide the knowledge to apply to cell-scaffold combinations in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chi-Chang Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - Feng-Chen Kao
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Edmund C So
- Department of Anesthesiology, Tainan Municipal An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Yang-Kao Wang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
- Medical Device Innovation Center, National Cheng-Kung University, Tainan, Taiwan
| |
Collapse
|
44
|
Saracino GAA, Gelain F. Modelling and analysis of early aggregation events of BMHP1-derived self-assembling peptides. J Biomol Struct Dyn 2013; 32:759-75. [PMID: 23730849 DOI: 10.1080/07391102.2013.790848] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the increasing use and development of peptide-based scaffolds in different fields including that of regenerative medicine, the understanding of the factors governing the self-assembly process and the relationship between sequence and properties have not yet been fully understood. BMHP1-derived self-assembling peptides (SAPs) have been developed and characterized showing that biotinylation at the N-terminal cap corresponds to better performing assembly and scaffold biomechanics. In this study, the effects of biotinylation on the self-assembly dynamics of seven BMHP1-derived SAPs have been investigated by molecular dynamics simulations. We confirmed that these SAPs self-assemble into β-structures and that proline acts as a β-breaker of the assembled aggregates. In biotinylated peptides, the formation of ordered β-structured aggregates is triggered by both the establishment of a dense and dynamic H-bonds network and the formation of a 'hydrophobic wall' available to interact with other peptides. Such conditions result from the peculiar chemical composition of the biotinyl-cap, given by the synergic cooperation of the uracil function of the ureido ring with the high hydrophobic portion consisting of the thiophenyl ring and valeryl chain. The inbuilt propensity of biotinylated peptides towards the formation of ordered small aggregates makes them ideal precursors of higher hierarchically organized self-assembled nanostructures as experimentally observed.
Collapse
Affiliation(s)
- Gloria Anna Ada Saracino
- a Center of Nanomedicine and Tissue Engineering A. O. Ospedale Niguarda Ca' Granda , Milan , 20162 Italy
| | | |
Collapse
|
45
|
Liu X, Wang X, Wang X, Ren H, He J, Qiao L, Cui FZ. Functionalized self-assembling peptide nanofiber hydrogels mimic stem cell niche to control human adipose stem cell behavior in vitro. Acta Biomater 2013; 9:6798-805. [PMID: 23380207 DOI: 10.1016/j.actbio.2013.01.027] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 01/21/2013] [Accepted: 01/25/2013] [Indexed: 10/27/2022]
Abstract
A class of designer functionalized self-assembling peptide nanofiber scaffolds developed from self-assembling peptide RADA16-I (AcN-RADARADARADARADA-CONH2) has become increasingly attractive not only for studying spatial behaviors of cells, but also for developing approaches for a wide range of medical applications including regenerative medicine, rapid hemostasis and cell therapy. In this study, we report three functionalized self-assembling peptide hydrogels that serve as a three-dimensional (3-D) artificial microenvironment to control human adipose stem cell (hASC) behavior in vitro. Short peptide motifs SKPPGTSS (bone marrow homing motif), FHRRIKA (heparin-binding motif) and PRGDSGYRGDS (two-unit RGD cell adhesion motif) were used to extend the C-terminus of RADA16-I to obtain functionalized peptides. Atomic force microscopy confirmed the formation of self-assembling nanofibers in the mixture of RADA16-I peptide and functionalized peptides. The behaviors of hASCs cultured in 3-D peptide hydrogels, including migration, proliferation and growth factor-secretion ability, were studied. Our results showed that the functionalized peptide hydrogels were suitable 3-D scaffolds for hASC growth with higher cell proliferation, migration and the secretion of angiogenic growth factors compared with tissue culture plates and pure RADA16-I scaffolds. The present study suggests that these functionalized designer peptide hydrogels not only have promising applications for diverse tissue engineering and regenerative medicine applications as stem cell delivery vehicles, but also could be a biomimetic 3-D system to study nanobiomaterial-stem cell interactions and to direct stem cell behaviors.
Collapse
|
46
|
Bábíčková J, Tóthová Ľ, Boor P, Celec P. In vivo phage display--a discovery tool in molecular biomedicine. Biotechnol Adv 2013; 31:1247-59. [PMID: 23623852 DOI: 10.1016/j.biotechadv.2013.04.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/15/2013] [Accepted: 04/15/2013] [Indexed: 12/13/2022]
Abstract
In vivo phage display is a high-throughput method for identifying target ligands specific for different vascular beds. Targeting is possible due to the heterogeneous expression of receptors and other antigens in a particular vascular bed. Such expression is additionally influenced by the physiological or pathological status of the vasculature. In vivo phage display represents a technique that is usable in both, vascular mapping and targeted drug development. In this review, several important methodological aspects of in vivo phage display experiments are discussed. These include choosing an appropriate phage library, an appropriate animal model and the route of phage library administration. In addition, peptides or antibodies identified by in vivo phage display homing to specific types of vascular beds, including the altered vasculature present in several types of diseases are summarized. Still, confirmation in independent experiments and reproduction of identified sequences are needed for enhancing the clinical applicability of in vivo phage display research.
Collapse
Affiliation(s)
- Janka Bábíčková
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia; Division of Nephrology, RWTH University, Aachen, Germany
| | | | | | | |
Collapse
|
47
|
Reetz J, Herchenröder O, Schmidt A, Pützer BM. Vector Technology and Cell Targeting: Peptide-Tagged Adenoviral Vectors as a Powerful Tool for Cell Specific Targeting. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
48
|
Saracino GAA, Cigognini D, Silva D, Caprini A, Gelain F. Nanomaterials design and tests for neural tissue engineering. Chem Soc Rev 2012; 42:225-62. [PMID: 22990473 DOI: 10.1039/c2cs35065c] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanostructured scaffolds recently showed great promise in tissue engineering: nanomaterials can be tailored at the molecular level and scaffold morphology may more closely resemble features of extracellular matrix components in terms of porosity, framing and biofunctionalities. As a consequence, both biomechanical properties of scaffold microenvironments and biomaterial-protein interactions can be tuned, allowing for improved transplanted cell engraftment and better controlled diffusion of drugs. Easier said than done, a nanotech-based regenerative approach encompasses different fields of know-how, ranging from in silico simulations, nanomaterial synthesis and characterization at the nano-, micro- and mesoscales to random library screening methods (e.g. phage display), in vitro cellular-based experiments and validation in animal models of the target injury. All of these steps of the "assembly line" of nanostructured scaffolds are tightly interconnected both in their standard analysis techniques and in their most recent breakthroughs: indeed their efforts have to jointly provide the deepest possible analyses of the diverse facets of the challenging field of neural tissue engineering. The purpose of this review is therefore to provide a critical overview of the recent advances in and drawbacks and potential of each mentioned field, contributing to the realization of effective nanotech-based therapies for the regeneration of peripheral nerve transections, spinal cord injuries and brain traumatic injuries. Far from being the ultimate overview of such a number of topics, the reader will acknowledge the intrinsic complexity of the goal of nanotech tissue engineering for a conscious approach to the development of a regenerative therapy and, by deciphering the thread connecting all steps of the research, will gain the necessary view of its tremendous potential if each piece of stone is correctly placed to work synergically in this impressive mosaic.
Collapse
Affiliation(s)
- Gloria A A Saracino
- Center for Nanomedicine and Tissue Engineering, A.O. Ospedale Niguarda Cà Granda, Milan, 20162, Italy
| | | | | | | | | |
Collapse
|
49
|
Sun L, Zhao X. A self-assembling peptide RADA16-I integrated with spider fibroin uncrystalline motifs. Int J Nanomedicine 2012; 7:571-80. [PMID: 22346352 PMCID: PMC3277437 DOI: 10.2147/ijn.s27428] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mechanical strength of nanofiber scaffolds formed by the self-assembling peptide RADA16-I or its derivatives is not very good and limits their application. To address this problem, we inserted spidroin uncrystalline motifs, which confer incomparable elasticity and hydrophobicity to spider silk GGAGGS or GPGGY, into the C-terminus of RADA16-I to newly design two peptides: R3 (n-RADARADARADARADA-GGAGGS-c) and R4 (n-RADARADARADARADA-GPGGY-c), and then observed the effect of these motifs on biophysical properties of the peptide. Atomic force microscopy, transmitting electron microscopy, and circular dichroism spectroscopy confirm that R3 and R4 display β-sheet structure and self-assemble into long nanofibers. Compared with R3, the β-sheet structure and nanofibers formed by R4 are more stable; they change to random coil and unordered aggregation at higher temperature. Rheology measurements indicate that novel peptides form hydrogel when induced by DMEM, and the storage modulus of R3 and R4 hydrogel is 0.5 times and 3 times higher than that of RADA16-I, respectively. Furthermore, R4 hydrogel remarkably promotes growth of liver cell L02 and liver cancer cell SMCC7721 compared with 2D culture, determined by MTT assay. Novel peptides still have potential as hydrophobic drug carriers; they can stabilize pyrene microcrystals in aqueous solution and deliver this into a lipophilic environment, identified by fluorescence emission spectra. Altogether, the spider fibroin motif GPGGY most effectively enhances mechanical strength and hydrophobicity of the peptide. This study provides a new method in the design of nanobiomaterials and helps us to understand the role of the amino acid sequence in nanofiber formation.
Collapse
Affiliation(s)
- Lijuan Sun
- West China Hospital Laboratory of Nanomedicine and Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, Chengdu 610041, Sichuan, China
| | | |
Collapse
|
50
|
Cigognini D, Satta A, Colleoni B, Silva D, Donegà M, Antonini S, Gelain F. Evaluation of early and late effects into the acute spinal cord injury of an injectable functionalized self-assembling scaffold. PLoS One 2011; 6:e19782. [PMID: 21611127 PMCID: PMC3097206 DOI: 10.1371/journal.pone.0019782] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 04/04/2011] [Indexed: 12/03/2022] Open
Abstract
The complex physiopathological events occurring after spinal cord injury (SCI) make this devastating trauma still incurable. Self-assembling peptides (SAPs) are nanomaterials displaying some appealing properties for application in regenerative medicine because they mimic the structure of the extra-cellular matrix (ECM), are reabsorbable, allow biofunctionalizations and can be injected directly into the lesion. In this study we evaluated the putative neurorigenerative properties of RADA16-4G-BMHP1 SAP, proved to enhance in vitro neural stem cells survival and differentiation. This SAP (RADA16-I) has been functionalized with a bone marrow homing motif (BMHP1) and optimized via the insertion of a 4-glycine-spacer that ameliorates scaffold stability and exposure of the biomotifs. We injected the scaffold immediately after contusion in the rat spinal cord, then we evaluated the early effects by semi-quantitative RT-PCR and the late effects by histological analysis. Locomotor recovery over 8 weeks was assessed using Basso, Beattie, Bresnahan (BBB) test. Gene expression analysis showed that at 7 days after lesion the functionalized SAP induced a general upregulation of GAP-43, trophic factors and ECM remodelling proteins, whereas 3 days after SCI no remarkable changes were observed. Hystological analysis revealed that 8 weeks after SCI our scaffold increased cellular infiltration, basement membrane deposition and axon regeneration/sprouting within the cyst. Moreover the functionalized SAP showed to be compatible with the surrounding nervous tissue and to at least partially fill the cavities. Finally SAP injection resulted in a statistically significant improvement of both hindlimbs' motor performance and forelimbs-hindlimbs coordination. Altogether, these results indicate that RADA16-4G-BMHP1 induced favourable reparative processes, such as matrix remodelling, and provided a physical and trophic support to nervous tissue ingrowth. Thus this biomaterial, eventually combined with cells and growth factors, may constitute a promising biomimetic scaffold for regenerative applications in the injured central nervous system.
Collapse
Affiliation(s)
- Daniela Cigognini
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
- Center for Nanomedicine and Tissue Engineering, Niguarda Ca' Granda Hospital, Milano, Italy
| | - Alessandro Satta
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
| | - Bianca Colleoni
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
| | - Diego Silva
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
- Center for Nanomedicine and Tissue Engineering, Niguarda Ca' Granda Hospital, Milano, Italy
| | - Matteo Donegà
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
| | - Stefania Antonini
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
- Center for Nanomedicine and Tissue Engineering, Niguarda Ca' Granda Hospital, Milano, Italy
| | - Fabrizio Gelain
- Biotechnology and Biosciences Department, University of Milano-Bicocca, Milano, Italy
- Center for Nanomedicine and Tissue Engineering, Niguarda Ca' Granda Hospital, Milano, Italy
- Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
- * E-mail:
| |
Collapse
|