1
|
Li J, Zhang X, Peng ZX, Chen JH, Liang JH, Ke LQ, Huang D, Cheng WX, Lin S, Li G, Hou R, Zhong WZ, Lin ZJ, Qin L, Chen GQ, Zhang P. Metabolically activated energetic materials mediate cellular anabolism for bone regeneration. Trends Biotechnol 2024; 42:1745-1776. [PMID: 39237385 DOI: 10.1016/j.tibtech.2024.08.002] [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: 03/20/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
The understanding of cellular energy metabolism activation by engineered scaffolds remains limited, posing challenges for therapeutic applications in tissue regeneration. This study presents biosynthesized poly(3-hydroxybutyrate-co-4-hydroxybutyrate) [P(3HB-co-4HB)] and its major degradation product, 3-hydroxybutyrate (3HB), as endogenous bioenergetic fuels that augment cellular anabolism, thereby facilitating the progression of human bone marrow-derived mesenchymal stem cells (hBMSCs) towards osteoblastogenesis. Our research demonstrated that 3HB markedly boosts in vitro ATP production, elevating mitochondrial membrane potential and capillary-like tube formation. Additionally, it raises citrate levels in the tricarboxylic acid (TCA) cycle, facilitating the synthesis of citrate-containing apatite during hBMSCs osteogenesis. Furthermore, 3HB administration significantly increased bone mass in rats with osteoporosis induced by ovariectomy. The findings also showed that P(3HB-co-4HB) scaffold substantially enhances long-term vascularized bone regeneration in rat cranial defect models. These findings reveal a previously unknown role of 3HB in promoting osteogenesis of hBMSCs and highlight the metabolic activation of P(3HB-co-4HB) scaffold for bone regeneration.
Collapse
Affiliation(s)
- Jian Li
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China.
| | - Xu Zhang
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Zi-Xin Peng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jian-Hai Chen
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jian-Hui Liang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Li-Qing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Dan Huang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Wen-Xiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rui Hou
- Nam Yue Natural Medicine Co., Ltd., Macau, China
| | | | - Zheng-Jie Lin
- Department of Stomatology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, 518067, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Guo-Qiang Chen
- School of Life Sciences, Center of Synthetic and Systems Biology, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
2
|
Ozcicek I, Aysit N, Balcikanli Z, Ayturk NU, Aydeger A, Baydas G, Aydin MS, Altintas E, Erim UC. Development of BDNF/NGF/IKVAV Peptide Modified and Gold Nanoparticle Conductive PCL/PLGA Nerve Guidance Conduit for Regeneration of the Rat Spinal Cord Injury. Macromol Biosci 2024; 24:e2300453. [PMID: 38224015 DOI: 10.1002/mabi.202300453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Indexed: 01/16/2024]
Abstract
Spinal cord injuries are very common worldwide, leading to permanent nerve function loss with devastating effects in the affected patients. The challenges and inadequate results in the current clinical treatments are leading scientists to innovative neural regenerative research. Advances in nanoscience and neural tissue engineering have opened new avenues for spinal cord injury (SCI) treatment. In order for designed nerve guidance conduit (NGC) to be functionally useful, it must have ideal scaffold properties and topographic features that promote the linear orientation of damaged axons. In this study, it is aimed to develop channeled polycaprolactone (PCL)/Poly-D,L-lactic-co-glycolic acid (PLGA) hybrid film scaffolds, modify their surfaces by IKVAV pentapeptide/gold nanoparticles (AuNPs) or polypyrrole (PPy) and investigate the behavior of motor neurons on the designed scaffold surfaces in vitro under static/bioreactor conditions. Their potential to promote neural regeneration after implantation into the rat SCI by shaping the film scaffolds modified with neural factors into a tubular form is also examined. It is shown that channeled groups decorated with AuNPs highly promote neurite orientation under bioreactor conditions and also the developed optimal NGC (PCL/PLGA G1-IKVAV/BDNF/NGF-AuNP50) highly regenerates SCI. The results indicate that the designed scaffold can be an ideal candidate for spinal cord regeneration.
Collapse
Affiliation(s)
- Ilyas Ozcicek
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Nese Aysit
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Zeynep Balcikanli
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Nilufer Ulas Ayturk
- Department of Histology and Embryology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Canakkale, 17020, Turkey
| | - Asel Aydeger
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Gulsena Baydas
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Mehmet Serif Aydin
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Esra Altintas
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Umit Can Erim
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Analytical Chemistry, School of Pharmacy, Istanbul Medipol University, Istanbul, 34815, Turkey
| |
Collapse
|
3
|
Du J, Liu X, Marasini S, Wang Z, Dammen-Brower K, Yarema KJ, Jia X. Metabolically Glycoengineered Neural Stem Cells Boost Neural Repair After Cardiac Arrest. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309866. [PMID: 39071865 PMCID: PMC11281434 DOI: 10.1002/adfm.202309866] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Indexed: 07/30/2024]
Abstract
Cardiac arrest (CA)-induced cerebral ischemia remains challenging with high mortality and disability. Neural stem cell (NSC) engrafting is an emerging therapeutic strategy with considerable promise that, unfortunately, is severely compromised by limited cell functionality after in vivo transplantation. This groundbreaking report demonstrates that metabolic glycoengineering (MGE) using the "Ac5ManNTProp (TProp)" monosaccharide analog stimulates the Wnt/β-catenin pathway, improves cell adhesion, and enhances neuronal differentiation in human NSCs in vitro thereby substantially increasing the therapeutic potential of these cells. For the first time, MGE significantly enhances NSC efficacy for treating ischemic brain injury after asphyxia CA in rats. In particular, neurological deficit scores and neurobehavioral tests experience greater improvements when the therapeutic cells are pretreated with TProp than with "stand-alone" NSC therapy. Notably, the TProp-NSC group exhibits significantly stronger neuroprotective functions including enhanced differentiation, synaptic plasticity, and reduced microglia recruitment; furthermore, Wnt pathway agonists and inhibitors demonstrate a pivotal role for Wnt signaling in the process. These findings help establish MGE as a promising avenue for addressing current limitations associated with NSC transplantation via beneficially influencing neural regeneration and synaptic plasticity, thereby offering enhanced therapeutic options to boost brain recovery following global ischemia.
Collapse
Affiliation(s)
- Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiao Liu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Subash Marasini
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Zhuoran Wang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kris Dammen-Brower
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Kevin J. Yarema
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
4
|
Shlapakova LE, Surmeneva MA, Kholkin AL, Surmenev RA. Revealing an important role of piezoelectric polymers in nervous-tissue regeneration: A review. Mater Today Bio 2024; 25:100950. [PMID: 38318479 PMCID: PMC10840125 DOI: 10.1016/j.mtbio.2024.100950] [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: 10/05/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nerve injuries pose a drastic threat to nerve mobility and sensitivity and lead to permanent dysfunction due to low regenerative capacity of mature neurons. The electrical stimuli that can be provided by electroactive materials are some of the most effective tools for the formation of soft tissues, including nerves. Electric output can provide a distinctly favorable bioelectrical microenvironment, which is especially relevant for the nervous system. Piezoelectric biomaterials have attracted attention in the field of neural tissue engineering owing to their biocompatibility and ability to generate piezoelectric surface charges. In this review, an outlook of the most recent achievements in the field of piezoelectric biomaterials is described with an emphasis on piezoelectric polymers for neural tissue engineering. First, general recommendations for the design of an optimal nerve scaffold are discussed. Then, specific mechanisms determining nerve regeneration via piezoelectric stimulation are considered. Activation of piezoelectric responses via natural body movements, ultrasound, and magnetic fillers is also examined. The use of magnetoelectric materials in combination with alternating magnetic fields is thought to be the most promising due to controllable reproducible cyclic deformations and deep tissue permeation by magnetic fields without tissue heating. In vitro and in vivo applications of nerve guidance scaffolds and conduits made of various piezopolymers are reviewed too. Finally, challenges and prospective research directions regarding piezoelectric biomaterials promoting nerve regeneration are discussed. Thus, the most relevant scientific findings and strategies in neural tissue engineering are described here, and this review may serve as a guideline both for researchers and clinicians.
Collapse
Affiliation(s)
- Lada E. Shlapakova
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| | - Andrei L. Kholkin
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| |
Collapse
|
5
|
Okhovatian S, Shakeri A, Huyer LD, Radisic M. Elastomeric Polyesters in Cardiovascular Tissue Engineering and Organs-on-a-Chip. Biomacromolecules 2023; 24:4511-4531. [PMID: 37639715 PMCID: PMC10915885 DOI: 10.1021/acs.biomac.3c00387] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cardiovascular tissue constructs provide unique design requirements due to their functional responses to substrate mechanical properties and cyclic stretching behavior of cardiac tissue that requires the use of durable elastic materials. Given the diversity of polyester synthesis approaches, an opportunity exists to develop a new class of biocompatible, elastic, and immunomodulatory cardiovascular polymers. Furthermore, elastomeric polyester materials have the capability to provide tailored biomechanical synergy with native tissue and hence reduce inflammatory response in vivo and better support tissue maturation in vitro. In this review, we highlight underlying chemistry and design strategies of polyester elastomers optimized for cardiac tissue scaffolds. The major advantages of these materials such as their tunable elasticity, desirable biodegradation, and potential for incorporation of bioactive compounds are further expanded. Unique fabrication methods using polyester materials such as micromolding, 3D stamping, electrospinning, laser ablation, and 3D printing are discussed. Moreover, applications of these biomaterials in cardiovascular organ-on-a-chip devices and patches are analyzed. Finally, we outline unaddressed challenges in the field that need further study to enable the impactful translation of soft polyesters to clinical applications.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Locke Davenport Huyer
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
6
|
Aydeger A, Aysit N, Baydas G, Cakici C, Erim UC, Arpa MD, Ozcicek I. Design of IKVAV peptide/gold nanoparticle decorated, micro/nano-channeled PCL/PLGA film scaffolds for neuronal differentiation and neurite outgrowth. BIOMATERIALS ADVANCES 2023; 152:213472. [PMID: 37301056 DOI: 10.1016/j.bioadv.2023.213472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023]
Abstract
In the field of neural tissue engineering, intensive efforts are being made to develop tissue scaffolds that can support an effective functional recovery and neural development by guiding damaged axons and neurites. Micro/nano-channeled conductive biomaterials are considered a promising approach for repairing the injured neural tissues. Many studies have demonstrated that the micro/nano-channels and aligned nanofibers could guide the neurites to extend along the direction of alignment. However, an ideal biocompatible scaffold containing conductive arrays that could promote effective neural stem cell differentiation and development, and also stimulate high neurite guidance has not been fully developed. In the current study, we aimed to fabricate micro/nano-channeled polycaprolactone (PCL)/Poly-d,l-lactic-co-glycolic acid (PLGA) hybrid film scaffolds, decorate their surfaces with IKVAV pentapeptide/gold nanoparticles (AuNPs), and investigate the behavior of PC12 cells and neural stem cells (NSCs) on the developed biomaterial under static/bioreactor conditions. Here we show that channeled groups decorated with AuNPs highly promote neurite outgrowth and neuronal differentiation along linear lines in the presence of electrical stimulation, compared with the polypyrrole (PPy) coating, which has been used traditionally for many years. Hopefully, this newly developed channeled scaffold structure (PCL/PLGA-AuNPs-IKVAV) could help to support long-distance axonal regeneration and neuronal development after different neural damages.
Collapse
Affiliation(s)
- Asel Aydeger
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Nese Aysit
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Gulsena Baydas
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey; Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Cagri Cakici
- Department of Medical Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Umit Can Erim
- Department of Analytical Chemistry, School of Pharmacy, Istanbul Medipol University, Istanbul, Turkey
| | - Muhammet Davut Arpa
- Department of Pharmaceutical Technology, School of Pharmacy, Istanbul Medipol University, Istanbul, Turkey
| | - Ilyas Ozcicek
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
7
|
Li J, Zhang X, Udduttula A, Fan ZS, Chen JH, Sun AR, Zhang P. Microbial-Derived Polyhydroxyalkanoate-Based Scaffolds for Bone Tissue Engineering: Biosynthesis, Properties, and Perspectives. Front Bioeng Biotechnol 2022; 9:763031. [PMID: 34993185 PMCID: PMC8724543 DOI: 10.3389/fbioe.2021.763031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/17/2021] [Indexed: 01/15/2023] Open
Abstract
Polyhydroxyalkanoates (PHAs) are a class of structurally diverse natural biopolyesters, synthesized by various microbes under unbalanced culture conditions. PHAs as biomedical materials have been fabricated in various forms to apply to tissue engineering for the past years due to their excellent biodegradability, inherent biocompatibility, modifiable mechanical properties, and thermo-processability. However, there remain some bottlenecks in terms of PHA production on a large scale, the purification process, mechanical properties, and biodegradability of PHA, which need to be further resolved. Therefore, scientists are making great efforts via synthetic biology and metabolic engineering tools to improve the properties and the product yields of PHA at a lower cost for the development of various PHA-based scaffold fabrication technologies to widen biomedical applications, especially in bone tissue engineering. This review aims to outline the biosynthesis, structures, properties, and the bone tissue engineering applications of PHA scaffolds with different manufacturing technologies. The latest advances will provide an insight into future outlooks in PHA-based scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Jian Li
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xu Zhang
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China.,Department of Chemical Engineering, Tsinghua University, Beijing, China
| | - Anjaneyulu Udduttula
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhi Shan Fan
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jian Hai Chen
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Antonia RuJia Sun
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Peng Zhang
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
8
|
Blanco FG, Hernández N, Rivero-Buceta V, Maestro B, Sanz JM, Mato A, Hernández-Arriaga AM, Prieto MA. From Residues to Added-Value Bacterial Biopolymers as Nanomaterials for Biomedical Applications. NANOMATERIALS 2021; 11:nano11061492. [PMID: 34200068 PMCID: PMC8228158 DOI: 10.3390/nano11061492] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022]
Abstract
Bacterial biopolymers are naturally occurring materials comprising a wide range of molecules with diverse chemical structures that can be produced from renewable sources following the principles of the circular economy. Over the last decades, they have gained substantial interest in the biomedical field as drug nanocarriers, implantable material coatings, and tissue-regeneration scaffolds or membranes due to their inherent biocompatibility, biodegradability into nonhazardous disintegration products, and their mechanical properties, which are similar to those of human tissues. The present review focuses upon three technologically advanced bacterial biopolymers, namely, bacterial cellulose (BC), polyhydroxyalkanoates (PHA), and γ-polyglutamic acid (PGA), as models of different carbon-backbone structures (polysaccharides, polyesters, and polyamides) produced by bacteria that are suitable for biomedical applications in nanoscale systems. This selection models evidence of the wide versatility of microorganisms to generate biopolymers by diverse metabolic strategies. We highlight the suitability for applied sustainable bioprocesses for the production of BC, PHA, and PGA based on renewable carbon sources and the singularity of each process driven by bacterial machinery. The inherent properties of each polymer can be fine-tuned by means of chemical and biotechnological approaches, such as metabolic engineering and peptide functionalization, to further expand their structural diversity and their applicability as nanomaterials in biomedicine.
Collapse
Affiliation(s)
- Francisco G. Blanco
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
| | - Natalia Hernández
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
| | - Virginia Rivero-Buceta
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
| | - Beatriz Maestro
- Host-Parasite Interplay in Pneumococcal Infection Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain; (B.M.); (J.M.S.)
| | - Jesús M. Sanz
- Host-Parasite Interplay in Pneumococcal Infection Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain; (B.M.); (J.M.S.)
| | - Aránzazu Mato
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
| | - Ana M. Hernández-Arriaga
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
| | - M. Auxiliadora Prieto
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), 28040 Madrid, Spain; (F.G.B.); (N.H.); (V.R.-B.); (A.M.); (A.M.H.-A.)
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Biological Research Centre Margarita Salas, CIB-CSIC, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
9
|
Mohandas SP, Balan L, Gopi J, Anoop BS, Mohan P S, Philip R, Cubelio SS, Singh ISB. Biocompatibility of polyhydroxybutyrate-co-hydroxyvalerate films generated from Bacillus cereus MCCB 281 for medical applications. Int J Biol Macromol 2021; 176:244-252. [PMID: 33548322 DOI: 10.1016/j.ijbiomac.2021.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 01/13/2023]
Abstract
Polyhydroxyalkanoates (PHAs) are natural polyesters produced by microorganisms as a source of intracellular energy reserves. These polymers have been extensively studied for tissue engineering and drug delivery applications due to their desirable material properties. Solvent-cast film of poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), produced by Bacillus cereus MCCB 281 was characterized to study the surface morphology, roughness, thermal and mechanical properties. PHBV films were slightly hydrophilic with an average surface roughness of 43.66 nm. In vitro cell viability and proliferation studies on PHBV film surface investigated using L929 fibroblasts showed good cell attachment and proliferation. Hemocompatibility of PHBV evaluated by hemolysis assay, in vitro platelet adhesion and coagulation assays demonstrated good blood compatibility for use as blood contact graft materials. Therefore, PHBV produced from the marine bacterium favoured cellular growth of L929 fibroblasts indicating its potential to be used as a biomaterial substrate for cell adhesion in tissue engineering and medical applications.
Collapse
Affiliation(s)
- Sowmya P Mohandas
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India
| | - Linu Balan
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India
| | - Jayanath Gopi
- Dept. of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India
| | - B S Anoop
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India
| | - Sooraj Mohan P
- Department of Environmental Engineering, National I-Lan University, Taiwan
| | - Rosamma Philip
- Dept. of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India
| | | | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, India.
| |
Collapse
|
10
|
Zhou N, Ma X, Hu W, Ren P, Zhao Y, Zhang T. Effect of RGD content in poly(ethylene glycol)-crosslinked poly(methyl vinyl ether-alt-maleic acid) hydrogels on the expansion of ovarian cancer stem-like cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111477. [PMID: 33255056 DOI: 10.1016/j.msec.2020.111477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022]
Abstract
The extracellular matrix (ECM) affects cell behaviors, such as survival, proliferation, motility, invasion, and differentiation. The arginine-glycine-aspartic acid (RGD) sequence is present in several ECM proteins, such as fibronectin, collagen type I, fibrinogen, laminin, vitronectin, and osteopontin. It is very critical to develop ECM-like substrates with well-controlled features for the investigation of influence of RGD on the behavior of tumor cells. In this study, poly(ethylene glycol) (PEG)-crosslinked poly(methyl vinyl ether-alt-maleic acid) (P(MVE-alt-MA)) hydrogels (PEMM) with different RGD contents were synthesized, fully characterized, and established as in vitro culture platforms to investigate the effects of RGD content on cancer stem cell (CSC) enrichment. The morphology, proliferation, and viability of SK-OV-3 ovarian cancer cells cultured on hydrogels with different RGD contents, the expression of CSC markers and malignant signaling pathway-related genes, and drug resistance were systematically evaluated. The cell aggregates formed on the hydrogel surface with a lower RGD content acquired certain CSC-like properties, thus drug resistance was enhanced. In contrast, the drug sensitivity of cells on the higher RGD content surface increased because of less CSC-like properties. However, the presence of RGD in the stiff hydrogels (PEMM2) had less effect on the stemness expression than did its presence in the soft hydrogels (PEMM1). The results suggest that RGD content and matrix stiffness can lead to synergetic effects on the expression of cancer cell stemness and the epithelial-mesenchymal transition (EMT), interleukin-6 (IL-6), and Wnt pathways.
Collapse
Affiliation(s)
- Naizhen Zhou
- State Key Lab of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaoe Ma
- State Key Lab of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Wanjun Hu
- State Key Lab of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Pengfei Ren
- State Key Lab of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Youliang Zhao
- Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Tianzhu Zhang
- State Key Lab of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
11
|
Mato A, Blanco FG, Maestro B, Sanz JM, Pérez-Gil J, Prieto MA. Dissecting the Polyhydroxyalkanoate-Binding Domain of the PhaF Phasin: Rational Design of a Minimized Affinity Tag. Appl Environ Microbiol 2020; 86:e00570-20. [PMID: 32303541 PMCID: PMC7267194 DOI: 10.1128/aem.00570-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022] Open
Abstract
Phasin PhaF from Pseudomonas putida consists of a modular protein whose N-terminal domain (BioF) has been demonstrated to be responsible for binding to the polyhydroxyalkanoate (PHA) granule. BioF has been exploited for biotechnological purposes as an affinity tag in the functionalization of PHA beads with fusion proteins both in vivo and in vitro The structural model of this domain suggests an amphipathic α-helical conformation with the hydrophobic residues facing the PHA granule. In this work, we analyzed the mean hydrophobicity and the hydrophobic moment of the native BioF tag to rationally design shorter versions that maintain affinity for the granule. Hybrid proteins containing the green fluorescent protein (GFP) fused to the BioF derivatives were studied for in vivo localization on PHA, stability on the surface of the PHA granule against pH, temperature, and ionic strength, and their possible influence on PHA synthesis. Based on the results obtained, a minimized BioF tag for PHA functionalization has been proposed (MinP) that retains similar binding properties but possesses an attractive biotechnological potential derived from its reduced size. The MinP tag was further validated by analyzing the functionality and stability of the fusion proteins MinP-β-galactosidase and MinP-CueO from Escherichia coliIMPORTANCE Polyhydroxyalkanoates (PHAs) are biocompatible, nontoxic, and biodegradable biopolymers with exceptional applications in the industrial and medical fields. The complex structure of the PHA granule can be exploited as a toolbox to display molecules of interest on their surface. Phasins, the most abundant group of proteins on the granule, have been employed as anchoring tags to obtain functionalized PHA beads for high-affinity bioseparation, enzyme immobilization, diagnostics, or cell targeting. Here, a shorter module based on the previously designed BioF tag has been demonstrated to maintain the affinity for the PHA granule, with higher stability and similar functionalization efficiency. The use of a 67% shorter peptide, which maintains the binding properties of the entire protein, constitutes an advantage for the immobilization of recombinant proteins on the PHA surface both in vitro and in vivo.
Collapse
Affiliation(s)
- Aranzazu Mato
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), Madrid, Spain
| | - Francisco G Blanco
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), Madrid, Spain
| | - Beatriz Maestro
- Host-Parasite Interplay in Pneumococcal Infection Group, Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Jesús M Sanz
- Host-Parasite Interplay in Pneumococcal Infection Group, Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Jesús Pérez-Gil
- Biochemical and Molecular Biology Department, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Madrid, Spain
| | - M Auxiliadora Prieto
- Polymer Biotechnology Group, Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Interdisciplinary Platform for Sustainable Plastics towards a Circular Economy-Spanish National Research Council (SusPlast-CSIC), Madrid, Spain
| |
Collapse
|
12
|
Liu D, Pavathuparambil Abdul Manaph N, Al-Hawwas M, Bobrovskaya L, Xiong LL, Zhou XF. Coating Materials for Neural Stem/Progenitor Cell Culture and Differentiation. Stem Cells Dev 2020; 29:463-474. [PMID: 32106778 DOI: 10.1089/scd.2019.0288] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) have a potential to treat various neurological diseases, such as Parkinson's Disease, Alzheimer's Disease, and Spinal Cord Injury. However, the limitation of NSPC sources and the difficulty to maintain their stemness or to differentiate them into specific therapeutic cells are the main hurdles for clinical research and application. Thus, for obtaining a therapeutically relevant number of NSPCs in vitro, it is important to understand factors regulating their behaviors and to establish a protocol for stable NSPC proliferation and differentiation. Coating materials for cell culture, such as Matrigel, laminin, collagen, and other coating materials, can significantly affect NSPC characteristics. This article provides a review of coating materials for NSPC culturing in both two dimensions and three dimensions, and their functions in NSPC proliferation and differentiation, and presents a useful guide to select coating materials for researchers.
Collapse
Affiliation(s)
- Donghui Liu
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | | | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Liu-Lin Xiong
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
13
|
Ghane N, Beigi MH, Labbaf S, Nasr-Esfahani MH, Kiani A. Design of hydrogel-based scaffolds for the treatment of spinal cord injuries. J Mater Chem B 2020; 8:10712-10738. [DOI: 10.1039/d0tb01842b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hydrogel-based scaffold design approaches for the treatment of spinal cord injuries.
Collapse
Affiliation(s)
- Nazanin Ghane
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Mohammad-Hossein Beigi
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Sheyda Labbaf
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | | | - Amirkianoosh Kiani
- Silicon Hall: Micro/Nano Manufacturing Facility
- Faculty of Engineering and Applied Science
- Ontario Tech University
- Ontario
- Canada
| |
Collapse
|
14
|
Voinova V, Bonartseva G, Bonartsev A. Effect of poly(3-hydroxyalkanoates) as natural polymers on mesenchymal stem cells. World J Stem Cells 2019; 11:764-786. [PMID: 31692924 PMCID: PMC6828591 DOI: 10.4252/wjsc.v11.i10.764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/17/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are stromal multipotent stem cells that can differentiate into multiple cell types, including fibroblasts, osteoblasts, chondrocytes, adipocytes, and myoblasts, thus allowing them to contribute to the regeneration of various tissues, especially bone tissue. MSCs are now considered one of the most promising cell types in the field of tissue engineering. Traditional petri dish-based culture of MSCs generate heterogeneity, which leads to inconsistent efficacy of MSC applications. Biodegradable and biocompatible polymers, poly(3-hydroxyalkanoates) (PHAs), are actively used for the manufacture of scaffolds that serve as carriers for MSC growth. The growth and differentiation of MSCs grown on PHA scaffolds depend on the physicochemical properties of the polymers, the 3D and surface microstructure of the scaffolds, and the biological activity of PHAs, which was discovered in a series of investigations. The mechanisms of the biological activity of PHAs in relation to MSCs remain insufficiently studied. We suggest that this effect on MSCs could be associated with the natural properties of bacteria-derived PHAs, especially the most widespread representative poly(3-hydroxybutyrate) (PHB). This biopolymer is present in the bacteria of mammalian microbiota, whereas endogenous poly(3-hydroxybutyrate) is found in mammalian tissues. The possible association of PHA effects on MSCs with various biological functions of poly(3-hydroxybutyrate) in bacteria and eukaryotes, including in humans, is discussed in this paper.
Collapse
Affiliation(s)
- Vera Voinova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
| | - Garina Bonartseva
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia
| | - Anton Bonartsev
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia
| |
Collapse
|
15
|
Jung HR, Jeon JM, Yi DH, Song HS, Yang SY, Choi TR, Bhatia SK, Yoon JJ, Kim YG, Brigham CJ, Yang YH. Poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) terpolymer production from volatile fatty acids using engineered Ralstonia eutropha. Int J Biol Macromol 2019; 138:370-378. [DOI: 10.1016/j.ijbiomac.2019.07.091] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 01/05/2023]
|
16
|
Tarazona NA, Machatschek R, Schulz B, Prieto MA, Lendlein A. Molecular Insights into the Physical Adsorption of Amphiphilic Protein PhaF onto Copolyester Surfaces. Biomacromolecules 2019; 20:3242-3252. [DOI: 10.1021/acs.biomac.9b00069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Natalia A. Tarazona
- Institute of Biomaterial
Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513 Teltow, Germany
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Rainhard Machatschek
- Institute of Biomaterial
Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513 Teltow, Germany
| | - Burkhard Schulz
- Institute of Biomaterial
Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14469 Potsdam, Germany
| | - M. Auxiliadora Prieto
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Andreas Lendlein
- Institute of Biomaterial
Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14469 Potsdam, Germany
| |
Collapse
|
17
|
Luo D, Ruan S, Liu A, Kong X, Lee IS, Chen C. Laminin functionalized biomimetic apatite to regulate the adhesion and proliferation behaviors of neural stem cells. Int J Nanomedicine 2018; 13:6223-6233. [PMID: 30349246 PMCID: PMC6188167 DOI: 10.2147/ijn.s176596] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Functionalizing biomaterial substrates with biological signals shows promise in regulating neural stem cell (NSC) behaviors through mimicking cellular microenvironment. However, diverse methods for immobilizing biological molecules yields promising results but with many problems. Biomimetic apatite is an excellent carrier due to its non-toxicity, good biocompatibility, biodegradability, and favorable affinity to plenty of molecules. Therefore, it may provide a promising alternative in regulating NSC behaviors. Methods Biomimetic apatite immobilized with the extracellular protein - laminin (LN) was prepared through coprecipitation process in modified Dulbecco's phosphate-buffered saline (DPBS) containing LN. The amount of coprecipitated LN and their release kinetics were examined. The adhesion and proliferation behaviors of NSC on biomimetic apatite immobilized with LN were investigated. Results The coprecipitation approach provided well retention of LN within biomimetic apatite up to 28 days, and supported the adhesion and proliferation of NSCs without cytotoxicity. For long-term cultivation, NSCs formed neurosphere-like aggregates on non-functionalized biomimetic apatite. A monolayer of proliferated NSCs on biomimetic apatite with coprecipitated LN was observed and even more stable than the positive control of LN coated tissue-culture treated polystyrene (TCP). Conclusion The simple and reproducible method of coprecipitation suggests that biomimetic apatite is an ideal carrier to functionalize materials with biological molecules for neural-related applications.
Collapse
Affiliation(s)
- Dandan Luo
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - Shichao Ruan
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - Aiping Liu
- Center for Optoelectronics Materials and Devices, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Xiangdong Kong
- College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - In-Seop Lee
- College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China, .,Institute of Natural Sciences, Yonsei University, Seoul 03722, Korea,
| | - Cen Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China, .,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, People's Republic of China,
| |
Collapse
|
18
|
Chen WS, Guo LY, Masroujeh AM, Augustine AM, Tsai CK, Chin TY, Chen-Yang YW, Yang ML. A Single-Step Surface Modification of Electrospun Silica Nanofibers Using a Silica Binding Protein Fused with an RGD Motif for Enhanced PC12 Cell Growth and Differentiation. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E927. [PMID: 29848981 PMCID: PMC6024934 DOI: 10.3390/ma11060927] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/09/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022]
Abstract
In this study, a previously known high-affinity silica binding protein (SB) was genetically engineered to fuse with an integrin-binding peptide (RGD) to create a recombinant protein (SB-RGD). SB-RGD was successfully expressed in Escherichia coli and purified using silica beads through a simple and fast centrifugation method. A further functionality assay showed that SB-RGD bound to the silica surface with an extremely high affinity that required 2 M MgCl₂ for elution. Through a single-step incubation, the purified SB-RGD proteins were noncovalently coated onto an electrospun silica nanofiber (SNF) substrate to fabricate the SNF-SB-RGD substrate. SNF-SB-RGD was characterized by a combination of scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy, and immunostaining fluorescence microscopy. As PC12 cells were seeded onto the SNF-SB-RGD surface, significantly higher cell viability and longer neurite extensions were observed when compared to those on the control surfaces. These results indicated that SB-RGD could serve as a noncovalent coating biologic to support and promote neuron growth and differentiation on silica-based substrates for neuronal tissue engineering. It also provides proof of concept for the possibility to genetically engineer protein-based signaling molecules to noncovalently modify silica-based substrates as bioinspired material.
Collapse
Affiliation(s)
- Wen Shuo Chen
- Department of Chemistry, Center for Nanotechnology, Center for Biomedical Technology, Chung Yuan Christian University, Chung Li 32023, Taiwan.
| | - Ling Yu Guo
- Department of Chemistry, Center for Nanotechnology, Center for Biomedical Technology, Chung Yuan Christian University, Chung Li 32023, Taiwan.
| | | | | | - Cheng Kang Tsai
- Department of Chemistry, Center for Nanotechnology, Center for Biomedical Technology, Chung Yuan Christian University, Chung Li 32023, Taiwan.
| | - Ting Yu Chin
- Department of Bioscience Technology, Chung Yuan Christian University, Chung Li, 32023, Taiwan.
| | - Yui Whei Chen-Yang
- Department of Chemistry, Center for Nanotechnology, Center for Biomedical Technology, Chung Yuan Christian University, Chung Li 32023, Taiwan.
| | - Mong-Lin Yang
- Department of Science, Concordia University Saint Paul, Saint Paul, MN 55104, USA.
| |
Collapse
|
19
|
Chen GQ, Zhang J. Microbial polyhydroxyalkanoates as medical implant biomaterials. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1-18. [DOI: 10.1080/21691401.2017.1371185] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, Beijing, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Center for Nano and Micro Mechanics, Tsinghua University, Beijing, China
- Department of Chemical Engineering, MOE Key Lab of Industrial Biocatalysis, Tsinghua University, Beijing, China
| | - Junyu Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Zhu L, Wang K, Ma T, Huang L, Xia B, Zhu S, Yang Y, Liu Z, Quan X, Luo K, Kong D, Huang J, Luo Z. Noncovalent Bonding of RGD and YIGSR to an Electrospun Poly(ε-Caprolactone) Conduit through Peptide Self-Assembly to Synergistically Promote Sciatic Nerve Regeneration in Rats. Adv Healthc Mater 2017; 6. [PMID: 28140528 DOI: 10.1002/adhm.201600860] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Indexed: 12/18/2022]
Abstract
The nerve conduit with biofunctionalities can regulate neurite outgrowth, as well as the migration, proliferation, and myelination activity of Schwann cells. In the present study, polycaprolactone (PCL) conduits are coated with Naphthalene-phenylalanine-phenylalanine-glycine-arginine-glycine-aspartic (Nap-FFGRGD) and Naphthalene-phenylalanine-phenylalanine-glycine-cysteine-aspartic-proline-glycine-tyrosine-isoleucine-glycine-serine-arginine (Nap-FFGCDPGYIGSR) by self-assembly. In vitro studies demonstrate that arginine-glycine-aspartic (RGD) and tyrosine-isoleucine-glycine-serine-arginine (YIGSR) are capable of synergistically enhancing the ability of PCL to support the adhesion and proliferation of Schwann cells, as well as increasing neurite outgrowth from dorsal root ganglions explants. This synergistic effect may occur via the activation of both the phosphoinositide 3-kinase/protein kinase B and mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathways. RGD/YIGSR modifications demonstrate beneficial effects across a 15 mm sciatic nerve gap in axonal regeneration and functional recovery. In addition, increased vascularization is observed in the RGD/YIGSR-PCL group, which might contribute to their beneficial effects on nerve regeneration. These findings indicate the potential of the RGD/YIGSR-PCL conduit to promote axonal regeneration and functional recovery, making the RGD/YIGSR-PCL conduit an attractive candidate for the treatment of a critical nerve defect.
Collapse
Affiliation(s)
- Lei Zhu
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive Materials; Ministry of Education; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin); College of Life Science; Nankai University; Tianjin 300071 China
| | - Teng Ma
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Liangliang Huang
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Bing Xia
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Shu Zhu
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Yafeng Yang
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Zhongyang Liu
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Xin Quan
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Kai Luo
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive Materials; Ministry of Education; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin); College of Life Science; Nankai University; Tianjin 300071 China
| | - Jinghui Huang
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Zhuojing Luo
- Institute of Orthopaedics; Xijing Hospital; The Fourth Military Medical University; Xi'an Shaanxi 710032 China
| |
Collapse
|
21
|
Huang L, Wang G. The Effects of Different Factors on the Behavior of Neural Stem Cells. Stem Cells Int 2017; 2017:9497325. [PMID: 29358957 PMCID: PMC5735681 DOI: 10.1155/2017/9497325] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/18/2017] [Indexed: 02/07/2023] Open
Abstract
The repair of central nervous system (CNS) injury has been a worldwide problem in the biomedical field. How to reduce the damage to the CNS and promote the reconstruction of the damaged nervous system structure and function recovery has always been the concern of nerve tissue engineering. Multiple differentiation potentials of neural stem cell (NSC) determine the application value for the repair of the CNS injury. Thus, how to regulate the behavior of NSCs becomes the key to treating the CNS injury. So far, a large number of researchers have devoted themselves to searching for a better way to regulate the behavior of NSCs. This paper summarizes the effects of different factors on the behavior of NSCs in the past 10 years, especially on the proliferation and differentiation of NSCs. The final purpose of this review is to provide a more detailed theoretical basis for the clinical repair of the CNS injury by nerve tissue engineering.
Collapse
Affiliation(s)
- Lixiang Huang
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Gan Wang
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China
| |
Collapse
|
22
|
Lee SY. Applications of Microbial Biopolymers in Display Technology. CONSEQUENCES OF MICROBIAL INTERACTIONS WITH HYDROCARBONS, OILS, AND LIPIDS: PRODUCTION OF FUELS AND CHEMICALS 2017. [PMCID: PMC7123360 DOI: 10.1007/978-3-319-50436-0_377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Microorganisms produce a variety of different polymers such as polyamides, polysaccharides, and polyesters. The polyesters, the polyhydroxyalkanoates (PHAs), are the most extensively studied polymers in regard to their use in display technology. The material properties of bacterial PHAs in combination with their biocompatibility and biodegradability make them attractive substrates for use in display technology applications. By translationally fusing bioactive molecules to a gene encoding a PHA-binding domain, the appropriate functionalization for a given application can be achieved such that the need for chemical immobilization is circumvented. By separately extracting and processing the biopolymer, using it to coat a surface, and then treating this surface with the fusion proteins, surface functionalization for immunodiagnostic microarray or tissue engineering applications can be accomplished. Conversely, by expressing the fusion protein directly in the PHA-producing organisms, one-step production of functionalized beads can be achieved. Such beads have been demonstrated in diverse applications, including fluorescence-activated cell sorting, enzyme-linked immunosorbent assays, microarrays, diagnostic skin test for tuberculosis, vaccines, protein purification, and affinity bioseparation.
Collapse
Affiliation(s)
- Sang Yup Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea (Republic of)
| |
Collapse
|
23
|
Yang R, Xu C, Wang T, Wang Y, Wang J, Quan D, Deng DYB. PTMAc-PEG-PTMAc hydrogel modified by RGDC and hyaluronic acid promotes neural stem cells' survival and differentiation in vitro. RSC Adv 2017. [DOI: 10.1039/c7ra06614g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The enhancement of the biological properties of hydrogels by surface modifying with bioactive molecules is of great significance, especially for the treatment of central nervous system injury by combining engrafted cells.
Collapse
Affiliation(s)
- Ruirui Yang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Caixia Xu
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Tao Wang
- PCFM Lab
- GD HPPC Lab
- Research Center of Engineering and Technology for Functional Biomaterials of Guangdong
- School of Chemistry
- Sun Yat-Sen University
| | - Yuanqi Wang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Jingnan Wang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Daping Quan
- PCFM Lab
- GD HPPC Lab
- Research Center of Engineering and Technology for Functional Biomaterials of Guangdong
- School of Chemistry
- Sun Yat-Sen University
| | - David Y. B. Deng
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| |
Collapse
|
24
|
Enhancement of Neural Stem Cell Survival, Proliferation, Migration, and Differentiation in a Novel Self-Assembly Peptide Nanofibber Scaffold. Mol Neurobiol 2016; 54:8050-8062. [PMID: 27878763 DOI: 10.1007/s12035-016-0295-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/08/2016] [Indexed: 12/31/2022]
Abstract
Considerable efforts have been made to combine biologically active molecules into the self-assembling peptide in order to improve cells growth, survival, and differentiation. In this study, a novel three-dimensional scaffold (RADA4GGSIKVAV; R-GSIK) was designed by adding glycine and serine between RADA4 and IKVAV to promote the strength of the peptide. The cell adhesion, viability, proliferation, migration, and differentiation of rat embryonic neural stem cells (NSCs) in R-GSIK were investigated and compared to laminin-coated, two-dimensional, and Puramatrix cultures. The scanning electron microscopy studies of the R-GSIK showed an open porous structure and a suitable surface area available for cell interaction. R-GSIK promoted the cell adhesion, viability, proliferation, and migration compared to the other cultures. In addition, the R-GSIK enhanced NSCs differentiation into neuronal cells. The NSCs injected in R-GSIK had a lower glial differentiation rate than in the Puramatrix. The results suggest that R-GSIK holds great promise for cell therapies and neuronal tissue repair.
Collapse
|
25
|
Vallejo-Giraldo C, Pugliese E, Larrañaga A, Fernandez-Yague MA, Britton JJ, Trotier A, Tadayyon G, Kelly A, Rago I, Sarasua JR, Dowd E, Quinlan LR, Pandit A, Biggs MJP. Polyhydroxyalkanoate/carbon nanotube nanocomposites: flexible electrically conducting elastomers for neural applications. Nanomedicine (Lond) 2016; 11:2547-63. [DOI: 10.2217/nnm-2016-0075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Medium chain length-polyhydroxyalkanoate/multi-walled carbon nanotube (MWCNTs) nanocomposites with a range of mechanical and electrochemical properties were fabricated via assisted dispersion and solvent casting, and their suitability as neural interface biomaterials was investigated. Materials & methods: Mechanical and electrical properties of medium chain length-polyhydroxyalkanoate/MWCNTs nanocomposite films were evaluated by tensile test and electrical impedance spectroscopy, respectively. Primary rat mesencephalic cells were seeded on the composites and quantitative immunostaining of relevant neural biomarkers, and electrical stimulation studies were performed. Results: Incorporation of MWCNTs to the polymeric matrix modulated the mechanical and electrical properties of resulting composites, and promoted differential cell viability, morphology and function as a function of MWCNT concentration. Conclusion: This study demonstrates the feasibility of a green thermoplastic MWCNTs nanocomposite for potential use in neural interfacing applications.
Collapse
Affiliation(s)
- Catalina Vallejo-Giraldo
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Eugenia Pugliese
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Aitor Larrañaga
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Mining-Metallurgy Engineering & Materials Science & POLYMAT, School of Engineering, University of the Basque Country (UPV/EHU) 480130 Bilbao, Spain
| | - Marc A Fernandez-Yague
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - James J Britton
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Alexandre Trotier
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Ghazal Tadayyon
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Adriona Kelly
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Ilaria Rago
- Department of Physics, University of Trieste, Via Valerio 2-34127, Trieste, Italy
| | - Jose-Ramon Sarasua
- Department of Mining-Metallurgy Engineering & Materials Science & POLYMAT, School of Engineering, University of the Basque Country (UPV/EHU) 480130 Bilbao, Spain
| | - Eilís Dowd
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Physics, University of Trieste, Via Valerio 2-34127, Trieste, Italy
| | - Leo R Quinlan
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Pharmacology, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| | - Manus JP Biggs
- CÚRAM – Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Department of Biomedical Engineering, National University of Ireland, Galway, Ireland
| |
Collapse
|
26
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
27
|
Rana D, Ramasamy K, Leena M, Jiménez C, Campos J, Ibarra P, Haidar ZS, Ramalingam M. Surface functionalization of nanobiomaterials for application in stem cell culture, tissue engineering, and regenerative medicine. Biotechnol Prog 2016; 32:554-67. [DOI: 10.1002/btpr.2262] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/16/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Deepti Rana
- Centre for Stem Cell Research (CSCR); A Unit of Institute for Stem Cell Biology and Regenerative Medicine-Bengaluru, Stem Cell Nanotechnology Lab, Christian Medical College Campus; Vellore 632002 India
| | - Keerthana Ramasamy
- Centre for Stem Cell Research (CSCR); A Unit of Institute for Stem Cell Biology and Regenerative Medicine-Bengaluru, Stem Cell Nanotechnology Lab, Christian Medical College Campus; Vellore 632002 India
| | - Maria Leena
- Dept. of Nanoscience and Technology; Karunya University; Coimbatore 641114 India
| | - Constanza Jiménez
- BioMAT'X, Facultad De Odontología; Universidad De Los Andes; Mons. Álvaro Del Portillo Santiago 12.455 Chile
- Centro De Investigación Biomédica (CIB), Facultad De Medicina; Universidad De Los Andes; Mons. Álvaro Del Portillo Santiago 12.455 Chile
| | - Javier Campos
- BioMAT'X, Facultad De Odontología; Universidad De Los Andes; Mons. Álvaro Del Portillo Santiago 12.455 Chile
- Plan De Mejoramiento Institucional (PMI) En Innovación-I+D+I, Universidad De Los Andes; Santiago 12.455 Chile
| | - Paula Ibarra
- BioMAT'X, Facultad De Odontología; Universidad De Los Andes; Mons. Álvaro Del Portillo Santiago 12.455 Chile
- Plan De Mejoramiento Institucional (PMI) En Innovación-I+D+I, Universidad De Los Andes; Santiago 12.455 Chile
| | - Ziyad S. Haidar
- BioMAT'X, Facultad De Odontología; Universidad De Los Andes; Mons. Álvaro Del Portillo Santiago 12.455 Chile
- Plan De Mejoramiento Institucional (PMI) En Innovación-I+D+I, Universidad De Los Andes; Santiago 12.455 Chile
| | - Murugan Ramalingam
- Centre for Stem Cell Research (CSCR); A Unit of Institute for Stem Cell Biology and Regenerative Medicine-Bengaluru, Stem Cell Nanotechnology Lab, Christian Medical College Campus; Vellore 632002 India
- WPI-Advanced Institute for Materials Research, Tohoku University; Sendai 980-8577 Japan
| |
Collapse
|
28
|
Sun Y, Li W, Wu X, Zhang N, Zhang Y, Ouyang S, Song X, Fang X, Seeram R, Xue W, He L, Wu W. Functional Self-Assembling Peptide Nanofiber Hydrogels Designed for Nerve Degeneration. ACS APPLIED MATERIALS & INTERFACES 2016; 8:2348-59. [PMID: 26720334 DOI: 10.1021/acsami.5b11473] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Self-assembling peptide (SAP) RADA16-I (Ac-(RADA)4-CONH2) has been suffering from a main drawback associated with low pH, which damages cells and host tissues upon direct exposure. In this study, we presented a strategy to prepare nanofiber hydrogels from two designer SAPs at neutral pH. RADA16-I was appended with functional motifs containing cell adhesion peptide RGD and neurite outgrowth peptide IKVAV. The two SAPs were specially designed to have opposite net charges at neutral pH, the combination of which created a nanofiber hydrogel (-IKVAV/-RGD) characterized by significantly higher G' than G″ in a viscoelasticity examination. Circular dichroism, Fourier transform infrared spectroscopy, and Raman measurements were performed to investigate the secondary structure of the designer SAPs, indicating that both the hydrophobic/hydrophilic properties and electrostatic interactions of the functional motifs play an important role in the self-assembling behavior of the designer SAPs. The neural progenitor cells (NPCs)/stem cells (NSCs) fully embedded in the 3D-IKVAV/-RGD nanofiber hydrogel survived, whereas those embedded within the RADA 16-I hydrogel hardly survived. Moreover, the -IKVAV/-RGD nanofiber hydrogel supported NPC/NSC neuron and astrocyte differentiation in a 3D environment without adding extra growth factors. Studies of three nerve injury models, including sciatic nerve defect, intracerebral hemorrhage, and spinal cord transection, indicated that the designer -IKVAV/-RGD nanofiber hydrogel provided a more permissive environment for nerve regeneration than the RADA 16-I hydrogel. Therefore, we reported a new mechanism that might be beneficial for the synthesis of SAPs for in vitro 3D cell culture and nerve regeneration.
Collapse
Affiliation(s)
- Yuqiao Sun
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong, 510632, China
| | - Wen Li
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
| | - Xiaoli Wu
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
| | - Na Zhang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration (GHMICR), Jinan University , Guangzhou, Guangdong, 510632, China
| | - Yongnu Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong, 510632, China
| | - Songying Ouyang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Xiyong Song
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Xinyu Fang
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
| | - Ramakrishna Seeram
- Guangdong-Hongkong-Macau Institute of CNS Regeneration (GHMICR), Jinan University , Guangzhou, Guangdong, 510632, China
- Center for Nanofibers and Nanotechnology, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore , Singapore , 117576
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong, 510632, China
| | - Liumin He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong, 510632, China
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
| | - Wutian Wu
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration (GHMICR), Jinan University , Guangzhou, Guangdong, 510632, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong , Pokfulam, Hong Kong SAR, 000000, PR China
| |
Collapse
|
29
|
Dinjaski N, Prieto MA. Smart polyhydroxyalkanoate nanobeads by protein based functionalization. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:885-99. [PMID: 25720989 PMCID: PMC7106125 DOI: 10.1016/j.nano.2015.01.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 10/11/2014] [Accepted: 01/05/2015] [Indexed: 11/29/2022]
Abstract
The development of innovative medicines and personalized biomedical approaches calls for new generation easily tunable biomaterials that can be manufactured applying straightforward and low-priced technologies. Production of functionalized bacterial polyhydroxyalkanoate (PHA) nanobeads by harnessing their natural carbon-storage granule production system is a thrilling recent development. This branch of nanobiotechnology employs proteins intrinsically binding the PHA granules as tags to immobilize recombinant proteins of interest and design functional nanocarriers for wide range of applications. Additionally, the implementation of new methodological platforms regarding production of endotoxin free PHA nanobeads using Gram-positive bacteria opened new avenues for biomedical applications. This prompts serious considerations of possible exploitation of bacterial cell factories as alternatives to traditional chemical synthesis and sources of novel bioproducts that could dramatically expand possible applications of biopolymers. From the Clinical Editor In the 21st century, we are coming into the age of personalized medicine. There is a growing use of biomaterials in the clinical setting. In this review article, the authors describe the use of natural polyhydroxyalkanoate (PHA) nanoparticulates, which are formed within bacterial cells and can be easily functionalized. The potential uses would include high-affinity bioseparation, enzyme immobilization, protein delivery, diagnostics etc. The challenges of this approach remain the possible toxicity from endotoxin and the high cost of production.
Collapse
Affiliation(s)
- Nina Dinjaski
- Polymer Biotechnology Lab, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - M Auxiliadora Prieto
- Polymer Biotechnology Lab, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain.
| |
Collapse
|
30
|
Preparation and characterisation of poly(hydroxyalkanoate)/Ganoderma lucidum fibre composites: mechanical and biological properties. Polym Bull (Berl) 2015. [DOI: 10.1007/s00289-015-1307-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
31
|
de Luca AC, Lacour SP, Raffoul W, di Summa PG. Extracellular matrix components in peripheral nerve repair: how to affect neural cellular response and nerve regeneration? Neural Regen Res 2015; 9:1943-8. [PMID: 25598773 PMCID: PMC4283273 DOI: 10.4103/1673-5374.145366] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2014] [Indexed: 01/09/2023] Open
Abstract
Peripheral nerve injury is a serious problem affecting significantly patients’ life. Autografts are the “gold standard” used to repair the injury gap, however, only 50% of patients fully recover from the trauma. Artificial conduits are a valid alternative to repairing peripheral nerve. They aim at confining the nerve environment throughout the regeneration process, and providing guidance to axon outgrowth. Biocompatible materials have been carefully designed to reduce inflammation and scar tissue formation, but modifications of the inner lumen are still required in order to optimise the scaffolds. Biomicking the native neural tissue with extracellular matrix fillers or coatings showed great promises in repairing longer gaps and extending cell survival. In addition, extracellular matrix molecules provide a platform to further bind growth factors that can be released in the system over time. Alternatively, conduit fillers can be used for cell transplantation at the injury site, reducing the lag time required for endogenous Schwann cells to proliferate and take part in the regeneration process. This review provides an overview on the importance of extracellular matrix molecules in peripheral nerve repair.
Collapse
Affiliation(s)
- Alba C de Luca
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Stephanie P Lacour
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Wassim Raffoul
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Pietro G di Summa
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
32
|
Bai S, Zhang W, Lu Q, Ma Q, Kaplan DL, Zhu H. Silk Nanofiber Hydrogels with Tunable Modulus to Regulate Nerve Stem Cell Fate. J Mater Chem B 2014; 2:6590-6600. [PMID: 25530851 PMCID: PMC4269376 DOI: 10.1039/c4tb00878b] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Reconstruction of damaged nerves remains a significant unmet challenge in clinical medicine. To foster improvements, the control of neural stem cell (NSC) behaviors, including migration, proliferation and differentiation are critical factors to consider. Topographical and mechanical stimulation based on the control of biomaterial features is a promising approach, which are usually studied separately. The synergy between topography and mechanical rigidity could offer new insights into the control of neural cell fate if they could be utilized concurrently in studies. To achieve this need, silk fibroin self-assembled nanofibers with a beta-sheet-enriched structure are formed into hydrogels. Stiffness is tuned using different annealing processes to enable mechanical control without impacting the nanofiber topography. Compared with nonannealed nanofibers, NSCs on methanol annealed nanofibers with stiffness similar to nerve tissues differentiate into neurons with the restraint of glial differentiation, without the influence of specific differentiation biochemical factors. These results demonstrate that combining topographic and mechanical cues provides the control of nerve cell behaviors, with potential for neurogenerative repair strategies.
Collapse
Affiliation(s)
- ShuMeng Bai
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - WenMin Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, the Second Affiliated Hospital, Soochow University, Suzhou 215123, People’s Republic of China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
- Jiangsu Province Key Laboratory of Stem Cell Research, Medical College, Soochow University, Suzhou 215006, People’s Republic of China
| | - QuanHong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, the Second Affiliated Hospital, Soochow University, Suzhou 215123, People’s Republic of China
| | - David L. Kaplan
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - HeSun Zhu
- Research Center of Materials Science, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| |
Collapse
|
33
|
Lee IC, Wu YC. Assembly of polyelectrolyte multilayer films on supported lipid bilayers to induce neural stem/progenitor cell differentiation into functional neurons. ACS APPLIED MATERIALS & INTERFACES 2014; 6:14439-50. [PMID: 25111699 DOI: 10.1021/am503750w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The key factors affecting the success of neural engineering using neural stem/progenitor cells (NSPCs) are the neuron quantity, the guidance of neurite outgrowth, and the induction of neurons to form functional synapses at synaptic junctions. Herein, a biomimetic material comprising a supported lipid bilayer (SLB) with adsorbed sequential polyelectrolyte multilayer (PEM) films was fabricated to induce NSPCs to form functional neurons without the need for serum and growth factors in a short-term culture. SLBs are suitable artificial substrates for neural engineering due to their structural similarity to synaptic membranes. In addition, PEM film adsorption provides protection for the SLB as well as the ability to vary the surface properties to evaluate the effects of physical and mechanical signals on NSPC differentiation. Our results revealed that NSPCs were inducible on SLB-PEM films consisting of up to eight alternating layers. In addition, the process outgrowth length, the percentage of differentiated neurons, and the synaptic function were regulated by the number of layers and the surface charge of the outermost layer. The average process outgrowth length was greater than 500 μm on SLB-PLL/PLGA (n = 7.5) after only 3 days of culture. Moreover, the quantity and quality of the differentiated neurons were obviously enhanced on the SLB-PEM system compared with those on the PEM-only substrates. These results suggest that the PEM films can induce NSPC adhesion and differentiation and that an SLB base may enhance neuron differentiation and trigger the formation of functional synapses.
Collapse
Affiliation(s)
- I-Chi Lee
- Graduate Institute of Biochemical and Biomedical Engineering, Chang-Gung University , No. 259, Wenhua First Road, Guishan Township, Taoyuan County, 33302, Taiwan (R.O.C.)
| | | |
Collapse
|
34
|
Carriers in cell-based therapies for neurological disorders. Int J Mol Sci 2014; 15:10669-723. [PMID: 24933636 PMCID: PMC4100175 DOI: 10.3390/ijms150610669] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/19/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023] Open
Abstract
There is a pressing need for long-term neuroprotective and neuroregenerative therapies to promote full function recovery of injuries in the human nervous system resulting from trauma, stroke or degenerative diseases. Although cell-based therapies are promising in supporting repair and regeneration, direct introduction to the injury site is plagued by problems such as low transplanted cell survival rate, limited graft integration, immunorejection, and tumor formation. Neural tissue engineering offers an integrative and multifaceted approach to tackle these complex neurological disorders. Synergistic therapeutic effects can be obtained from combining customized biomaterial scaffolds with cell-based therapies. Current scaffold-facilitated cell transplantation strategies aim to achieve structural and functional rescue via offering a three-dimensional permissive and instructive environment for sustainable neuroactive factor production for prolonged periods and/or cell replacement at the target site. In this review, we intend to highlight important considerations in biomaterial selection and to review major biodegradable or non-biodegradable scaffolds used for cell transplantation to the central and peripheral nervous system in preclinical and clinical trials. Expanded knowledge in biomaterial properties and their prolonged interaction with transplanted and host cells have greatly expanded the possibilities for designing suitable carrier systems and the potential of cell therapies in the nervous system.
Collapse
|