1
|
Li H, Yu L, Li Z, Li S, Liu Y, Qu G, Chen K, Huang L, Li Z, Ren J, Wu X, Huang J. A Narrative Review of Bioactive Hydrogel Microspheres: Ingredients, Modifications, Fabrications, Biological Functions, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500426. [PMID: 40103506 DOI: 10.1002/smll.202500426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/02/2025] [Indexed: 03/20/2025]
Abstract
Hydrogel microspheres are important in regenerative medicine and tissue engineering, acting as cargos of cells, drugs, growth factors, bio-inks for 3D printing, and medical devices. The antimicrobial and anti-inflammatory characteristics of hydrogel microspheres are good for treating injured tissues. However, the biological properties of hydrogel microspheres should be modified for optimal treatment of various body parts with different physiological and biochemical environments. In addition, specific preparation methods are required to produce customized hydrogel microspheres with different shapes and sizes for various clinical applications. Herein, the advances in hydrogel microspheres for biomedical applications are reviewed. Synthesis methods for hydrogel precursor solutions, manufacturing methods, and strategies for enhancing the biological functions of these hydrogel microspheres are described. The involvement of bioactive hydrogel microspheres in tissue repair is also discussed. This review anticipates fostering more insights into the design, production, and application of hydrogel microspheres in biomedicine.
Collapse
Affiliation(s)
- Haohui Li
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Yu
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ze Li
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Sicheng Li
- School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Ye Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Guiwen Qu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Kang Chen
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Luqiao Huang
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zongan Li
- Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing, NARI School of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, 210042, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jinjian Huang
- Research Institute of General Surgery, Jinling Hospital, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Medicine, Nanjing University, Nanjing, 210093, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
2
|
Wei Y, Liang Y, Qi K, Gu Z, Yan B, Xie H. Exploring the application of piezoelectric ceramics in bone regeneration. J Biomater Appl 2024; 39:409-420. [PMID: 39152927 DOI: 10.1177/08853282241274528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
Piezoelectric ceramics are piezoelectric materials with polycrystalline structure and have been widely used in many fields such as medical imaging and sound sensors. As knowledge about this kind of material develops, researchers find piezoelectric ceramics possess favorable piezoelectricity, biocompatibility, mechanical properties, porous structure and antibacterial effect and endeavor to apply piezoelectric ceramics to the field of bone tissue engineering. However, clinically no piezoelectric ceramics have been exercised so far. Therefore, in this paper we present a comprehensive review of the research and development of various piezoelectric ceramics including barium titanate, potassium sodium niobate and zinc oxide ceramics and aims to explore the application of piezoelectric ceramics in bone regeneration by providing a detailed overview of the current knowledge and research of piezoelectric ceramics in bone tissue regeneration.
Collapse
Affiliation(s)
- Yige Wei
- State Key Laboratory of Oral Diseases, National Center for Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaxian Liang
- State Key Laboratory of Oral Diseases, National Center for Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kailong Qi
- State Key Laboratory of Oral Diseases, National Center for Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Bing Yan
- State Key Laboratory of Oral Diseases, National Center for Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Center for Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Banik O, Salve AL, Kumar P, Kumar S, Banoth E. Electrically conductive nanomaterials: transformative applications in biomedical engineering-a review. NANOTECHNOLOGY 2024; 36:022001. [PMID: 39389095 DOI: 10.1088/1361-6528/ad857d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
In recent years, significant advancements in nanotechnology have improved the various disciplines of scientific fields. Nanomaterials, like, carbon-based (carbon nanotubes, graphene), metallic, metal oxides, conductive polymers, and 2D materials (MXenes) exhibit exceptional electrical conductivity, mechanical strength, flexibility, thermal property and chemical stability. These materials hold significant capability in transforming material science and biomedical engineering by enabling the creation of more efficient, miniaturized, and versatile devices. The indulgence of nanotechnology with conductive materials in biological fields promises a transformative innovation across various industries, from bioelectronics to environmental regulations. The conductivity of nanomaterials with a suitable size and shape exhibits unique characteristics, which provides a platform for realization in bioelectronics as biosensors, tissue engineering, wound healing, and drug delivery systems. It can be explored for state-of-the-art cardiac, skeletal, nerve, and bone scaffold fabrication while highlighting their proof-of-concept in the development of biosensing probes and medical imaging. This review paper highlights the significance and application of the conductive nanomaterials associated with conductivity and their contribution towards a new perspective in improving the healthcare system globally.
Collapse
Affiliation(s)
- Oindrila Banik
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Amol Lalchand Salve
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Prasoon Kumar
- BioDesign and Medical Devices, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Santosh Kumar
- Department of Electronics and Communication Engineering, Centre of Excellence for Nanotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh 522302, India
| | - Earu Banoth
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
4
|
Kohestani AA, Xu Z, Baştan FE, Boccaccini AR, Pishbin F. Electrically conductive coatings in tissue engineering. Acta Biomater 2024; 186:30-62. [PMID: 39128796 DOI: 10.1016/j.actbio.2024.08.007] [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: 04/14/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Recent interest in tissue engineering (TE) has focused on electrically conductive biomaterials. This has been inspired by the characteristics of the cells' microenvironment where signalling is supported by electrical stimulation. Numerous studies have demonstrated the positive influence of electrical stimulation on cell excitation to proliferate, differentiate, and deposit extracellular matrix. Even without external electrical stimulation, research shows that electrically active scaffolds can improve tissue regeneration capacity. Tissues like bone, muscle, and neural contain electrically excitable cells that respond to electrical cues provided by implanted biomaterials. To introduce an electrical pathway, TE scaffolds can incorporate conductive polymers, metallic nanoparticles, and ceramic nanostructures. However, these materials often do not meet implantation criteria, such as maintaining mechanical durability and degradation characteristics, making them unsuitable as scaffold matrices. Instead, depositing conductive layers on TE scaffolds has shown promise as an efficient alternative to creating electrically conductive structures. A stratified scaffold with an electroactive surface synergistically excites the cells through active top-pathway, with/without electrical stimulation, providing an ideal matrix for cell growth, proliferation, and tissue deposition. Additionally, these conductive coatings can be enriched with bioactive or pharmaceutical components to enhance the scaffold's biomedical performance. This review covers recent developments in electrically active biomedical coatings for TE. The physicochemical and biological properties of conductive coating materials, including polymers (polypyrrole, polyaniline and PEDOT:PSS), metallic nanoparticles (gold, silver) and inorganic (ceramic) particles (carbon nanotubes, graphene-based materials and Mxenes) are examined. Each section explores the conductive coatings' deposition techniques, deposition parameters, conductivity ranges, deposit morphology, cell responses, and toxicity levels in detail. Furthermore, the applications of these conductive layers, primarily in bone, muscle, and neural TE are considered, and findings from in vitro and in vivo investigations are presented. STATEMENT OF SIGNIFICANCE: Tissue engineering (TE) scaffolds are crucial for human tissue replacement and acceleration of healing. Neural, muscle, bone, and skin tissues have electrically excitable cells, and their regeneration can be enhanced by electrically conductive scaffolds. However, standalone conductive materials often fall short for TE applications. An effective approach involves coating scaffolds with a conductive layer, finely tuning surface properties while leveraging the scaffold's innate biological and physical support. Further enhancement is achieved by modifying the conductive layer with pharmaceutical components. This review explores the under-reviewed topic of conductive coatings in tissue engineering, introducing conductive biomaterial coatings and analyzing their biological interactions. It provides insights into enhancing scaffold functionality for tissue regeneration, bridging a critical gap in current literature.
Collapse
Affiliation(s)
- Abolfazl Anvari Kohestani
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran 11155-4563 Tehran, Iran
| | - Zhiyan Xu
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Fatih Erdem Baştan
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany; Thermal Spray Research and Development Laboratory, Metallurgical and Materials Engineering Department, Sakarya University, Esentepe Campus, 54187, Turkey
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany.
| | - Fatemehsadat Pishbin
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran 11155-4563 Tehran, Iran.
| |
Collapse
|
5
|
Chen Z, Xiao N, Luo L, Zhang L, Yin F, Hu W, Wu Z, Chen Y, Luo K, Xu X. Nanosilicates facilitate periodontal regeneration potential by activating the PI3K-AKT signaling pathway in periodontal ligament cells. J Nanobiotechnology 2024; 22:532. [PMID: 39223550 PMCID: PMC11370094 DOI: 10.1186/s12951-024-02798-6] [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: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The recent development of nanobiomaterials has shed some light on the field of periodontal tissue regeneration. Laponite (LAP), an artificially synthesized two-dimensional (2D) disk-shaped nanosilicate, has garnered substantial attention in regenerative biomedical applications owing to its distinctive structure, exceptional biocompatibility and bioactivity. This study endeavors to comprehensively evaluate the influence of LAP on periodontal regeneration. The effects of LAP on periodontal ligament cells (PDLCs) on osteogenesis, cementogenesis and angiogenesis were systematically assessed, and the potential mechanism was explored through RNA sequencing. The results indicated that LAP improved osteogenic and cementogenic differentiation of PDLCs, the regulatory effects of LAP on PDLCs were closely correlated with activation of PI3K-AKT signaling pathway. Moreover, LAP enhanced angiogenesis indirectly via manipulating paracrine of PDLCs. Then, LAP was implanted into rat periodontal defect to confirm its regenerative potential. Both micro-CT and histological analysis indicated that LAP could facilitate periodontal tissue regeneration in vivo. These findings provide insights into the bioactivity and underlying mechanism of LAP on PDLCs, highlighting it might be a potential therapeutic option in periodontal therapy.
Collapse
Affiliation(s)
- Ziqin Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Nianqi Xiao
- Gannan Health Vocational College, Ganzhou, Jiangxi, 341000, P.R. China
| | - Lan Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Fan Yin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Weiqiang Hu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| |
Collapse
|
6
|
Fallert L, Urigoitia-Asua A, Cipitria A, Jimenez de Aberasturi D. Dynamic 3D in vitro lung models: applications of inorganic nanoparticles for model development and characterization. NANOSCALE 2024; 16:10880-10900. [PMID: 38787741 DOI: 10.1039/d3nr06672j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Being a vital organ exposed to the external environment, the lung is susceptible to a plethora of pathogens and pollutants. This is reflected in high incidences of chronic respiratory diseases, which remain a leading cause of mortality world-wide and pose a persistent global burden. It is thus of paramount importance to improve our understanding of these pathologies and provide better therapeutic options. This necessitates the development of representative and physiologically relevant in vitro models. Advances in bioengineering have enabled the development of sophisticated models that not only capture the three-dimensional architecture of the cellular environment but also incorporate the dynamics of local biophysical stimuli. However, such complex models also require novel approaches that provide reliable characterization. Within this review we explore how 3D bioprinting and nanoparticles can serve as multifaceted tools to develop such dynamic 4D printed in vitro lung models and facilitate their characterization in the context of pulmonary fibrosis and breast cancer lung metastasis.
Collapse
Affiliation(s)
- Laura Fallert
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
| | - Ane Urigoitia-Asua
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
- POLYMAT, Basque Centre for Macromolecular Design and Engineering, 20018 Donostia-San Sebastián, Spain
| | - Amaia Cipitria
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Dorleta Jimenez de Aberasturi
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), 20014 Donostia-San Sebastián, Spain
| |
Collapse
|
7
|
Khan T, Vadivel G, Ramasamy B, Murugesan G, Sebaey TA. Biodegradable Conducting Polymer-Based Composites for Biomedical Applications-A Review. Polymers (Basel) 2024; 16:1533. [PMID: 38891481 PMCID: PMC11175044 DOI: 10.3390/polym16111533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
In recent years, researchers have increasingly directed their focus toward the biomedical field, driven by the goal of engineering polymer systems that possess a unique combination of both electrical conductivity and biodegradability. This convergence of properties holds significant promise, as it addresses a fundamental requirement for biomedical applications: compatibility with biological environments. These polymer systems are viewed as auspicious biomaterials, precisely because they meet this critical criterion. Beyond their biodegradability, these materials offer a range of advantageous characteristics. Their exceptional processability enables facile fabrication into various forms, and their chemical stability ensures reliability in diverse physiological conditions. Moreover, their low production costs make them economically viable options for large-scale applications. Notably, their intrinsic electrical conductivity further distinguishes them, opening up possibilities for applications that demand such functionality. As the focus of this review, a survey into the use of biodegradable conducting polymers in tissue engineering, biomedical implants, and antibacterial applications is conducted.
Collapse
Affiliation(s)
- Tabrej Khan
- Department of Engineering Management, College of Engineering, Prince Sultan University, Riyadh 11586, Saudi Arabia
| | - Gayathri Vadivel
- Department of Physics, KPR Institute of Engineering and Technology, Coimbatore 641407, Tamil Nadu, India
| | - Balan Ramasamy
- Department of Physics, Government Arts and Science College, Mettupalayam 641104, Tamil Nadu, India
| | - Gowtham Murugesan
- Department of Physics, Kongunadu Arts and Science College, Coimbatore 641029, Tamil Nadu, India
| | - Tamer A. Sebaey
- Department of Engineering Management, College of Engineering, Prince Sultan University, Riyadh 11586, Saudi Arabia
- Department of Mechanical Design and Production Engineering, Faculty of Engineering, Zagazig University, Zagazig 44519, Sharkia, Egypt
| |
Collapse
|
8
|
Raja IS, Kim B, Han DW. Nanofibrous Material-Reinforced Printable Ink for Enhanced Cell Proliferation and Tissue Regeneration. Bioengineering (Basel) 2024; 11:363. [PMID: 38671784 PMCID: PMC11047974 DOI: 10.3390/bioengineering11040363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The three-dimensional (3D) printing of biomaterials, cells, and bioactive components, including growth factors, has gained interest among researchers in the field of tissue engineering (TE) with the aim of developing many scaffolds to sustain size, shape fidelity, and structure and retain viable cells inside a network. The biocompatible hydrogel employed in 3D printing should be soft enough to accommodate cell survival. At the same time, the gel should be mechanically strong to avoid the leakage of cells into the surrounding medium. Considering these basic criteria, researchers have developed nanocomposite-based printable inks with suitable mechanical and electroconductive properties. These nanomaterials, including carbon family nanomaterials, transition metal dichalcogenides, and polymeric nanoparticles, act as nanofillers and dissipate stress across polymeric networks through their electroactive interactions. Nanofiber-reinforced printable ink is one kind of nanocomposite-based ink that comprises dispersed nanofiber components in a hydrogel matrix. In this current review, we compile various TE applications of nanofiber-reinforced printable ink and describe the 3D-printing parameters, classification, and impact of cross-linkage. Furthermore, we discuss the challenges and future perspectives in this field.
Collapse
Affiliation(s)
| | - Bongju Kim
- Dental Life Science Research Institute, Seoul National University Dental Hospital, Seoul 03080, Republic of Korea;
| | - Dong-Wook Han
- Institute of Nano-Bio Convergence, Pusan National University, Busan 46241, Republic of Korea
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
9
|
Sacchi M, Sauter-Starace F, Mailley P, Texier I. Resorbable conductive materials for optimally interfacing medical devices with the living. Front Bioeng Biotechnol 2024; 12:1294238. [PMID: 38449676 PMCID: PMC10916519 DOI: 10.3389/fbioe.2024.1294238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/02/2024] [Indexed: 03/08/2024] Open
Abstract
Implantable and wearable bioelectronic systems are arising growing interest in the medical field. Linking the microelectronic (electronic conductivity) and biological (ionic conductivity) worlds, the biocompatible conductive materials at the electrode/tissue interface are key components in these systems. We herein focus more particularly on resorbable bioelectronic systems, which can safely degrade in the biological environment once they have completed their purpose, namely, stimulating or sensing biological activity in the tissues. Resorbable conductive materials are also explored in the fields of tissue engineering and 3D cell culture. After a short description of polymer-based substrates and scaffolds, and resorbable electrical conductors, we review how they can be combined to design resorbable conductive materials. Although these materials are still emerging, various medical and biomedical applications are already taking shape that can profoundly modify post-operative and wound healing follow-up. Future challenges and perspectives in the field are proposed.
Collapse
Affiliation(s)
- Marta Sacchi
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
- Université Paris-Saclay, CEA, JACOB-SEPIA, Fontenay-aux-Roses, France
| | - Fabien Sauter-Starace
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| | - Pascal Mailley
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| | - Isabelle Texier
- Université Grenoble Alpes, CEA, LETI-DTIS (Département des Technologies pour l’Innovation en Santé), Grenoble, France
| |
Collapse
|
10
|
Afsharian MH, Mahdavian R, Jafari S, Allahverdi A, Soleymani H, Naderi-Manesh H. Investigation of synergic effects of nanogroove topography and polyaniline-chitosan nanocomposites on PC12 cell differentiation and axonogenesis. iScience 2024; 27:108828. [PMID: 38303727 PMCID: PMC10831943 DOI: 10.1016/j.isci.2024.108828] [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: 07/06/2023] [Revised: 10/09/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Axonal damage is the main characteristic of neurodegenerative diseases. This research was focused on remodeling cell morphology and developing a semi-tissue nanoenvironment via mechanobiological stimuli. The combination of nanogroove topography and polyaniline-chitosan enabled the manipulation of the cells by changing the morphology of PC12 cells to spindle shape and inducing the early stage of signal transduction, which is vital for differentiation. The nanosubstarte embedded with nanogooves induced PC12 cells to elongate their morphology and increase their size by 51% as compared with controls. In addition, the use of an electroconductive nanocomposite alongside nanogrooves resulted in the differentiation of PC12 cells into neurons with an average length of 193 ± 7 μm for each axon and an average number of seven axons for each neurite. Our results represent a combined tool to initiate a promising future for cell reprogramming by inducing cell differentiation and specific cellular morphology in many cases, including neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammad Hossein Afsharian
- Department of Biophysics, Faculty of Biological Sciences Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O. Box: 14115-111, Tehran, Iran
| | - Reza Mahdavian
- Department of Biophysics, Faculty of Biological Sciences Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O. Box: 14115-111, Tehran, Iran
| | - Samira Jafari
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abdollah Allahverdi
- Department of Biophysics, Faculty of Biological Sciences Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O. Box: 14115-111, Tehran, Iran
| | - Hossein Soleymani
- Department of Biophysics, Faculty of Biological Sciences Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O. Box: 14115-111, Tehran, Iran
| | - Hossein Naderi-Manesh
- Department of Biophysics, Faculty of Biological Sciences Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O. Box: 14115-111, Tehran, Iran
| |
Collapse
|
11
|
Truong LB, Medina-Cruz D, Mostafavi E. Gold nanoparticles for delivery of nucleic acid constructs for cancer treatment. GOLD NANOPARTICLES FOR DRUG DELIVERY 2024:141-165. [DOI: 10.1016/b978-0-443-19061-2.00005-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
赵 少, 郝 晓, 菅 炎, 王 一, 刘 伟, 邵 欣, 樊 俊, 徐 松. [A dual-crosslinked injectable hydrogel derived from muscular decellularized matrix promoting myoblasts proliferation and myogenic differentiation]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2023; 37:1514-1522. [PMID: 38130196 PMCID: PMC10739670 DOI: 10.7507/1002-1892.202306054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/18/2023] [Indexed: 12/23/2023]
Abstract
Objective To investigate the feasibility of a dual-crosslinked injectable hydrogel derived from acellular musclar matrix (AMM) for promoting myoblasts proliferation and myogenic differentiation. Methods Firstly, hyaluronic acid was oxidized with NaIO 4 and methylated to prepare methacrylamidated oxidized hyaluronic acid (MOHA). Then, AMM obtained by washing enzymatically treated muscle tissue was aminolyzed to prepare aminated AMM (AAMM). MOHA hydrogel and AAMM were crosslinked using Schiff based reaction and UV radiation to prepare a dual-crosslinked MOHA/AAMM injectable hydrogel. Fourier transform infrared spectroscopy (FTIR) was used to characterize MOHA, AAMM, and MOHA/AAMM hydrogels. The injectability of MOHA/AAMM hydrogel were evaluated by manual injection, and the gelation performance was assessed by UV crosslinking. The rheological properties and Young's modulus of the hydrogel were examined through mechanical tests. The degradation rate of the hydrogel was assessed by immersing it in PBS. The active components of the hydrogel were verified using immunofluorescence staining and ELISA assay kits. The promotion of cell proliferation by the hydrogel was tested using live/dead staining and cell counting kit 8 (CCK-8) assays after co-culturing with C2C12 myoblasts for 9 days. The effect of the hydrogel on myogenic differentiation was evaluated by immunofluorescence staining and real time quantitative polymerase chain reaction (RT-qPCR). Results FTIR spectra confirmed the successful preparation of MOHA/AAMM hydrogel. The hydrogel exhibited good injectability and gelation ability. Compared to MOHA hydrogel, MOHA/AAMM hydrogel exhibited higher viscosity and Young's modulus, a reduced degradation rate, and contained a higher amount of collagen (including collagen type Ⅰ and collagen type Ⅲ) as well as bioactive factors (including epidermal growth factor, fibroblast growth factor 2, vascular endothelial growth factor, and insulin-like growth factor 1). The live/dead cell staining and CCK-8 assay indicated that with prolonged incubation time, there was a significant increase in viable cells and a decrease in dead cells in the C2C12 myoblasts within the MOHA/AAMM hydrogel. Compared with MOHA hydrogel, the difference was significant at each time point ( P<0.05). Immunofluorescence staining and RT-qPCR analysis demonstrated that the deposition of IGF-1 and expression levels of myogenic-related genes (including Myogenin, Troponin T, and myosin heavy chain) in the MOHA/AAMM group were significantly higher than those in the MOHA group ( P<0.05). Conclusion The MOHA/AAMM hydrogel prepared based on AMM can promote myoblasts proliferation and myogenic differentiation, providing a novel dual-crosslinked injectable hydrogel for muscle tissue engineering.
Collapse
Affiliation(s)
- 少华 赵
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 晓亮 郝
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 炎鹏 菅
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 一公 王
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 伟杰 刘
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 欣慰 邵
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 俊 樊
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| | - 松山 徐
- 河南科技大学附属许昌市中心医院整形美容科(河南许昌 461000)Department of Plastic Surgery, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang Henan, 461000, P. R. China
| |
Collapse
|
13
|
Brooks AK, Ramsey RG, Zhang N, Yadavalli VK. Tunable Light-Actuated Interpenetrating Networks of Silk Fibroin and Gelatin for Tissue Engineering and Flexible Biodevices. ACS Biomater Sci Eng 2023; 9:5793-5803. [PMID: 37698556 DOI: 10.1021/acsbiomaterials.3c00741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Soft materials with tunable properties are valuable for applications such as tissue engineering, electronic skins, and human-machine interfaces. Materials that are nature-derived offer additional advantages such as biocompatibility, biodegradability, low-cost sourcing, and sustainability. However, these materials often have contrasting properties that limit their use. For example, silk fibroin (SF) has high mechanical strength but lacks processability and cell-adhesive domains. Gelatin, derived from collagen, has excellent biological properties, but is fragile and lacks stability. To overcome these limitations, composites of gelatin and SF have been explored. However, mechanically robust self-supported matrices and electrochemically active or micropatterned substrates were not demonstrated. In this study, we present a composite of photopolymerizable SF and photogelatin, termed photofibrogel (PFG). By incorporating photoreactive properties in both SF and gelatin, control over material properties can be achieved. The PFG composite can be easily and rapidly formed into free-standing, high-resolution architectures with tunable properties. By optimizing the ratio of SF to gelatin, properties such as swelling, mechanical behavior, enzymatic degradation, and patternability are tailored. The PFG composite allows for macroscale and microscale patterning without significant swelling, enabling the fabrication of structures using photolithography and laser cutting techniques. PFG can be patterned with electrically conductive materials, making it suitable for cell guidance and stimulation. The versatility, mechanical robustness, bioactivity, and electrochemical properties of PFG are shown for skeletal muscle tissue engineering using C2C12 cells as a model. Overall, such composite biomaterials with tunable properties have broad potential in flexible bioelectronics, wound healing, regenerative medicine, and food systems.
Collapse
|
14
|
Bianconi S, Oliveira KMC, Klein KL, Wolf J, Schaible A, Schröder K, Barker J, Marzi I, Leppik L, Henrich D. Pretreatment of Mesenchymal Stem Cells with Electrical Stimulation as a Strategy to Improve Bone Tissue Engineering Outcomes. Cells 2023; 12:2151. [PMID: 37681884 PMCID: PMC10487010 DOI: 10.3390/cells12172151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Electrical stimulation (EStim), whether used alone or in combination with bone tissue engineering (BTE) approaches, has been shown to promote bone healing. In our previous in vitro studies, mesenchymal stem cells (MSCs) were exposed to EStim and a sustained, long-lasting increase in osteogenic activity was observed. Based on these findings, we hypothesized that pretreating MSC with EStim, in 2D or 3D cultures, before using them to treat large bone defects would improve BTE treatments. Critical size femur defects were created in 120 Sprague-Dawley rats and treated with scaffold granules seeded with MSCs that were pre-exposed or not (control group) to EStim 1 h/day for 7 days in 2D (MSCs alone) or 3D culture (MSCs + scaffolds). Bone healing was assessed at 1, 4, and 8 weeks post-surgery. In all groups, the percentage of new bone increased, while fibrous tissue and CD68+ cell count decreased over time. However, these and other healing features, like mineral density, bending stiffness, the amount of new bone and cartilage, and the gene expression of osteogenic markers, did not significantly differ between groups. Based on these findings, it appears that the bone healing environment could counteract the long-term, pro-osteogenic effects of EStim seen in our in vitro studies. Thus, EStim seems to be more effective when administered directly and continuously at the defect site during bone healing, as indicated by our previous studies.
Collapse
Affiliation(s)
- Santiago Bianconi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Karla M. C. Oliveira
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Kari-Leticia Klein
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Jakob Wolf
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Alexander Schaible
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Katrin Schröder
- Vascular Research Centre, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - John Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany;
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Liudmila Leppik
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| |
Collapse
|
15
|
Barbosa F, Garrudo FFF, Marques AC, Cabral JMS, Morgado J, Ferreira FC, Silva JC. Novel Electroactive Mineralized Polyacrylonitrile/PEDOT:PSS Electrospun Nanofibers for Bone Repair Applications. Int J Mol Sci 2023; 24:13203. [PMID: 37686010 PMCID: PMC10488027 DOI: 10.3390/ijms241713203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Bone defect repair remains a critical challenge in current orthopedic clinical practice, as the available therapeutic strategies only offer suboptimal outcomes. Therefore, bone tissue engineering (BTE) approaches, involving the development of biomimetic implantable scaffolds combined with osteoprogenitor cells and native-like physical stimuli, are gaining widespread interest. Electrical stimulation (ES)-based therapies have been found to actively promote bone growth and osteogenesis in both in vivo and in vitro settings. Thus, the combination of electroactive scaffolds comprising conductive biomaterials and ES holds significant promise in improving the effectiveness of BTE for clinical applications. The aim of this study was to develop electroconductive polyacrylonitrile/poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PAN/PEDOT:PSS) nanofibers via electrospinning, which are capable of emulating the native tissue's fibrous extracellular matrix (ECM) and providing a platform for the delivery of exogenous ES. The resulting nanofibers were successfully functionalized with apatite-like structures to mimic the inorganic phase of the bone ECM. The conductive electrospun scaffolds presented nanoscale fiber diameters akin to those of collagen fibrils and displayed bone-like conductivity. PEDOT:PSS incorporation was shown to significantly promote scaffold mineralization in vitro. The mineralized electroconductive nanofibers demonstrated improved biological performance as observed by the significantly enhanced proliferation of both human osteoblast-like MG-63 cells and human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs). Moreover, mineralized PAN/PEDOT:PSS nanofibers up-regulated bone marker genes expression levels of hBM-MSCs undergoing osteogenic differentiation, highlighting their potential as electroactive biomimetic BTE scaffolds for innovative bone defect repair strategies.
Collapse
Affiliation(s)
- Frederico Barbosa
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (F.B.); (F.F.F.G.); (J.M.S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Fábio F. F. Garrudo
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (F.B.); (F.F.F.G.); (J.M.S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Department of Bioengineering and Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
| | - Ana C. Marques
- Departament of Chemical Engineering and CERENA—Center for Natural Resources and the Environment, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (F.B.); (F.F.F.G.); (J.M.S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge Morgado
- Department of Bioengineering and Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (F.B.); (F.F.F.G.); (J.M.S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - João C. Silva
- Department of Bioengineering and iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (F.B.); (F.F.F.G.); (J.M.S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
16
|
Mondal A, Nayak AK, Chakraborty P, Banerjee S, Nandy BC. Natural Polymeric Nanobiocomposites for Anti-Cancer Drug Delivery Therapeutics: A Recent Update. Pharmaceutics 2023; 15:2064. [PMID: 37631276 PMCID: PMC10459560 DOI: 10.3390/pharmaceutics15082064] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is one of the most common lethal diseases and the leading cause of mortality worldwide. Effective cancer treatment is a global problem, and subsequent advancements in nanomedicine are useful as substitute management for anti-cancer agents. Nanotechnology, which is gaining popularity, enables fast-expanding delivery methods in science for curing diseases in a site-specific approach, utilizing natural bioactive substances because several studies have established that natural plant-based bioactive compounds can improve the effectiveness of chemotherapy. Bioactive, in combination with nanotechnology, is an exceptionally alluring and recent development in the fight against cancer. Along with their nutritional advantages, natural bioactive chemicals may be used as chemotherapeutic medications to manage cancer. Alginate, starch, xanthan gum, pectin, guar gum, hyaluronic acid, gelatin, albumin, collagen, cellulose, chitosan, and other biopolymers have been employed successfully in the delivery of medicinal products to particular sites. Due to their biodegradability, natural polymeric nanobiocomposites have garnered much interest in developing novel anti-cancer drug delivery methods. There are several techniques to create biopolymer-based nanoparticle systems. However, these systems must be created in an affordable and environmentally sustainable way to be more readily available, selective, and less hazardous to increase treatment effectiveness. Thus, an extensive comprehension of the various facets and recent developments in natural polymeric nanobiocomposites utilized to deliver anti-cancer drugs is imperative. The present article provides an overview of the latest research and developments in natural polymeric nanobiocomposites, particularly emphasizing their applications in the controlled and targeted delivery of anti-cancer drugs.
Collapse
Affiliation(s)
- Arijit Mondal
- Department of Pharmaceutical Chemistry, M.R. College of Pharmaceutical Sciences and Research, Balisha 743 234, India
| | - Amit Kumar Nayak
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar 751 003, India;
| | - Prithviraj Chakraborty
- Department of Pharmaceutics, Royal School of Pharmacy, The Assam Royal Global University, Guwahati 781 035, India;
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, India;
| | - Bankim Chandra Nandy
- Department of Pharmaceutics, M.R. College of Pharmaceutical Sciences and Research, Balisha 743 234, India;
| |
Collapse
|
17
|
Saghebasl S, Nobakht A, Saghebasl H, Hayati S, Naturi O, Rahbarghazi R. Sandwich-like electro-conductive polyurethane-based gelatin/soybean oil nanofibrous scaffolds with a targeted release of simvastatin for cardiac tissue engineering. J Biol Eng 2023; 17:42. [PMID: 37415188 DOI: 10.1186/s13036-023-00364-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Cardiac tissue engineering (CTE) is a promising way for the restoration of injured cardiac tissue in the healthcare system. The development of biodegradable scaffolds with appropriate chemical, electrical, mechanical, and biological properties is an unmet need for the success of CTE. Electrospinning is a versatile technique that has shown potential applications in CTE. Herein, four different types of multifunctional scaffolds, including synthetic-based poly (glycerol sebacate)-polyurethane (PGU), PGU-Soy scaffold, and a series of trilayer scaffolds containing two outer layers of PGU-Soy and a middle (inner) layer of gelatin (G) as a natural and biodegradable macromolecule without simvastatin (S) and with simvastatin (GS), an anti-inflammatory agent, were fabricated in the sandwich-like structure using electrospinning technique. This approach offers a combination of the advantages of both synthetic and natural polymers to enhance the bioactivity and the cell-to-cell and cell-to-matrix intercommunication. An in vitro drug release analysis was performed after the incorporation of soybean oil (Soy) and G. Soy is used as a semiconducting material was introduced to improve the electrical conductivity of nanofibrous scaffolds. The physicochemical properties, contact angle, and biodegradability of the electrospun scaffolds were also assessed. Moreover, the blood compatibility of nanofibrous scaffolds was studied through activated partial thromboplastin time (APTT), prothrombin time (PT), and hemolytic assay. The results showed that all scaffolds exhibited defect-free morphologies with mean fiber diameters in the range of 361 ± 109 to 417 ± 167 nm. A delay in blood clotting was observed, demonstrating the anticoagulant nature of nanofibrous scaffolds. Furthermore, rat cardiomyoblast cell lines (H9C2) were cultured on scaffolds for 7 days, and the morphology and cell arrangement were monitored. Data indicated an appropriate cytocompatibility. Of note, in the PGU-Soy/GS nanofibrous scaffold, a high survival rate was indicated compared to other groups. Our findings exhibited that the simvastatin-loaded polymeric system had positive effects on cardiomyoblasts attachment and growth and could be utilized as a drug release carrier in the field of CTE.
Collapse
Affiliation(s)
- Solmaz Saghebasl
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Nobakht
- Research Center of Biosciences & Biotechnology (RCBB), University of Tabriz, Tabriz, Iran
| | - Hesam Saghebasl
- Faculty of Medicine, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Sanya Hayati
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Asaro GA, Solazzo M, Suku M, Spurling D, Genoud K, Gonzalez JG, Brien FJO, Nicolosi V, Monaghan MG. MXene functionalized collagen biomaterials for cardiac tissue engineering driving iPSC-derived cardiomyocyte maturation. NPJ 2D MATERIALS AND APPLICATIONS 2023; 7:44. [PMID: 38665478 PMCID: PMC11041746 DOI: 10.1038/s41699-023-00409-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 06/15/2023] [Indexed: 04/28/2024]
Abstract
Electroconductive biomaterials are gaining significant consideration for regeneration in tissues where electrical functionality is of crucial importance, such as myocardium, neural, musculoskeletal, and bone tissue. In this work, conductive biohybrid platforms were engineered by blending collagen type I and 2D MXene (Ti3C2Tx) and afterwards covalently crosslinking; to harness the biofunctionality of the protein component and the increased stiffness and enhanced electrical conductivity (matching and even surpassing native tissues) that two-dimensional titanium carbide provides. These MXene platforms were highly biocompatible and resulted in increased proliferation and cell spreading when seeded with fibroblasts. Conversely, they limited bacterial attachment (Staphylococcus aureus) and proliferation. When neonatal rat cardiomyocytes (nrCMs) were cultured on the substrates increased spreading and viability up to day 7 were studied when compared to control collagen substrates. Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were seeded and stimulated using electric-field generation in a custom-made bioreactor. The combination of an electroconductive substrate with an external electrical field enhanced cell growth, and significantly increased cx43 expression. This in vitro study convincingly demonstrates the potential of this engineered conductive biohybrid platform for cardiac tissue regeneration.
Collapse
Affiliation(s)
- Giuseppe A. Asaro
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
| | - Matteo Solazzo
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
| | - Meenakshi Suku
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland
| | - Dahnan Spurling
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- School of Chemistry, Trinity College Dublin, Dublin, 2 Ireland
| | - Katelyn Genoud
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, 2 Ireland
| | - Javier Gutierrez Gonzalez
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, 2 Ireland
| | - Fergal J. O’ Brien
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, 2 Ireland
| | - Valeria Nicolosi
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- School of Chemistry, Trinity College Dublin, Dublin, 2 Ireland
| | - Michael G. Monaghan
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin, 2 Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, 2 Ireland
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland
| |
Collapse
|
19
|
Sharma A, Kokil GR, He Y, Lowe B, Salam A, Altalhi TA, Ye Q, Kumeria T. Inorganic/organic combination: Inorganic particles/polymer composites for tissue engineering applications. Bioact Mater 2023; 24:535-550. [PMID: 36714332 PMCID: PMC9860401 DOI: 10.1016/j.bioactmat.2023.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Biomaterials have ushered the field of tissue engineering and regeneration into a new era with the development of advanced composites. Among these, the composites of inorganic materials with organic polymers present unique structural and biochemical properties equivalent to naturally occurring hybrid systems such as bones, and thus are highly desired. The last decade has witnessed a steady increase in research on such systems with the focus being on mimicking the peculiar properties of inorganic/organic combination composites in nature. In this review, we discuss the recent progress on the use of inorganic particle/polymer composites for tissue engineering and regenerative medicine. We have elaborated the advantages of inorganic particle/polymer composites over their organic particle-based composite counterparts. As the inorganic particles play a crucial role in defining the features and regenerative capacity of such composites, the review puts a special emphasis on the various types of inorganic particles used in inorganic particle/polymer composites. The inorganic particles that are covered in this review are categorised into two broad types (1) solid (e.g., calcium phosphate, hydroxyapatite, etc.) and (2) porous particles (e.g., mesoporous silica, porous silicon etc.), which are elaborated in detail with recent examples. The review also covers other new types of inorganic material (e.g., 2D inorganic materials, clays, etc.) based polymer composites for tissue engineering applications. Lastly, we provide our expert analysis and opinion of the field focusing on the limitations of the currently used inorganic/organic combination composites and the immense potential of new generation of composites that are in development.
Collapse
Affiliation(s)
- Astha Sharma
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Ganesh R. Kokil
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- School of Pharmacy, University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China
| | - Baboucarr Lowe
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Arwa Salam
- Chemistry Department, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Tariq A. Altalhi
- Chemistry Department, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- School of Pharmacy, University of Queensland, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
20
|
Majeed S, Saravanan M, Danish M, Zakariya NA, Ibrahim MNM, Rizvi EH, NisaAndrabi SU, Barabadi H, Mohanta YK, Mostafavi E. Bioengineering of green-synthesized TAT peptide-functionalized silver nanoparticles for apoptotic cell-death mediated therapy of breast adenocarcinoma. Talanta 2023. [DOI: 10.1016/j.talanta.2022.124026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Cruz DM, Mostafavi E, Vernet-Crua A, O’Connell CP, Barabadi H, Mobini S, Cholula-Díaz JL, Guisbiers G, García-Martín JM, Webster TJ. Green nanotechnology and nanoselenium for biomedical applications. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00001-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
22
|
Peidavosi N, Azami M, Beheshtizadeh N, Ramazani Saadatabadi A. Piezoelectric conductive electrospun nanocomposite PCL/Polyaniline/Barium Titanate scaffold for tissue engineering applications. Sci Rep 2022; 12:20828. [PMID: 36460783 PMCID: PMC9718788 DOI: 10.1038/s41598-022-25332-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Recent trends in tissue engineering technology have switched to electrical potentials generated through bioactive scaffolds regarding their appropriate effects on cell behaviors. Preparing a piezo-electrical stimuli scaffold with high electrical conductivity for bone and cartilage tissue regeneration is the ultimate goal of the present study. Here, Barium Titanate nanoparticles (BaTiO3 NPs) were used as piezoelectric material and highly conductive binary doped Polyaniline nanoparticles (PANI NPs) were synthesized by oxidative polymerization. Polycaprolactone (PCL) was applied as carrier substrate polymer and conductive spun nanofibrous scaffolds of PCL/PANI composites were prepared in two different amounts of PANI (3 and 5 wt.%). The conductivity of PCL/PANI nanofibers has been analyzed by standard four probes test. Based on the obtained results, the PCL/PANI5 (with 5 wt.% PANI) was selected due to the superior electrical conductivity of 8.06 × 10-4 s cm - 1. Moreover, the piezoelectric nanofibrous scaffolds of PCL/BT composite were electrospun in three different amounts of BT (20, 30, and 40 wt.%). To investigate the synergic effect of conductive PANI and piezoelectric BT, ternary nanocomposite scaffolds of PCL/PANI/BT were prepared using the dual jet electrospinning technique. The piezoelectric properties have been analyzed by determining the produced voltage. The morphological assessment, contact angle, mechanical test, and MTT assay have been conducted to evaluate other properties including biocompatibility of nanofibrous scaffolds. The PCL/PANI5/BT40 composite resulted in an unprecedented voltage of 1.9 Volt. SEM results confirm that BT NPs have been distributed and embedded inside PCL fibers quite appropriately. Also, the chosen scaffolds were homogeneously intertwined and possessed an average fiber diameter of 288 ± 180 nm, and a contact angle of 92 ± 7°, making it a desirable surface for cell attachment and protein interactions. Moreover, Young's modulus, ultimate tensile stress, and elongation were obtained as 11 ± 1 MPa, 5 ± 0.6 MPa, and 109 ± 15% respectively. Obtained results assert the novel potential of piezo-electrical stimuli conductive nanocomposite scaffold for tissue engineering applications.
Collapse
Affiliation(s)
- Naeemeh Peidavosi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
23
|
Mostafavi E, Medina-Cruz D, Truong LB, Kaushik A, Iravani S. Selenium-based nanomaterials for biosensing applications. MATERIALS ADVANCES 2022; 3:7742-7756. [PMID: 36353516 PMCID: PMC9619417 DOI: 10.1039/d2ma00756h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/11/2022] [Indexed: 05/03/2023]
Abstract
The unique chemical and physical features of nanomaterials make them ideal for developing new and better sensing devices, particularly biosensors. Various types of nanoparticles, including metal, oxide, and semiconductor nanostructures, have been utilized to manufacture biosensors, and each kind of nanoparticle plays a unique role in the sensing system. Nanoparticles provide critical roles such as immobilizing biomolecules, catalyzing electrochemical processes, enhancing electron transport between electrode surfaces and proteins, identifying biomolecules, and even functioning as the reactant for the catalytic reaction. Among all the potential nanosystems to be used in biosensors, selenium nanoparticle (SeNP) features have sparked a growing interest in their use in bridging biological recognition events and signal transduction, as well as in developing biosensing devices with novel applications for identification, quantification, and study of different analytes of biological relevance. The optical, physical, and chemical characteristics of differently shaped SeNPs opened up a world of possibilities for developing biosensors of biomedical interest. The outstanding biocompatibility, conductivity, catalytic characteristics, high surface-to-volume ratio, and high density of SeNPs have enabled their widespread use in developing electrochemical biosensors with superior analytical performance compared to other designs of biosensors. This review summarizes recent and ongoing advances, current challenges, and future research perspectives on real-world applications of Se-based nanobiosensors to detect biologically relevant analytes such as hydrogen peroxide, heavy metals, or glucose. Due to the superior properties and multifunctionality of Se-NPs biosensors, these structures can open up considerable new horizons in the future of healthcare and medicine.
Collapse
Affiliation(s)
- Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine Stanford CA 94305 USA
- Department of Medicine, Stanford University School of Medicine Stanford CA 94305 USA
| | - David Medina-Cruz
- Chemical Engineering Department, Northeastern University Boston MA 02115 USA
| | - Linh B Truong
- Chemical Engineering Department, Northeastern University Boston MA 02115 USA
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University Lakeland FL-33805 USA
- School of Engineering, University of Petroleum and Energy Studies (UPES) Dehradun Uttarakhand India
| | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
24
|
McCorry MC, Reardon KF, Black M, Williams C, Babakhanova G, Halpern JM, Sarkar S, Swami NS, Mirica KA, Boermeester S, Underhill A. Sensor technologies for quality control in engineered tissue manufacturing. Biofabrication 2022; 15:10.1088/1758-5090/ac94a1. [PMID: 36150372 PMCID: PMC10283157 DOI: 10.1088/1758-5090/ac94a1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022]
Abstract
The use of engineered cells, tissues, and organs has the opportunity to change the way injuries and diseases are treated. Commercialization of these groundbreaking technologies has been limited in part by the complex and costly nature of their manufacture. Process-related variability and even small changes in the manufacturing process of a living product will impact its quality. Without real-time integrated detection, the magnitude and mechanism of that impact are largely unknown. Real-time and non-destructive sensor technologies are key for in-process insight and ensuring a consistent product throughout commercial scale-up and/or scale-out. The application of a measurement technology into a manufacturing process requires cell and tissue developers to understand the best way to apply a sensor to their process, and for sensor manufacturers to understand the design requirements and end-user needs. Furthermore, sensors to monitor component cells' health and phenotype need to be compatible with novel integrated and automated manufacturing equipment. This review summarizes commercially relevant sensor technologies that can detect meaningful quality attributes during the manufacturing of regenerative medicine products, the gaps within each technology, and sensor considerations for manufacturing.
Collapse
Affiliation(s)
- Mary Clare McCorry
- Advanced Regenerative Manufacturing Institute, Manchester, NH 03101, United States of America
| | - Kenneth F Reardon
- Chemical and Biological Engineering and Biomedical Engineering, Colorado State University, Fort Collins, CO 80521, United States of America
| | - Marcie Black
- Advanced Silicon Group, Lowell, MA 01854, United States of America
| | - Chrysanthi Williams
- Access Biomedical Solutions, Trinity, Florida 34655, United States of America
| | - Greta Babakhanova
- National Institute of Standards and Technology, Gaithersburg, MD 20899, United States of America
| | - Jeffrey M Halpern
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824, United States of America
- Materials Science and Engineering Program, University of New Hampshire, Durham, NH 03824, United States of America
| | - Sumona Sarkar
- National Institute of Standards and Technology, Gaithersburg, MD 20899, United States of America
| | - Nathan S Swami
- Electrical and Computer Engineering, University of Virginia, Charlottesville, VA 22904, United States of America
| | - Katherine A Mirica
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, United States of America
| | - Sarah Boermeester
- Advanced Regenerative Manufacturing Institute, Manchester, NH 03101, United States of America
| | - Abbie Underhill
- Scientific Bioprocessing Inc., Pittsburgh, PA 15238, United States of America
| |
Collapse
|
25
|
Chen H, Zhang Y, Yu T, Song G, Xu T, Xin T, Lin Y, Han B. Nano-Based Drug Delivery Systems for Periodontal Tissue Regeneration. Pharmaceutics 2022; 14:2250. [PMID: 36297683 PMCID: PMC9612159 DOI: 10.3390/pharmaceutics14102250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2022] Open
Abstract
Periodontitis is a dysbiotic biofilm-induced and host-mediated inflammatory disease of tooth supporting tissues that leads to progressive destruction of periodontal ligament and alveolar bone, thereby resulting in gingival recession, deep periodontal pockets, tooth mobility and exfoliation, and aesthetically and functionally compromised dentition. Due to the improved biopharmaceutical and pharmacokinetic properties and targeted and controlled drug release, nano-based drug delivery systems have emerged as a promising strategy for the treatment of periodontal defects, allowing for increased efficacy and safety in controlling local inflammation, establishing a regenerative microenvironment, and regaining bone and attachments. This review provides an overview of nano-based drug delivery systems and illustrates their practical applications, future prospects, and limitations in the field of periodontal tissue regeneration.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunfan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tingting Yu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Guangying Song
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianmin Xu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianyi Xin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yifan Lin
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Bing Han
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
26
|
Preparation and Properties of Electrospun PLLA/PTMC Scaffolds. Polymers (Basel) 2022; 14:polym14204406. [PMID: 36297984 PMCID: PMC9611888 DOI: 10.3390/polym14204406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/03/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
Poly(L-lactide) (PLLA) and PLLA/poly(trimethylene carbonate) (PTMC) scaffolds characterised by different PLLA:PTMC mass ratios (10:0, 9:1, 8:2, 7:3, 6:4 and 5:5) were prepared via electrospinning. The results showed that increasing the PTMC content in the spinning solution caused the following effects: (1) the diameter of the prepared PLLA/PTMC electrospun fibres gradually increased from 188.12 ± 48.87 nm (10:0) to 584.01 ± 60.68 nm (5:5), (2) electrospun fibres with uniform diameters and no beads could be prepared at the PTMC contents of >30%, (3) the elastic modulus of the fibre initially increased and then decreased, reaching a maximum value of 74.49 ± 8.22 Mpa (5:5) and (4) the elongation at the breaking point of the fibres increased gradually from 24.71% to 344.85%. Compared with the PLLA electrospun fibrous membrane, the prepared PLLA/PTMC electrospun fibrous membrane exhibited considerably improved mechanical properties while maintaining good histocompatibility.
Collapse
|
27
|
Alamdari SG, Alibakhshi A, de la Guardia M, Baradaran B, Mohammadzadeh R, Amini M, Kesharwani P, Mokhtarzadeh A, Oroojalian F, Sahebkar A. Conductive and Semiconductive Nanocomposite-Based Hydrogels for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200526. [PMID: 35822350 DOI: 10.1002/adhm.202200526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/26/2022] [Indexed: 01/27/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide and the most common cause is myocardial infarction. Therefore, appropriate approaches should be used to repair damaged heart tissue. Recently, cardiac tissue engineering approaches have been extensively studied. Since the creation of the nature of cardiovascular tissue engineering, many advances have been made in cellular and scaffolding technologies. Due to the hydrated and porous structures of the hydrogel, they are used as a support matrix to deliver cells to the infarct tissue. In heart tissue regeneration, bioactive and biodegradable hydrogels are required by simulating native tissue microenvironments to support myocardial wall stress in addition to preserving cells. Recently, the use of nanostructured hydrogels has increased the use of nanocomposite hydrogels and has revolutionized the field of cardiac tissue engineering. Therefore, to overcome the limitation of the use of hydrogels due to their mechanical fragility, various nanoparticles of polymers, metal, and carbon are used in tissue engineering and create a new opportunity to provide hydrogels with excellent properties. Here, the types of synthetic and natural polymer hydrogels, nanocarbon-based hydrogels, and other nanoparticle-based materials used for cardiac tissue engineering with emphasis on conductive nanostructured hydrogels are briefly introduced.
Collapse
Affiliation(s)
- Sania Ghobadi Alamdari
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, Burjassot, Valencia, 46100, Spain
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Reza Mohammadzadeh
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran.,Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran
| |
Collapse
|
28
|
Ho TH, Yang CH, Jiang ZE, Lin HY, Chen YF, Wang TL. NIR-Triggered Generation of Reactive Oxygen Species and Photodynamic Therapy Based on Mesoporous Silica-Coated LiYF 4 Upconverting Nanoparticles. Int J Mol Sci 2022; 23:ijms23158757. [PMID: 35955888 PMCID: PMC9368848 DOI: 10.3390/ijms23158757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
To date, the increase in reactive oxygen species (ROS) production for effectual photodynamic therapy (PDT) treatment still remains challenging. In this study, a facile and effective approach is utilized to coat mesoporous silica (mSiO2) shell on the ligand-free upconversion nanoparticles (UCNPs) based on the LiYF4 host material. Two kinds of mesoporous silica-coated UCNPs (UCNP@mSiO2) that display green emission (doped with Ho3+) and red emission (doped with Er3+), respectively, were successfully synthesized and well characterized. Three photosensitizers (PSs), merocyanine 540 (MC 540), rose bengal (RB), and chlorin e6 (Ce6), with the function of absorption of green or red emission, were selected and loaded into the mSiO2 shell of both UCNP@mSiO2 nanomaterials. A comprehensive study for the three UCNP@mSiO2/PS donor/acceptor pairs was performed to investigate the efficacy of fluorescence resonance energy transfer (FRET), ROS generation, and in vitro PDT using a MCF-7 cell line. ROS generation detection showed that as compared to the oleate-capped and ligand-free UCNP/PS pairs, the UCNP@mSiO2/PS nanocarrier system demonstrated more pronounced ROS generation due to the UCNP@mSiO2 nanoparticles in close vicinity to PS molecules and a higher loading capacity of the photosensitizer. As a result, the three LiYF4 UCNP@mSiO2/PS nanoplatforms displayed more prominent therapeutic efficacies in PDT by using in vitro cytotoxicity tests.
Collapse
Affiliation(s)
- Tsung-Han Ho
- Department of Chemical and Materials Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 807, Taiwan
| | - Chien-Hsin Yang
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Zheng-En Jiang
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tzong-Liu Wang
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 811, Taiwan
- Correspondence: ; Tel.: +886-7-5919278
| |
Collapse
|
29
|
Zarepour A, Bal Öztürk A, Koyuncu Irmak D, Yaşayan G, Gökmen A, Karaöz E, Zarepour A, Zarrabi A, Mostafavi E. Combination Therapy Using Nanomaterials and Stem Cells to Treat Spinal Cord Injuries. Eur J Pharm Biopharm 2022; 177:224-240. [PMID: 35850168 DOI: 10.1016/j.ejpb.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
As a part of the central nervous system, the spinal cord (SC) provides most of the communications between the brain and other parts of the body. Any damage to SC interrupts this communication, leading to serious problems, which may remain for the rest of their life. Due to its significant impact on patients' quality of life and its exorbitant medical costs, SC injury (SCI) is known as one of the most challengeable diseases in the world. Thus, it is critical to introduce highly translatable therapeutic platforms for SCI treatment. So far, different strategies have been introduced, among which utilizing various types of stem cells is one of the most interesting ones. The capability of stem cells to differentiate into several types of cell lines makes them promising candidates for the regeneration of injured tissues. One of the other interesting and novel strategies for SCI treatment is the application of nanomaterials, which could appear as a carrier for therapeutic agents or as a platform for culturing the cells. Combining these two approaches, stem cells and nanomaterials, could provide promising therapeutic strategies for SCI management. Accordingly, in this review we have summarized some of the recent advancements in which the applications of different types of stem cells and nanomaterials, alone and in combination forms, were evaluated for SCI treatment.
Collapse
Affiliation(s)
- Arezou Zarepour
- Radiology Department, Kashan University of Medical Sciences, Kashan, Isfahan, Iran
| | - Ayça Bal Öztürk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey; Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Zeytinburnu, Turkey
| | | | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Aylin Gökmen
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Bahcesehir University, Besiktas, Istanbul, Turkey
| | - Erdal Karaöz
- Liv Hospital, Center for Regenerative Medicine and Stem Cell Manufacturing (LivMedCell), İstanbul, Turkey
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Porrang S, Davaran S, Rahemi N, Allahyari S, Mostafavi E. How Advancing are Mesoporous Silica Nanoparticles? A Comprehensive Review of the Literature. Int J Nanomedicine 2022; 17:1803-1827. [PMID: 35498391 PMCID: PMC9043011 DOI: 10.2147/ijn.s353349] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
The application of mesoporous silica nanoparticles (MSNs) is ubiquitous in various sciences. MSNs possess unique features, including the diversity in manufacturing by different synthesis methods and from different sources, structure controllability, pore design capabilities, pore size tunability, nanoparticle size distribution adjustment, and the ability to create diverse functional groups on their surface. These characteristics have led to various types of MSNs as a unique system for drug delivery. In this review, first, the synthesis of MSNs by different methods via using different sources were studied. Then, the parameters affecting their physicochemical properties and functionalization have been discussed. Finally, the last decade's novel strategies, including surface functionalization, drug delivery, and cancer treatment, based on the MSNs in drug delivery and cancer therapy have been addressed.
Collapse
Affiliation(s)
- Sahar Porrang
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Centre for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Rahemi
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Somaiyeh Allahyari
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
31
|
Saghebasl S, Akbarzadeh A, Gorabi AM, Nikzamir N, SeyedSadjadi M, Mostafavi E. Biodegradable functional macromolecules as promising scaffolds for cardiac tissue engineering. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Solmaz Saghebasl
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Abolfazl Akbarzadeh
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Universal Scientific Education and Research Network (USERN) Tabriz Iran
| | - Armita Mahdavi Gorabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Nasrin Nikzamir
- Department of Chemistry, Science and Research Branch Islamic Azad University Tehran Iran
| | | | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford California USA
- Department of Medicine Stanford University School of Medicine Stanford California USA
| |
Collapse
|
32
|
Broad-Spectrum Theranostics and Biomedical Application of Functionalized Nanomaterials. Polymers (Basel) 2022; 14:polym14061221. [PMID: 35335551 PMCID: PMC8956086 DOI: 10.3390/polym14061221] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology is an important branch of science in therapies known as “nanomedicine” and is the junction of various fields such as material science, chemistry, biology, physics, and optics. Nanomaterials are in the range between 1 and 100 nm in size and provide a large surface area to volume ratio; thus, they can be used for various diseases, including cardiovascular diseases, cancer, bacterial infections, and diabetes. Nanoparticles play a crucial role in therapy as they can enhance the accumulation and release of pharmacological agents, improve targeted delivery and ultimately decrease the intensity of drug side effects. In this review, we discussthe types of nanomaterials that have various biomedical applications. Biomolecules that are often conjugated with nanoparticles are proteins, peptides, DNA, and lipids, which can enhance biocompatibility, stability, and solubility. In this review, we focus on bioconjugation and nanoparticles and also discuss different types of nanoparticles including micelles, liposomes, carbon nanotubes, nanospheres, dendrimers, quantum dots, and metallic nanoparticles and their crucial role in various diseases and clinical applications. Additionally, we review the use of nanomaterials for bio-imaging, drug delivery, biosensing tissue engineering, medical devices, and immunoassays. Understandingthe characteristics and properties of nanoparticles and their interactions with the biological system can help us to develop novel strategies for the treatment, prevention, and diagnosis of many diseases including cancer, pulmonary diseases, etc. In this present review, the importance of various kinds of nanoparticles and their biomedical applications are discussed in much detail.
Collapse
|
33
|
Ashrafizadeh M, Saebfar H, Gholami MH, Hushmandi K, Zabolian A, Bikarannejad P, Hashemi M, Daneshi S, Mirzaei S, Sharifi E, Kumar AP, Khan H, Heydari Sheikh Hossein H, Vosough M, Rabiee N, Thakur Kumar V, Makvandi P, Mishra YK, Tay FR, Wang Y, Zarrabi A, Orive G, Mostafavi E. Doxorubicin-loaded graphene oxide nanocomposites in cancer medicine: Stimuli-responsive carriers, co-delivery and suppressing resistance. Expert Opin Drug Deliv 2022; 19:355-382. [PMID: 35152815 DOI: 10.1080/17425247.2022.2041598] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The application of doxorubicin (DOX) in cancer therapy has been limited due to its drug resistance and poor internalization. Graphene oxide (GO) nanostructures have the capacity for DOX delivery while promoting its cytotoxicity in cancer. AREAS COVERED The favorable characteristics of GO nanocomposites, preparation method, and application in cancer therapy are described. Then, DOX resistance in cancer is discussed. The GO-mediated photothermal therapy and DOX delivery for cancer suppression are described. Preparation of stimuli-responsive GO nanocomposites, surface functionalization, hybrid nanoparticles, and theranostic applications are emphasized in DOX chemotherapy. EXPERT OPINION Graphene oxide nanoparticle-based photothermal therapy maximizes the anti-cancer activity of DOX against cancer cells. Apart from DOX delivery, GO nanomaterials are capable of loading anti-cancer agents and genetic tools to minimize drug resistance and enhance the cytolytic impact of DOX in cancer eradication. To enhance DOX accumulation in cancer cells, stimuli-responsive (redox-, light-, enzyme- and pH-sensitive) GO nanoparticles have been developed for DOX delivery. Further development of targeted delivery of DOX-loaded GO nanomaterials against cancer cells may be achieved by surface modification of polymers such as polyethylene glycol, hyaluronic acid, and chitosan. Doxorubicin-loaded GO nanoparticles have demonstrated theranostic potential for simultaneous diagnosis and therapy. Hybridization of GO with other nanocarriers such as silica and gold nanoparticles further broadens their potential anti-cancer therapy applications.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Hamidreza Saebfar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hossein Gholami
- DVM. Graduated, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Pooria Bikarannejad
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Alan Prem Kumar
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.,Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Navid Rabiee
- Department of Chemistry, Sharif University of Technology, Tehran, Iran.,School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Vijay Thakur Kumar
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh EH9 3JG, U.K.,School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, 6400 Sønderborg, Denmark
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, USA
| | - Yuzhuo Wang
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain.,University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHUFundación Eduardo Anitua). Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
34
|
Suwardi A, Wang F, Xue K, Han MY, Teo P, Wang P, Wang S, Liu Y, Ye E, Li Z, Loh XJ. Machine Learning-Driven Biomaterials Evolution. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2102703. [PMID: 34617632 DOI: 10.1002/adma.202102703] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/09/2021] [Indexed: 06/13/2023]
Abstract
Biomaterials is an exciting and dynamic field, which uses a collection of diverse materials to achieve desired biological responses. While there is constant evolution and innovation in materials with time, biomaterials research has been hampered by the relatively long development period required. In recent years, driven by the need to accelerate materials development, the applications of machine learning in materials science has progressed in leaps and bounds. The combination of machine learning with high-throughput theoretical predictions and high-throughput experiments (HTE) has shifted the traditional Edisonian (trial and error) paradigm to a data-driven paradigm. In this review, each type of biomaterial and their key properties and use cases are systematically discussed, followed by how machine learning can be applied in the development and design process. The discussions are classified according to various types of materials used including polymers, metals, ceramics, and nanomaterials, and implants using additive manufacturing. Last, the current gaps and potential of machine learning to further aid biomaterials discovery and application are also discussed.
Collapse
Affiliation(s)
- Ady Suwardi
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - FuKe Wang
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Kun Xue
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Ming-Yong Han
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Peili Teo
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Pei Wang
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Shijie Wang
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Ye Liu
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Enyi Ye
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| |
Collapse
|
35
|
Islam M, Lantada AD, Mager D, Korvink JG. Carbon-Based Materials for Articular Tissue Engineering: From Innovative Scaffolding Materials toward Engineered Living Carbon. Adv Healthc Mater 2022; 11:e2101834. [PMID: 34601815 PMCID: PMC11469261 DOI: 10.1002/adhm.202101834] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 12/14/2022]
Abstract
Carbon materials constitute a growing family of high-performance materials immersed in ongoing scientific technological revolutions. Their biochemical properties are interesting for a wide set of healthcare applications and their biomechanical performance, which can be modulated to mimic most human tissues, make them remarkable candidates for tissue repair and regeneration, especially for articular problems and osteochondral defects involving diverse tissues with very different morphologies and properties. However, more systematic approaches to the engineering design of carbon-based cell niches and scaffolds are needed and relevant challenges should still be overcome through extensive and collaborative research. In consequence, this study presents a comprehensive description of carbon materials and an explanation of their benefits for regenerative medicine, focusing on their rising impact in the area of osteochondral and articular repair and regeneration. Once the state-of-the-art is illustrated, innovative design and fabrication strategies for artificially recreating the cellular microenvironment within complex articular structures are discussed. Together with these modern design and fabrication approaches, current challenges, and research trends for reaching patients and creating social and economic impacts are examined. In a closing perspective, the engineering of living carbon materials is also presented for the first time and the related fundamental breakthroughs ahead are clarified.
Collapse
Affiliation(s)
- Monsur Islam
- Karlsruhe Institute of TechnologyInstitute of Microstructure TechnologyHermann‐von‐Helmholtz‐Platz 1Eggenstein‐Leopoldshafen76344Germany
| | - Andrés Díaz Lantada
- Department of Mechanical EngineeringUniversidad Politécnica de MadridJosé Gutiérrez Abascal 2Madrid28006Spain
| | - Dario Mager
- Karlsruhe Institute of TechnologyInstitute of Microstructure TechnologyHermann‐von‐Helmholtz‐Platz 1Eggenstein‐Leopoldshafen76344Germany
| | - Jan G. Korvink
- Karlsruhe Institute of TechnologyInstitute of Microstructure TechnologyHermann‐von‐Helmholtz‐Platz 1Eggenstein‐Leopoldshafen76344Germany
| |
Collapse
|
36
|
Serna JA, Rueda-Gensini L, Céspedes-Valenzuela DN, Cifuentes J, Cruz JC, Muñoz-Camargo C. Recent Advances on Stimuli-Responsive Hydrogels Based on Tissue-Derived ECMs and Their Components: Towards Improving Functionality for Tissue Engineering and Controlled Drug Delivery. Polymers (Basel) 2021; 13:3263. [PMID: 34641079 PMCID: PMC8512780 DOI: 10.3390/polym13193263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Due to their highly hydrophilic nature and compositional versatility, hydrogels have assumed a protagonic role in the development of physiologically relevant tissues for several biomedical applications, such as in vivo tissue replacement or regeneration and in vitro disease modeling. By forming interconnected polymeric networks, hydrogels can be loaded with therapeutic agents, small molecules, or cells to deliver them locally to specific tissues or act as scaffolds for hosting cellular development. Hydrogels derived from decellularized extracellular matrices (dECMs), in particular, have gained significant attention in the fields of tissue engineering and regenerative medicine due to their inherently high biomimetic capabilities and endowment of a wide variety of bioactive cues capable of directing cellular behavior. However, these hydrogels often exhibit poor mechanical stability, and their biological properties alone are not enough to direct the development of tissue constructs with functional phenotypes. This review highlights the different ways in which external stimuli (e.g., light, thermal, mechanical, electric, magnetic, and acoustic) have been employed to improve the performance of dECM-based hydrogels for tissue engineering and regenerative medicine applications. Specifically, we outline how these stimuli have been implemented to improve their mechanical stability, tune their microarchitectural characteristics, facilitate tissue morphogenesis and enable precise control of drug release profiles. The strategic coupling of the bioactive features of dECM-based hydrogels with these stimulation schemes grants considerable advances in the development of functional hydrogels for a wide variety of applications within these fields.
Collapse
Affiliation(s)
| | | | | | | | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| |
Collapse
|
37
|
Akbari E, Mousazadeh H, Hanifehpour Y, Mostafavi E, Gorabi AM, Nejati K, keyhanvar P, Pazoki-Toroudi H, Mohammadhosseini M, Akbarzadeh A. Co-Loading of Cisplatin and Methotrexate in Nanoparticle-Based PCL-PEG System Enhances Lung Cancer Chemotherapy Effects. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02101-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
38
|
Mostafavi E, Medina-Cruz D, Vernet-Crua A, Chen J, Cholula-Díaz JL, Guisbiers G, Webster TJ. Green nanomedicine: the path to the next generation of nanomaterials for diagnosing brain tumors and therapeutics? Expert Opin Drug Deliv 2021; 18:715-736. [PMID: 33332168 DOI: 10.1080/17425247.2021.1865306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Current brain cancer treatments, based on radiotherapy and chemotherapy, are sometimes successful, but they are not free of drawbacks.Areas covered: Traditional methods for the treatment of brain tumors are discussed here with new solutions presented, among which the application of nanotechnology has demonstrated promising results over the past decade. The traditional synthesis of nanostructures, which relies on the use of physicochemical methodologies are discussed, and their associated concerns in terms of environmental and health impact due to the production of toxic by-products, need for toxic catalysts, and their lack of biocompatibility are presented. An overview of the current situation for treating brain tumors using nanotechnological-based approaches is introduced, and some of the latest advances in the application of green nanomaterials (NMs) for the effective targeting of brain tumors are presented.Expert opinion: Green nanotechnology is introduced as a potential solution to toxic NMs through the application of environmentally friendly and cost-effective protocols using living organisms and biomolecules. The current status of this field, such as those involving clinical trials, is included, and the possible limitations of green-NMs and potential ways to avoid those limitations are discussed so that the field can potentially evolve.
Collapse
Affiliation(s)
- Ebrahim Mostafavi
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.,Stanford Cardiovascular Institute, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - David Medina-Cruz
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Ada Vernet-Crua
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Junjiang Chen
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | | | - Gregory Guisbiers
- Department of Physics and Astronomy, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
39
|
Augustine R, Dan P, Hasan A, Khalaf IM, Prasad P, Ghosal K, Gentile C, McClements L, Maureira P. Stem cell-based approaches in cardiac tissue engineering: controlling the microenvironment for autologous cells. Biomed Pharmacother 2021; 138:111425. [PMID: 33756154 DOI: 10.1016/j.biopha.2021.111425] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/08/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is one of the leading causes of mortality worldwide. Cardiac tissue engineering strategies focusing on biomaterial scaffolds incorporating cells and growth factors are emerging as highly promising for cardiac repair and regeneration. The use of stem cells within cardiac microengineered tissue constructs present an inherent ability to differentiate into cell types of the human heart. Stem cells derived from various tissues including bone marrow, dental pulp, adipose tissue and umbilical cord can be used for this purpose. Approaches ranging from stem cell injections, stem cell spheroids, cell encapsulation in a suitable hydrogel, use of prefabricated scaffold and bioprinting technology are at the forefront in the field of cardiac tissue engineering. The stem cell microenvironment plays a key role in the maintenance of stemness and/or differentiation into cardiac specific lineages. This review provides a detailed overview of the recent advances in microengineering of autologous stem cell-based tissue engineering platforms for the repair of damaged cardiac tissue. A particular emphasis is given to the roles played by the extracellular matrix (ECM) in regulating the physiological response of stem cells within cardiac tissue engineering platforms.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | - Pan Dan
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, Nancy 54500, France; Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | | | - Parvathy Prasad
- International and Inter University Center for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560, India
| | - Kajal Ghosal
- Dr. B. C. Roy College of Pharmacy and AHS, Durgapur 713206, India
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW 2007, Australia; School of Medicine, Faculty of Medicine and Health, University of Sydney, NSW 2000, Australia; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Pablo Maureira
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, Nancy 54500, France
| |
Collapse
|
40
|
Casella A, Panitch A, Leach JK. Endogenous Electric Signaling as a Blueprint for Conductive Materials in Tissue Engineering. Bioelectricity 2021; 3:27-41. [PMID: 34476376 PMCID: PMC8370482 DOI: 10.1089/bioe.2020.0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bioelectricity plays an important role in cell behavior and tissue modulation, but is understudied in tissue engineering research. Endogenous electrical signaling arises from the transmembrane potential inherent to all cells and contributes to many cell behaviors, including migration, adhesion, proliferation, and differentiation. Electrical signals are also involved in tissue development and repair. Synthetic and natural conductive materials are under investigation for leveraging endogenous electrical signaling cues in tissue engineering applications due to their ability to direct cell differentiation, aid in maturing electroactive cell types, and promote tissue functionality. In this review, we provide a brief overview of bioelectricity and its impact on cell behavior, report recent literature using conductive materials for tissue engineering, and discuss opportunities within the field to improve experimental design when using conductive substrates.
Collapse
Affiliation(s)
- Alena Casella
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Surgery and UC Davis Health, Sacramento, California, USA
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| |
Collapse
|
41
|
Kato R, Lettow JH, Patel SN, Rowan SJ. Ion-Conducting Thermoresponsive Films Based on Polymer-Grafted Cellulose Nanocrystals. ACS APPLIED MATERIALS & INTERFACES 2020; 12:54083-54093. [PMID: 33201676 DOI: 10.1021/acsami.0c16059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mechanically robust, thermoresponsive, ion-conducting nanocomposite films are prepared from poly(2-phenylethyl methacrylate)-grafted cellulose nanocrystals (MxG-CNC-g-PPMA). One-component nanocomposite films of the polymer-grafted nanoparticle (PGN) MxG-CNC-g-PPMA are imbibed with 30 wt % imidazolium-based ionic liquid to produce flexible ion-conducting films. These films with 1-hexyl-3-methylimidazolium bis(trifluoromethylsulfonyl)imide (MxG-CNC-g-PPMA/[H]) not only display remarkable improvements in toughness (>25 times) and tensile strength (>70 times) relative to the corresponding films consisting of the ionic liquid imbibed in the two-component CNC/PPMA nanocomposite but also show higher ionic conductivity than the corresponding neat PPMA with the same weight percent of ionic liquid. Notably, the one-component film containing 1-ethyl-3-methylimidazolium bis(trifluoromethylsulfonyl)imide (MxG-CNC-g-PPMA/[E]) exhibits temperature-responsive ionic conduction. The ionic conductivity decreases at around 60 °C as a consequence of the lower critical solution temperature phase transition of the grafted polymer in the ionic liquid, which leads to phase separation. Moreover, holding the MxG-CNC-g-PPMA/[E] film at room temperature for 24 h returns the film to its original homogenous state. These materials exhibit properties relevant to thermal cutoff safety devices (e.g., thermal fuse) where a reduction in conductivity above a critical temperature is needed.
Collapse
|
42
|
Zaszczyńska A, Gradys A, Sajkiewicz P. Progress in the Applications of Smart Piezoelectric Materials for Medical Devices. Polymers (Basel) 2020; 12:E2754. [PMID: 33266424 PMCID: PMC7700596 DOI: 10.3390/polym12112754] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022] Open
Abstract
Smart piezoelectric materials are of great interest due to their unique properties. Piezoelectric materials can transform mechanical energy into electricity and vice versa. There are mono and polycrystals (piezoceramics), polymers, and composites in the group of piezoelectric materials. Recent years show progress in the applications of piezoelectric materials in biomedical devices due to their biocompatibility and biodegradability. Medical devices such as actuators and sensors, energy harvesting devices, and active scaffolds for neural tissue engineering are continually explored. Sensors and actuators from piezoelectric materials can convert flow rate, pressure, etc., to generate energy or consume it. This paper consists of using smart materials to design medical devices and provide a greater understanding of the piezoelectric effect in the medical industry presently. A greater understanding of piezoelectricity is necessary regarding the future development and industry challenges.
Collapse
Affiliation(s)
- Angelika Zaszczyńska
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawinskiego 5b St., 02-106 Warsaw, Poland; (A.G.); (P.S.)
| | | | | |
Collapse
|
43
|
Farokhi M, Mottaghitalab F, Saeb MR, Shojaei S, Zarrin NK, Thomas S, Ramakrishna S. Conductive Biomaterials as Substrates for Neural Stem Cells Differentiation towards Neuronal Lineage Cells. Macromol Biosci 2020; 21:e2000123. [PMID: 33015992 DOI: 10.1002/mabi.202000123] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 08/10/2020] [Indexed: 01/23/2023]
Abstract
The injuries and defects in the central nervous system are the causes of disability and death of an affected person. As of now, there are no clinically available methods to enhance neural structural regeneration and functional recovery of nerve injuries. Recently, some experimental studies claimed that the injuries in brain can be repaired by progenitor or neural stem cells located in the neurogenic sites of adult mammalian brain. Various attempts have been made to construct biomimetic physiological microenvironment for neural stem cells to control their ultimate fate. Conductive materials have been considered as one the best choices for nerve regeneration due to the capacity to mimic the microenvironment of stem cells and regulate the alignment, growth, and differentiation of neural stem cells. The review highlights the use of conductive biomaterials, e.g., polypyrrole, polyaniline, poly(3,4-ethylenedioxythiophene), multi-walled carbon nanotubes, single-wall carbon nanotubes, graphene, and graphite oxide, for controlling the neural stem cells activities in terms of proliferation and neuronal differentiation. The effects of conductive biomaterials in axon elongation and synapse formation for optimal repair of central nervous system injuries are also discussed.
Collapse
Affiliation(s)
- Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Mottaghitalab
- Nanotechnology Research CentreFaculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 14155-6451, Iran
| | | | - Shahrokh Shojaei
- Stem Cells Research CenterTissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, Tehran, Iran.,Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, 1316943551, Iran
| | - Negin Khaneh Zarrin
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Sabu Thomas
- School of Chemical Sciences, MG University, Kottayam, Kerala, 686560, India
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| |
Collapse
|
44
|
Rogers ZJ, Zeevi MP, Koppes R, Bencherif SA. Electroconductive Hydrogels for Tissue Engineering: Current Status and Future Perspectives. Bioelectricity 2020; 2:279-292. [PMID: 34476358 PMCID: PMC8370338 DOI: 10.1089/bioe.2020.0025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Over the past decade, electroconductive hydrogels, integrating both the biomimetic attributes of hydrogels and the electrochemical properties of conductive materials, have gained significant attention. Hydrogels, three-dimensional and swollen hydrophilic polymer networks, are an important class of tissue engineering (TE) scaffolds owing to their microstructural and mechanical properties, ability to mimic the native extracellular matrix, and promote tissue repair. However, hydrogels are intrinsically insulating and therefore unable to emulate the complex electrophysiological microenvironment of cardiac and neural tissues. To overcome this challenge, electroconductive materials, including carbon-based materials, nanoparticles, and polymers, have been incorporated within nonconductive hydrogels to replicate the electrical and biological characteristics of biological tissues. This review gives a brief introduction on the rational design of electroconductive hydrogels and their current applications in TE, especially for neural and cardiac regeneration. The recent progress and development trends of electroconductive hydrogels, their challenges, and clinical translatability, as well as their future perspectives, with a focus on advanced manufacturing technologies, are also discussed.
Collapse
Affiliation(s)
- Zachary J. Rogers
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Michael P. Zeevi
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Ryan Koppes
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Biomechanics and Bioengineering (BMBI), UTC CNRS UMR 7338, University of Technology of Compiègne, Compiègne, France
| |
Collapse
|