1
|
Camacho-Cardenosa M, Pulido-Escribano V, Estrella-Guisado G, Dorado G, Herrera-Martínez AD, Gálvez-Moreno MÁ, Casado-Díaz A. Bioprinted Hydrogels as Vehicles for the Application of Extracellular Vesicles in Regenerative Medicine. Gels 2025; 11:191. [PMID: 40136896 PMCID: PMC11941778 DOI: 10.3390/gels11030191] [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: 02/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Three-dimensional bioprinting is a new advance in tissue engineering and regenerative medicine. Bioprinting allows manufacturing three-dimensional (3D) structures that mimic tissues or organs. The bioinks used are mainly made of natural or synthetic polymers that must be biocompatible, printable, and biodegradable. These bioinks may incorporate progenitor cells, favoring graft implantation and regeneration of injured tissues. However, the natures of biomaterials, bioprinting processes, a lack of vascularization, and immune responses are factors that limit the viability and functionality of implanted cells and the regeneration of damaged tissues. These limitations can be addressed by incorporating extracellular vesicles (EV) into bioinks. Indeed, EV from progenitor cells may have regenerative capacities, being similar to those of their source cells. Therefore, their combinations with biomaterials can be used in cell-free therapies. Likewise, they can complement the manufacture of bioinks by increasing the viability, differentiation, and regenerative ability of incorporated cells. Thus, the main objective of this review is to show how the use of 3D bioprinting technology can be used for the application of EV in regenerative medicine by incorporating these nanovesicles into hydrogels used as bioinks. To this end, the latest advances derived from in vitro and in vivo studies have been described. Together, these studies show the high therapeutic potential of this strategy in regenerative medicine.
Collapse
Affiliation(s)
- Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Guadalupe Estrella-Guisado
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Gabriel Dorado
- Departamento Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain;
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| | - Aura D. Herrera-Martínez
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.C.-C.); (V.P.-E.); (G.E.-G.); (A.D.H.-M.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| |
Collapse
|
2
|
Orr A, Kalantarnia F, Nazir S, Bolandi B, Alderson D, O'Grady K, Hoorfar M, Julian LM, Willerth SM. Recent advances in 3D bioprinted neural models: A systematic review on the applications to drug discovery. Adv Drug Deliv Rev 2025; 218:115524. [PMID: 39900293 DOI: 10.1016/j.addr.2025.115524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/12/2024] [Accepted: 01/26/2025] [Indexed: 02/05/2025]
Abstract
The design of neural tissue models with architectural and biochemical relevance to native tissues opens the way for the fundamental study and development of therapies for many disorders with limited treatment options. Here, we systematically review the most recent literature on 3D bioprinted neural models, including their potential for use in drug screening. Neural tissues that model the central nervous system (CNS) from the relevant literature are reviewed with comprehensive summaries of each study, and discussion of the model types, bioinks and additives, cell types used, bioprinted construct shapes and culture time, and the characterization methods used. In this review, we accentuate the lack of standardization among characterization methods to analyze the functionality (including chemical, metabolic and other pathways) and mechanical relevance of the 3D bioprinted constructs, and discuss this as a critical area for future exploration. These gaps must be addressed for this technology to be applied for effective drug screening applications, despite its enormous potential for rapid and efficient drug screening. The future of biomimetic, 3D printed neural tissues is promising and evaluation of the in vivo relevance on multiple levels should be sought to adequately compare model performance and develop viable treatment options for neurodegenerative diseases, or other conditions that affect the CNS.
Collapse
Affiliation(s)
- Amanda Orr
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | | - Shama Nazir
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Behzad Bolandi
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Dominic Alderson
- Newcastle University Biosciences Institute, Newcastle-Upon-Tyne, NE2 4HH, UK
| | - Kerrin O'Grady
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA
| | - Mina Hoorfar
- Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Lisa M Julian
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; Centre for Advanced Materials and Technology, University of Victoria, Victoria, BC V8W 2Y2, Canada; School of Biomedical Engineering, University of British Columbia, Victoria, BC V6T 1Z4, Canada.
| |
Collapse
|
3
|
Bahraminasab M, Asgharzade S, Doostmohamadi A, Satari A, Hasannejad F, Arab S. Development of a hydrogel-based three-dimensional (3D) glioblastoma cell lines culture as a model system for CD73 inhibitor response study. Biomed Eng Online 2024; 23:127. [PMID: 39709472 DOI: 10.1186/s12938-024-01320-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 11/23/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Despite the development of various therapeutic approaches over the past decades, the treatment of glioblastoma multiforme (GBM) remains a major challenge. The extracellular adenosine-generating enzyme, CD73, is involved in the pathogenesis and progression of GBM, and targeting CD73 may represent a novel approach to treat this cancer. In this study, three-dimensional culture systems based on three hydrogel compositions were characterized and an optimal type was selected to simulate the GBM microenvironment. In addition, the effect of a CD73 inhibitor on GBM cell aggregates and spheroids was investigated as a potential therapeutic approach for this disease. METHODS Rheology measurements, Fourier transform infrared spectroscopy (FT-IR), scanning electron microscopy (SEM) and cell proliferation assays were performed to analyze the synthesized hydrogel and select an optimal formulation. The viability of tumor cells in the optimal hydrogel was examined histologically and by confocal microscopy. In addition, the sensitivity of the tumor cells to the CD73 inhibitor was investigated using a cell proliferation assay and real-time PCR. RESULTS The data showed that the hydrogel containing 5 wt% gelatin and 5 wt% sodium alginate had better rheological properties and higher cell viability. Therefore, it could provide a more suitable environment for GBM cells and better mimic the natural microenvironment. GBM cells treated with CD73 inhibitors significantly decreased the proliferation rate and expression of VEGF and HIF1-α in the optimal hydrogel. CONCLUSION Our current research demonstrates the great potential of CD73 inhibitor for clinical translation of cancer studies by analyzing the behavior and function of 3D tumor cells, and thus for more effective treatment protocols for GBM.
Collapse
Affiliation(s)
- Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Doostmohamadi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Atefeh Satari
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farkhonde Hasannejad
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
- Genetic Department, Breast Cancer Research Center, Moatamed Cancer Institute, ACECR, Tehran, Iran
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
4
|
Lai Y, Xiao X, Huang Z, Duan H, Yang L, Yang Y, Li C, Feng L. Photocrosslinkable Biomaterials for 3D Bioprinting: Mechanisms, Recent Advances, and Future Prospects. Int J Mol Sci 2024; 25:12567. [PMID: 39684279 DOI: 10.3390/ijms252312567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Constructing scaffolds with the desired structures and functions is one of the main goals of tissue engineering. Three-dimensional (3D) bioprinting is a promising technology that enables the personalized fabrication of devices with regulated biological and mechanical characteristics similar to natural tissues/organs. To date, 3D bioprinting has been widely explored for biomedical applications like tissue engineering, drug delivery, drug screening, and in vitro disease model construction. Among different bioinks, photocrosslinkable bioinks have emerged as a powerful choice for the advanced fabrication of 3D devices, with fast crosslinking speed, high resolution, and great print fidelity. The photocrosslinkable biomaterials used for light-based 3D printing play a pivotal role in the fabrication of functional constructs. Herein, this review outlines the general 3D bioprinting approaches related to photocrosslinkable biomaterials, including extrusion-based printing, inkjet printing, stereolithography printing, and laser-assisted printing. Further, the mechanisms, advantages, and limitations of photopolymerization and photoinitiators are discussed. Next, recent advances in natural and synthetic photocrosslinkable biomaterials used for 3D bioprinting are highlighted. Finally, the challenges and future perspectives of photocrosslinkable bioinks and bioprinting approaches are envisaged.
Collapse
Affiliation(s)
- Yushang Lai
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiong Xiao
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziwei Huang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongying Duan
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liping Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchu Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenxi Li
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Feng
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Qiu P, Wang L, Wang J, Wang X, Xu J, An X, Han F, Dong Z, Zhang J, Shi P, Niu Q. Adhesive chitosan-based hybrid biohydrogels for peripheral nerve injury repair. Front Cell Dev Biol 2024; 12:1499766. [PMID: 39610708 PMCID: PMC11602492 DOI: 10.3389/fcell.2024.1499766] [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: 09/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
With the rapid progress of industrialization, the incidence of peripheral nerve injuries caused by trauma has been continuously increasing. These injuries result in a significant number of disabilities and irreversible functional impairments, not only severely impacting the health and quality of life of patients but also placing a heavy economic burden on families and society. Effectively promoting peripheral nerve regeneration has thus become a key focus and challenge in current research. In recent years, hybrid biohydrogels with adhesive properties have gained widespread attention due to their excellent biocompatibility, mechanical stability, conductivity, and biodegradability. These materials can provide an optimal microenvironment to promote neuron adhesion and axonal extension while offering outstanding mechanical strength to meet the fixation requirements in clinical surgeries. This paper systematically reviews the application of adhesive hybrid biohydrogels in peripheral nerve injury repair, highlighting the latest research progress in promoting nerve regeneration and improving functional recovery, and discusses the challenges and future prospects for their clinical application.
Collapse
Affiliation(s)
- Pengjia Qiu
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Lei Wang
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Jing Wang
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Xingdong Wang
- Department of Orthopedics, Sichuan Gemflower Hospital, North Sichuan Medical College, Sichuan, China
| | - Jianchao Xu
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Xiaokai An
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Fengwang Han
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Zhao Dong
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Jiangtao Zhang
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Peiwen Shi
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| | - Qiang Niu
- Department of Orthopedics, Gaoyang County Hospital, Baoding, Hebei Province, China
| |
Collapse
|
6
|
Qavi I, Halder S, Tan G. Optimization of printability of bioinks with multi-response optimization (MRO) and artificial neural networks (ANN). PROGRESS IN ADDITIVE MANUFACTURING 2024. [DOI: 10.1007/s40964-024-00828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/28/2024] [Indexed: 01/06/2025]
|
7
|
Cadena MA, Sing A, Taylor K, Jin L, Ning L, Amoli MS, Singh Y, Lanjewar SN, Tomov ML, Serpooshan V, Sloan SA. A 3D Bioprinted Cortical Organoid Platform for Modeling Human Brain Development. Adv Healthc Mater 2024; 13:e2401603. [PMID: 38815975 PMCID: PMC11518656 DOI: 10.1002/adhm.202401603] [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: 05/03/2024] [Revised: 05/24/2024] [Indexed: 06/01/2024]
Abstract
The ability to promote three-dimensional (3D) self-organization of induced pluripotent stem cells into complex tissue structures called organoids presents new opportunities for the field of developmental biology. Brain organoids have been used to investigate principles of neurodevelopment and neuropsychiatric disorders and serve as a drug screening and discovery platform. However, brain organoid cultures are currently limited by a lacking ability to precisely control their extracellular environment. Here, this work employs 3D bioprinting to generate a high-throughput, tunable, and reproducible scaffold for controlling organoid development and patterning. Additionally, this approach supports the coculture of organoids and vascular cells in a custom architecture containing interconnected endothelialized channels. Printing fidelity and mechanical assessments confirm that fabricated scaffolds closely match intended design features and exhibit stiffness values reflective of the developing human brain. Using organoid growth, viability, cytoarchitecture, proliferation, and transcriptomic benchmarks, this work finds that organoids cultured within the bioprinted scaffold long-term are healthy and have expected neuroectodermal differentiation. Lastly, this work confirms that the endothelial cells (ECs) in printed channel structures can migrate toward and infiltrate into the embedded organoids. This work demonstrates a tunable 3D culturing platform that can be used to create more complex and accurate models of human brain development and underlying diseases.
Collapse
Affiliation(s)
- Melissa A. Cadena
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anson Sing
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kylie Taylor
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Mechanical Engineering, Cleveland State University, Cleveland, OH, United States
| | - Mehdi Salar Amoli
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Yamini Singh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - The Brain Organoid Hub
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Samantha N. Lanjewar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin L. Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, US
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
8
|
Eisenberg MT, Hustedt JW. Alginate Use in Orthopedics and Peripheral Nerve Repair: A Systematic Review. Cureus 2024; 16:e72480. [PMID: 39502971 PMCID: PMC11536484 DOI: 10.7759/cureus.72480] [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] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
The use of alginate, a derivative of seaweed, has been proposed for multiple orthopedic indications. We aimed to review the current use of alginate in orthopedics and to focus on the future applications of alginate for peripheral nerve repair. A comprehensive literature search was performed to identify biomechanical, laboratory, animal, and human studies where alginate has been utilized for orthopedic or nerve repair indications. A systematic review of orthopedic indications was conducted for safety and efficacy, and a specific focus was placed on alginate for use in peripheral nerve repair and reconstruction. Thirty-two studies were identified. Alginate has a strong history and safety profile for usage in orthopedic surgery. Its primary usage has been for the repair of articular cartilage, although it has also been used for disc regeneration of the lumbar spine and for cushioning joints in osteoarthritis. The primary indication in peripheral nerve repair is to create an environment that encourages Schwann cell migration and repair in nerve injuries while blocking fibrotic scar tissue formation by inhibiting the activity of fibroblasts. Alginate hydrogel may serve as a potential conduit for nerve regeneration in nerve injuries with small to medium-sized gaps.
Collapse
Affiliation(s)
- Matthew T Eisenberg
- Orthopedic Surgery, University of Arizona College of Medicine - Phoenix, Phoenix, USA
| | - Joshua W Hustedt
- Hand Surgery, University of Arizona College of Medicine - Phoenix, Phoenix, USA
| |
Collapse
|
9
|
Li QQ, Xu D, Dong QW, Song XJ, Chen YB, Cui YL. Biomedical potentials of alginate via physical, chemical, and biological modifications. Int J Biol Macromol 2024; 277:134409. [PMID: 39097042 DOI: 10.1016/j.ijbiomac.2024.134409] [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: 04/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Alginate is a linear polysaccharide with a modifiable structure and abundant functional groups, offers immense potential for tailoring diverse alginate-based materials to meet the demands of biomedical applications. Given the advancements in modification techniques, it is significant to analyze and summarize the modification of alginate by physical, chemical and biological methods. These approaches provide plentiful information on the preparation, characterization and application of alginate-based materials. Physical modification generally involves blending and physical crosslinking, while chemical modification relies on chemical reactions, mainly including acylation, sulfation, phosphorylation, carbodiimide coupling, nucleophilic substitution, graft copolymerization, terminal modification, and degradation. Chemical modified alginate contains chemically crosslinked alginate, grafted alginate and oligo-alginate. Biological modification associated with various enzymes to realize the hydrolysis or grafting. These diverse modifications hold great promise in fully harnessing the potential of alginate for its burgeoning biomedical applications in the future. In summary, this review provides a comprehensive discussion and summary of different modification methods applied to improve the properties of alginate while expanding its biomedical potentials.
Collapse
Affiliation(s)
- Qiao-Qiao Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xu-Jiao Song
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Yi-Bing Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
10
|
Li Y, Xu C, Zhou X, Li J, Xu S, Tu Y, Mu X, Huang J, Huang Q, Kang L, Wang H, Zhang M, Yuan Y, Wu C, Zhang J. DNA adductomics aided rapid screening of genotoxic impurities using nucleosides and 3D bioprinted human liver organoids. Talanta 2024; 273:125902. [PMID: 38508126 DOI: 10.1016/j.talanta.2024.125902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
Current genotoxicity assessment methods are mainly employed to verify the genotoxic safety of drugs, but do not allow for rapid screening of specific genotoxic impurities (GTIs). In this study, a new approach for the recognition of GTIs has been proposed. It is to expose the complex samples to an in vitro nucleoside incubation model, and then draw complete DNA adduct profiles to infer the structures of potential genotoxic impurities (PGIs). Subsequently, the genotoxicity is confirmed in human by 3D bioprinted human liver organoids. To verify the feasibility of the approach, lansoprazole chloride compound (Lanchlor), a PGI during the synthesis of lansoprazole, was selected as the model drug. After confirming genotoxicity by Comet assay, it was exposed to different models to map and compare the DNA adduct profiles by LC-MS/MS. The results showed Lanchlor could generate diverse DNA adducts, revealing firstly its genotoxicity at molecular mechanism of action. Furthermore, the largest variety and content of DNA adducts were observed in the nucleoside incubation model, while the human liver organoids exhibited similar results with rats. The results showed that the combination of DNA adductomics and 3D bioprinted organoids were useful for the rapid screening of GTIs.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Chen Xu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Xueting Zhou
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Jinhong Li
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Shiting Xu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuanbo Tu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Xue Mu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiajun Huang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Qing Huang
- Devision of Inspection Technology Research, Jiangsu Institute for Food and Drug Control, Nanjing, 210019, China
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, NSW, 2006, Australia
| | - Huaisong Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Mei Zhang
- Devision of Inspection Technology Research, Jiangsu Institute for Food and Drug Control, Nanjing, 210019, China
| | - Yaozuo Yuan
- Devision of Inspection Technology Research, Jiangsu Institute for Food and Drug Control, Nanjing, 210019, China.
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China.
| | - Junying Zhang
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
11
|
Clauzel J, Colitti N, Combeau M, Labriji W, Robert L, Brilhault A, Cirillo C, Desmoulin F, Raymond-Letron I, Loubinoux I. In vivo biocompatibility assessment of 3D printed bioresorbable polymers for brain tissue regeneration. A feasibility study. Regen Ther 2024; 26:941-955. [PMID: 39512739 PMCID: PMC11541680 DOI: 10.1016/j.reth.2024.10.004] [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: 07/23/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction The limited capacity of brain tissue to regenerate after acute injury, hampered by cell death, edema and inflammation, has led to an interest in promising and innovative approaches such as implantable regenerative scaffolds designed to improve brain plasticity. Leveraging the capabilities of bioprinting, these scaffolds can be tailored to match the intricate architecture of the brain. Methods In this methodological study, we performed in vivo biocompatibility assessments after a brain lesion on three distinct bioeliminable or bioresorbable materials: Poly(ethylene glycol) diacrylate (PEGDA), Polycaprolactone (PCL) and a PEGDA mixed with gelatin methacrylate (PEGDA-GelMA). Results A scaffold with a complex shape was printed with patterns, spatial resolution and porosity adapted to cerebral cortex reconstruction. In vivo evaluations were complemented by behavioral monitoring, affirming the safety of these materials. High-resolution T2 MRI imaging effectively captured scaffold structures and demonstrated their non-invasive utility in monitoring degradability. ASL MRI imaging quantified cerebral blood flow and was positively and significantly correlated with lectin immunofluorescent labeling. It may be used to non-invasively monitor progressive revascularization of implants.PEGDA produced an intense foreign-body response, encapsulated by a fibro-inflammatory barrier. On the other hand, PCL provoked a controlled inflammatory reaction and facilitated cell migration into the scaffold, although it induced a fibrotic response around PCL fibers. Conversely, the PEGDA-GelMA composite emerged as a promising candidate for intracerebral implantation. It facilitated the creation of a permissive glial layer, while also inducing neovascularization and attracting neuronal progenitors. Conclusion Behavior, MRI monitoring and histology allowed a thorough following of biomaterial biocompatibility. The collective findings position PEGDA-GelMA as a convincing biomaterial option as a basis for treating severe brain lesions, offering new avenues in the search for effective treatments.
Collapse
Affiliation(s)
- Julien Clauzel
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Nina Colitti
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Maylis Combeau
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Wafae Labriji
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Lorenne Robert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Adrien Brilhault
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Carla Cirillo
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Franck Desmoulin
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Isabelle Raymond-Letron
- LabHPEC, Université de Toulouse, ENVT, Toulouse, France
- Institut Restore, Université de Toulouse, CNRS U-5070, EFS, ENVT, Inserm U1301, Toulouse, France
| | - Isabelle Loubinoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| |
Collapse
|
12
|
Wang Z, Li S, Wu Z, Kang Y, Xie S, Cai Z, Shan X, Li Q. Pulsed electromagnetic field-assisted reduced graphene oxide composite 3D printed nerve scaffold promotes sciatic nerve regeneration in rats. Biofabrication 2024; 16:035013. [PMID: 38604162 DOI: 10.1088/1758-5090/ad3d8a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 04/11/2024] [Indexed: 04/13/2024]
Abstract
Peripheral nerve injuries can lead to sensory or motor deficits that have a serious impact on a patient's mental health and quality of life. Nevertheless, it remains a major clinical challenge to develop functional nerve conduits as an alternative to autologous grafts. We applied reduced graphene oxide (rGO) as a bioactive conductive material to impart electrophysiological properties to a 3D printed scaffold and the application of a pulsed magnetic field to excite the formation of microcurrents and induce nerve regeneration.In vitrostudies showed that the nerve scaffold and the pulsed magnetic field made no effect on cell survival, increased S-100βprotein expression, enhanced cell adhesion, and increased the expression level of nerve regeneration-related mRNAs.In vivoexperiments suggested that the protocol was effective in promoting nerve regeneration, resulting in functional recovery of sciatic nerves in rats, when they were damaged close to that of the autologous nerve graft, and increased expression of S-100β, NF200, and GAP43. These results indicate that rGO composite nerve scaffolds combined with pulsed magnetic field stimulation have great potential for peripheral nerve rehabilitation.
Collapse
Affiliation(s)
- Zichao Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Shijun Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Zongxi Wu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510030, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510030, People's Republic of China
| | - Yifan Kang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Zhigang Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Xiaofeng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| | - Qing Li
- National Center for Stomatology, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- Center of Digital Dentistry, Second Clinical Division, Peking University School and Hospital of Stomatology and National Center of Stomatology, Beijing 100081, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology and NHC Key Laboratory of Digital Stomatology and NMPA Key Laboratory for Dental Materials, Beijing 100081, People's Republic of China
| |
Collapse
|
13
|
Ramesh PA, Sethuraman S, Subramanian A. Multichannel Conduits with Fascicular Complementation: Significance in Long Segmental Peripheral Nerve Injury. ACS Biomater Sci Eng 2024; 10:2001-2021. [PMID: 38487853 DOI: 10.1021/acsbiomaterials.3c01868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Despite the advances in tissue engineering approaches, reconstruction of long segmental peripheral nerve defects remains unsatisfactory. Although autologous grafts with proper fascicular complementation have shown meaningful functional recovery according to the Medical Research Council Classification (MRCC), the lack of donor nerve for such larger defect sizes (>30 mm) has been a serious clinical issue. Further clinical use of hollow nerve conduits is limited to bridging smaller segmental defects of denuded nerve ends (<30 mm). Recently, bioinspired multichannel nerve guidance conduits (NGCs) gained attention as autograft substitutes as they mimic the fascicular connective tissue microarchitecture in promoting aligned axonal outgrowth with desirable innervation for complete sensory and motor function restoration. This review outlines the hierarchical organization of nerve bundles and their significance in the sensory and motor functions of peripheral nerves. This review also emphasizes the major challenges in addressing the longer nerve defects with the role of fascicular arrangement in the multichannel nerve guidance conduits and the need for fascicular matching to accomplish complete functional restoration, especially in treating long segmental nerve defects. Further, currently available fabrication strategies in developing multichannel nerve conduits and their inconsistency in existing preclinical outcomes captured in this review would seed a new process in designing an ideal larger nerve conduit for peripheral nerve repair.
Collapse
Affiliation(s)
- Preethy Amruthavarshini Ramesh
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology SASTRA Deemed University Thanjavur 613 401, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology SASTRA Deemed University Thanjavur 613 401, India
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology SASTRA Deemed University Thanjavur 613 401, India
| |
Collapse
|
14
|
Wang P, Zhu P, Yin W, Wu J, Zhang S. ICA/SDF-1α/PBMSCs loaded onto alginate and gelatin cross-linked scaffolds promote damaged cartilage repair. J Cell Mol Med 2024; 28:e18236. [PMID: 38509746 PMCID: PMC10955157 DOI: 10.1111/jcmm.18236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/13/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
A three-dimensional alginate-coated scaffold (GAIS) was constructed in the present study to showcase the multidifferentiation potential of peripheral blood mesenchymal stem cells (PBMSCs) and to investigate the role and mechanism by which Icariin (ICA)/stromal cell-derived factor (SDF-1α)/PBMSCs promote damaged articular repair. In addition, the ability of ICA, in combination with SDF-1α, to promote the migration and proliferation of stem cells was validated through the utilization of CCK-8 and migration experiments. The combination of ICA and SDF-1α inhibited the differentiation of PBMSCs into cartilage, as demonstrated by in vivo experiments and histological staining. Both PCR and western blot experiments showed that GAIS could upregulate the expression of particular genes in chondrocytes. In comparison to scaffolds devoid of alginate (G0), PBMSCs seeded into GAIS scaffolds exhibited a greater rate of proliferation, and the conditioned medium derived from scaffolds containing SDF-1α enhanced the capacity for cell migration. Moreover, after a 12-week treatment period, GAIS, when successfully transplanted into osteochondral defects of mice, was found to promote cartilage regeneration and repair. The findings, therefore, demonstrate that GAIS enhanced the in vitro capabilities of PBMSCs, including proliferation, migration, homing and chondrogenic differentiation. In addition, ICA and SDF-1α effectively collaborated to support cartilage formation in vivo. Thus, the ICA/SDF-1α/PBMSC-loaded biodegradable alginate-gelatin scaffolds showcase considerable potential for use in cartilage repair.
Collapse
Affiliation(s)
- Pengzhen Wang
- Guangzhou Institute of Traumatic SurgeryGuangzhou Red Cross Hospital of Jinan UniversityGuangzhouGuangdongChina
- Key Laboratory of Regenerative Medicine, Ministry of EducationJinan UniversityGuangzhouGuangdongChina
| | - Pingping Zhu
- Department of NeurologyGuangzhou Red Cross Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Wenhui Yin
- Department of CardiologyGuangzhou Red Cross Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Jian Wu
- Department of OtorhinolaryngologyGuangzhou Red Cross Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Shaoheng Zhang
- Department of CardiologyGuangzhou Red Cross Hospital of Jinan UniversityGuangzhouGuangdongChina
| |
Collapse
|
15
|
Zhou W, Rahman MSU, Sun C, Li S, Zhang N, Chen H, Han CC, Xu S, Liu Y. Perspectives on the Novel Multifunctional Nerve Guidance Conduits: From Specific Regenerative Procedures to Motor Function Rebuilding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307805. [PMID: 37750196 DOI: 10.1002/adma.202307805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury potentially destroys the quality of life by inducing functional movement disorders and sensory capacity loss, which results in severe disability and substantial psychological, social, and financial burdens. Autologous nerve grafting has been commonly used as treatment in the clinic; however, its rare donor availability limits its application. A series of artificial nerve guidance conduits (NGCs) with advanced architectures are also proposed to promote injured peripheral nerve regeneration, which is a complicated process from axon sprouting to targeted muscle reinnervation. Therefore, exploring the interactions between sophisticated NGC complexes and versatile cells during each process including axon sprouting, Schwann cell dedifferentiation, nerve myelination, and muscle reinnervation is necessary. This review highlights the contribution of functional NGCs and the influence of microscale biomaterial architecture on biological processes of nerve repair. Progressive NGCs with chemical molecule induction, heterogenous topographical morphology, electroactive, anisotropic assembly microstructure, and self-powered electroactive and magnetic-sensitive NGCs are also collected, and they are expected to be pioneering features in future multifunctional and effective NGCs.
Collapse
Affiliation(s)
- Weixian Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nuozi Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Charles C Han
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
16
|
Angolkar M, Paramshetti S, Gahtani RM, Al Shahrani M, Hani U, Talath S, Osmani RAM, Spandana A, Gangadharappa HV, Gundawar R. Pioneering a paradigm shift in tissue engineering and regeneration with polysaccharides and proteins-based scaffolds: A comprehensive review. Int J Biol Macromol 2024; 265:130643. [PMID: 38467225 DOI: 10.1016/j.ijbiomac.2024.130643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/16/2024] [Accepted: 03/03/2024] [Indexed: 03/13/2024]
Abstract
In the realm of modern medicine, tissue engineering and regeneration stands as a beacon of hope, offering the promise of restoring form and function to damaged or diseased organs and tissues. Central to this revolutionary field are biological macromolecules-nature's own blueprints for regeneration. The growing interest in bio-derived macromolecules and their composites is driven by their environmentally friendly qualities, renewable nature, minimal carbon footprint, and widespread availability in our ecosystem. Capitalizing on these unique attributes, specific composites can be tailored and enhanced for potential utilization in the realm of tissue engineering (TE). This review predominantly concentrates on the present research trends involving TE scaffolds constructed from polysaccharides, proteins and glycosaminoglycans. It provides an overview of the prerequisites, production methods, and TE applications associated with a range of biological macromolecules. Furthermore, it tackles the challenges and opportunities arising from the adoption of these biomaterials in the field of TE. This review also presents a novel perspective on the development of functional biomaterials with broad applicability across various biomedical applications.
Collapse
Affiliation(s)
- Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Reem M Gahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia.
| | - Mesfer Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia.
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India.
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India.
| | | | - Ravi Gundawar
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.
| |
Collapse
|
17
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [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: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
18
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
19
|
Mohapatra P, Gopikrishnan M, Doss C GP, Chandrasekaran N. How Precise are Nanomedicines in Overcoming the Blood-Brain Barrier? A Comprehensive Review of the Literature. Int J Nanomedicine 2024; 19:2441-2467. [PMID: 38482521 PMCID: PMC10932758 DOI: 10.2147/ijn.s442520] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2025] Open
Abstract
New nanotechnology strategies for enhancing drug delivery in brain disorders have recently received increasing attention from drug designers. The treatment of neurological conditions, including brain tumors, stroke, Parkinson's Disease (PD), and Alzheimer's disease (AD), may be greatly influenced by nanotechnology. Numerous studies on neurodegeneration have demonstrated the effective application of nanomaterials in the treatment of brain illnesses. Nanocarriers (NCs) have made it easier to deliver drugs precisely to where they are needed. Thus, the most effective use of nanomaterials is in the treatment of various brain diseases, as this amplifies the overall impact of medication and emphasizes the significance of nanotherapeutics through gene therapy, enzyme replacement therapy, and blood-barrier mechanisms. Recent advances in nanotechnology have led to the development of multifunctional nanotherapeutic agents, a promising treatment for brain disorders. This novel method reduces the side effects and improves treatment outcomes. This review critically assesses efficient nano-based systems in light of obstacles and outstanding achievements. Nanocarriers that transfer medications across the blood-brain barrier and nano-assisted therapies, including nano-immunotherapy, nano-gene therapy, nano enzyme replacement therapy, scaffolds, and 3D to 6D printing, have been widely explored for the treatment of brain disorders. This study aimed to evaluate existing literature regarding the use of nanotechnology in the development of drug delivery systems that can penetrate the blood-brain barrier (BBB) and deliver therapeutic agents to treat various brain disorders.
Collapse
Affiliation(s)
| | - Mohanraj Gopikrishnan
- Department of Integrative Biology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| | - George Priya Doss C
- Department of Integrative Biology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| | | |
Collapse
|
20
|
Sohrabi M, Hesaraki S, Shahrezaee M, Shams-Khorasani A, Roshanfar F, Glasmacher B, Heinemann S, Xu Y, Makvandi P. Antioxidant flavonoid-loaded nano-bioactive glass bone paste: in vitro apatite formation and flow behavior. NANOSCALE ADVANCES 2024; 6:1011-1022. [PMID: 38298585 PMCID: PMC10825906 DOI: 10.1039/d3na00941f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024]
Abstract
Non-cement pastes in the form of injectable materials have gained considerable attention in non-invasive regenerative medicine. Different osteoconductive bioceramics have been used as the solid phase of these bone pastes. Mesoporous bioactive glass can be used as an alternative bioceramic for paste preparation because of its osteogenic qualities. Plant-derived osteogenic agents can also be used in paste formulation to improve osteogenesis; however, their side effects on physical and physicochemical properties should be investigated. In this study, nano-bioactive glass powder was synthesized by a sol-gel method, loaded with different amounts of quercetin (0, 100, 150, and 200 μM), an antioxidant flavonoid with osteogenesis capacity. The loaded powder was then homogenized with a mixture of hyaluronic acid and sodium alginate solution to form a paste. We subsequently evaluated the rheological behavior, injectability, washout resistance, and in vitro bioactivity of the quercetin-loaded pastes. The washout resistance was found to be more than 96% after 14 days of immersion in simulated body fluid (SBF) as well as tris-buffered and citric acid-buffered solutions at 25 °C and 37 °C. All pastes exhibited viscoelastic behavior, in which the elastic modulus exceeded the viscous modulus. The pastes displayed shear-thinning behavior, in which viscosity was more influenced by angular frequency when the quercetin content increased. Results indicated that injectability was much improved using quercetin and the injection force was in the range 20-150 N. Following 14 days of SBF soaking, the formation of a nano-structured apatite phase on the surfaces of quercetin-loaded pastes was confirmed through scanning electron microscopy, X-ray diffractometry, and Fourier-transform infrared spectroscopy. Overall, quercetin, an antioxidant flavonoid osteogenic agent, can be loaded onto the nano-bioactive glass/hyaluronic acid/sodium alginate paste system to enhance injectability, rheological properties, and bioactivity.
Collapse
Affiliation(s)
- Mehri Sohrabi
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center Alborz Iran
| | - Saeed Hesaraki
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center Alborz Iran
| | | | - Alireza Shams-Khorasani
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center Alborz Iran
| | - Fahimeh Roshanfar
- Institute for Multiphase Processes (IMP), Leibniz University Hannover 30823 Garbsen Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) 30625 Hannover Germany
| | - Brigit Glasmacher
- Institute for Multiphase Processes (IMP), Leibniz University Hannover 30823 Garbsen Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) 30625 Hannover Germany
| | | | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital Quzhou China
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital 324000 Quzhou Zhejiang China
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University Rajpura-140401 Punjab India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University Chennai 600077 India
| |
Collapse
|
21
|
He W, Deng J, Ma B, Tao K, Zhang Z, Ramakrishna S, Yuan W, Ye T. Recent Advancements of Bioinks for 3D Bioprinting of Human Tissues and Organs. ACS APPLIED BIO MATERIALS 2024; 7:17-43. [PMID: 38091514 DOI: 10.1021/acsabm.3c00806] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
3D bioprinting is recognized as a promising biomanufacturing technology that enables the reproducible and high-throughput production of tissues and organs through the deposition of different bioinks. Especially, bioinks based on loaded cells allow for immediate cellularity upon printing, providing opportunities for enhanced cell differentiation for organ manufacturing and regeneration. Thus, extensive applications have been found in the field of tissue engineering. The performance of the bioinks determines the functionality of the entire printed construct throughout the bioprinting process. It is generally expected that bioinks should support the encapsulated cells to achieve their respective cellular functions and withstand normal physiological pressure exerted on the printed constructs. The bioinks should also exhibit a suitable printability for precise deposition of the constructs. These characteristics are essential for the functional development of tissues and organs in bioprinting and are often achieved through the combination of different biomaterials. In this review, we have discussed the cutting-edge outstanding performance of different bioinks for printing various human tissues and organs in recent years. We have also examined the current status of 3D bioprinting and discussed its future prospects in relieving or curing human health problems.
Collapse
Affiliation(s)
- Wen He
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jinjun Deng
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Binghe Ma
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Tao
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhi Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, National University of Singapore, Singapore 117576, Singapore
| | - Weizheng Yuan
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Ye
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
22
|
Khalvandi A, Tayebi L, Kamarian S, Saber-Samandari S, Song JI. Data-driven supervised machine learning to predict the compressive response of porous PVA/Gelatin hydrogels and in-vitro assessments: Employing design of experiments. Int J Biol Macromol 2023; 253:126906. [PMID: 37716655 DOI: 10.1016/j.ijbiomac.2023.126906] [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: 05/19/2023] [Revised: 07/01/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
The purpose of this study is to design and evaluate a series of porous hydrogels by considering three independent variables using the Box-Behnken method. Accordingly, concentrations of the constituent macromolecules of the hydrogels, Polyvinyl Alcohol and Gelatin, and concentration of the crosslinking agent are varied to fabricate sixteen different porous samples utilizing the lyophilization process. Subsequently, the porous hydrogels are subjected to a battery of tests, including Fourier Transform Infrared spectroscopy, morphology assessment, pore-size study, porosimetry, uniaxial compression, and swelling measurements. Additionally, in-vitro cell assessments are performed by culturing mouse fibroblast cells (L-929) on the hydrogels, where viability, proliferation, adhesion, and morphology of the L-929 cells are monitored over 24, 48, and 72 h to evaluate the biocompatibility of these biomaterials. To better understand the mechanical behavior of the hydrogels under compressive loadings, Deep Neural Networks (DNNs) are implemented to predict and capture their compressive stress-strain responses as a function of the constituent materials' concentrations and duration of the performed mechanical tests. Overall, this study emphasizes the importance of considering multiple variables in the design of porous hydrogels, provides a comprehensive evaluation of their mechanical and biological properties, and, particularly, implements DNNs in the prediction of the hydrogels' stress-strain responses.
Collapse
Affiliation(s)
- Ali Khalvandi
- Composites Research Laboratory (CRLab), Amirkabir University of Technology, Tehran, Iran; New Technologies Research Center, Amirkabir University of Technology, Tehran, Iran; Department of Mechanical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI 53233, United States
| | - Saeed Kamarian
- Mechanical Engineering Department, Changwon National University, Changwon, Republic of Korea
| | - Saeed Saber-Samandari
- Composites Research Laboratory (CRLab), Amirkabir University of Technology, Tehran, Iran; New Technologies Research Center, Amirkabir University of Technology, Tehran, Iran.
| | - Jung-Il Song
- Mechanical Engineering Department, Changwon National University, Changwon, Republic of Korea
| |
Collapse
|
23
|
Kale N, Edvall C, Ozoude C, Mallik S. In Vitro Tumor Mimetic Spheroid Model: Void Space within a Self-Detachable Cross-Linked Hydrogel. ACS APPLIED BIO MATERIALS 2023; 6:4682-4693. [PMID: 37867293 DOI: 10.1021/acsabm.3c00490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
The three-dimensional (3D) spheroid cell culture model is crucial in screening anticancer drugs in vitro and understanding tumor cell behavior. However, the current in vitro models require highly skilled techniques. Here, we present an in vitro, tumor-mimetic, self-detachable, cancer cell spheroid model that provides the confined space of a tumor microenvironment, convenient spheroid retrieval, immunostaining, treatment, and imaging. We formed a void space within alginate macrobeads by ionic disintegration at a specific region inside. The macrobeads were further destabilized with bovine serum albumin to retrieve the spheroid cultured within the void space. Quantitative analysis of the immunofluorescence images of the cultured spheroids showed enhanced expressions of the hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase-9 (CA-9), like monolayer cultures of cancer cells under hypoxic conditions (0.2% oxygen). Furthermore, adding CoCl2 to the cell culture media induces even higher amounts of HIF-1α and CA-9 in the cultured spheroids. In conclusion, the present work highlighted the in vitro spheroid model, which is closer to the tumor microenvironment and has user-friendly cell seeding, spheroid retrieval, and immunostaining steps.
Collapse
Affiliation(s)
- Narendra Kale
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Connor Edvall
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Chukwuebuka Ozoude
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Sanku Mallik
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
24
|
Liu Y, Yao X, Fan C, Zhang G, Luo X, Qian Y. Microfabrication and lab-on-a-chip devices promote in vitromodeling of neural interfaces for neuroscience researches and preclinical applications. Biofabrication 2023; 16:012002. [PMID: 37832555 DOI: 10.1088/1758-5090/ad032a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 10/13/2023] [Indexed: 10/15/2023]
Abstract
Neural tissues react to injuries through the orchestration of cellular reprogramming, generating specialized cells and activating gene expression that helps with tissue remodeling and homeostasis. Simplified biomimetic models are encouraged to amplify the physiological and morphological changes during neural regeneration at cellular and molecular levels. Recent years have witnessed growing interest in lab-on-a-chip technologies for the fabrication of neural interfaces. Neural system-on-a-chip devices are promisingin vitromicrophysiological platforms that replicate the key structural and functional characteristics of neural tissues. Microfluidics and microelectrode arrays are two fundamental techniques that are leveraged to address the need for microfabricated neural devices. In this review, we explore the innovative fabrication, mechano-physiological parameters, spatiotemporal control of neural cell cultures and chip-based neurogenesis. Although the high variability in different constructs, and the restriction in experimental and analytical access limit the real-life applications of microphysiological models, neural system-on-a-chip devices have gained considerable translatability for modeling neuropathies, drug screening and personalized therapy.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Guifeng Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xi Luo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| |
Collapse
|
25
|
Ciulla MG, Massironi A, Sugni M, Ensign MA, Marzorati S, Forouharshad M. Recent Advances in the Development of Biomimetic Materials. Gels 2023; 9:833. [PMID: 37888406 PMCID: PMC10606425 DOI: 10.3390/gels9100833] [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: 09/26/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
In this review, we focused on recent efforts in the design and development of materials with biomimetic properties. Innovative methods promise to emulate cell microenvironments and tissue functions, but many aspects regarding cellular communication, motility, and responsiveness remain to be explained. We photographed the state-of-the-art advancements in biomimetics, and discussed the complexity of a "bottom-up" artificial construction of living systems, with particular highlights on hydrogels, collagen-based composites, surface modifications, and three-dimensional (3D) bioprinting applications. Fast-paced 3D printing and artificial intelligence, nevertheless, collide with reality: How difficult can it be to build reproducible biomimetic materials at a real scale in line with the complexity of living systems? Nowadays, science is in urgent need of bioengineering technologies for the practical use of bioinspired and biomimetics for medicine and clinics.
Collapse
Affiliation(s)
- Maria G. Ciulla
- Department of Chemistry, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy
| | - Alessio Massironi
- Department of Environmental Science and Policy, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Michela Sugni
- Department of Environmental Science and Policy, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Matthew A. Ensign
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Stefania Marzorati
- Department of Environmental Science and Policy, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Mahdi Forouharshad
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
26
|
Zheng Y, Shariati K, Ghovvati M, Vo S, Origer N, Imahori T, Kaneko N, Annabi N. Hemostatic patch with ultra-strengthened mechanical properties for efficient adhesion to wet surfaces. Biomaterials 2023; 301:122240. [PMID: 37480758 DOI: 10.1016/j.biomaterials.2023.122240] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 07/24/2023]
Abstract
Controlling traumatic bleeding from damaged internal organs while effectively sealing the wound is critical for saving the lives of patients. Existing bioadhesives suffer from blood incompatibility, insufficient adhesion to wet surfaces, weak mechanical properties, and complex application procedures. Here, we engineered a ready-to-use hemostatic bioadhesive with ultra-strengthened mechanical properties and fatigue resistance, robust adhesion to wet tissues within a few seconds of gentle pressing, deformability to accommodate physiological function and action, and the ability to stop bleeding efficiently. The engineered hydrogel, which demonstrated high elasticity (>900%) and toughness (>4600 kJ/m3), was formed by fine-tuning a series of molecular interactions and crosslinking mechanisms involving N-hydroxysuccinimide (NHS) conjugated alginate (Alg-NHS), poly (ethylene glycol) diacrylate (PEGDA), tannic acid (TA), and Fe3+ ions. Dual adhesive moieties including mussel-inspired pyrogallol/catechol and NHS synergistically enhanced wet tissue adhesion (>400 kPa in a wound closure test). In conjunction with physical sealing, the high affinity of TA/Fe3+ for blood could further augment hemostasis. The engineered bioadhesive demonstrated excellent in vitro and in vivo biocompatibility as well as improved hemostatic efficacy as compared to commercial Surgicel®. Overall, the hydrogel design strategy described herein holds great promise for overcoming existing obstacles impeding clinical translation of engineered hemostatic bioadhesives.
Collapse
Affiliation(s)
- Yuting Zheng
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kaavian Shariati
- David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Steven Vo
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nolan Origer
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Taichiro Imahori
- Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Naoki Kaneko
- Division of Interventional Neuroradiology, Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
27
|
Sanaei K, Zamanian A, Mashayekhan S, Ramezani T. Formulation and Characterization of a Novel Oxidized Alginate-Gelatin-Silk Fibroin Bioink with the Aim of Skin Regeneration. IRANIAN BIOMEDICAL JOURNAL 2023; 27:280-93. [PMID: 37873644 PMCID: PMC10707813 DOI: 10.61186/ibj.27.5.280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/21/2023] [Indexed: 12/17/2023]
Abstract
Background In the present study, a novel bioink was suggested based on the oxidized alginate (OAlg), gelatin (GL), and silk fibroin (SF) hydrogels. Methods The composition of the bioink was optimized by the rheological and printability measurements, and the extrusion-based 3D bioprinting process was performed by applying the optimum OAlg-based bioink. Results The results demonstrated that the viscosity of bioink was continuously decreased by increasing the SF/GL ratio, and the bioink displayed a maximum achievable printability (92 ± 2%) at 2% (w/v) of SF and 4% (w/v) of GL. Moreover, the cellular behavior of the scaffolds investigated by MTT assay and live/dead staining confirmed the biocompatibility of the prepared bioink. Conclusion The bioprinted OAlg-GL-SF scaffold could have the potential for using in skin tissue engineering applications, which needs further exploration.
Collapse
Affiliation(s)
- Khadijeh Sanaei
- Department of Nanotechnology and Advanced Materials, Materials and Energy Research Center, Karaj, Iran
| | - Ali Zamanian
- Department of Nanotechnology and Advanced Materials, Materials and Energy Research Center, Karaj, Iran
| | - Shohreh Mashayekhan
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Tayebe Ramezani
- Faculty of biological sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
28
|
Magill E, Demartis S, Gavini E, Permana AD, Thakur RRS, Adrianto MF, Waite D, Glover K, Picco CJ, Korelidou A, Detamornrat U, Vora LK, Li L, Anjani QK, Donnelly RF, Domínguez-Robles J, Larrañeta E. Solid implantable devices for sustained drug delivery. Adv Drug Deliv Rev 2023; 199:114950. [PMID: 37295560 DOI: 10.1016/j.addr.2023.114950] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Implantable drug delivery systems (IDDS) are an attractive alternative to conventional drug administration routes. Oral and injectable drug administration are the most common routes for drug delivery providing peaks of drug concentrations in blood after administration followed by concentration decay after a few hours. Therefore, constant drug administration is required to keep drug levels within the therapeutic window of the drug. Moreover, oral drug delivery presents alternative challenges due to drug degradation within the gastrointestinal tract or first pass metabolism. IDDS can be used to provide sustained drug delivery for prolonged periods of time. The use of this type of systems is especially interesting for the treatment of chronic conditions where patient adherence to conventional treatments can be challenging. These systems are normally used for systemic drug delivery. However, IDDS can be used for localised administration to maximise the amount of drug delivered within the active site while reducing systemic exposure. This review will cover current applications of IDDS focusing on the materials used to prepare this type of systems and the main therapeutic areas of application.
Collapse
Affiliation(s)
- Elizabeth Magill
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Sara Demartis
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari, 07100, Italy
| | - Elisabetta Gavini
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, 07100, Italy
| | - Andi Dian Permana
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Hasanuddin, Makassar 90245, Indonesia
| | - Raghu Raj Singh Thakur
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK; Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Muhammad Faris Adrianto
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK; Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Airlangga University, Surabaya, East Java 60115, Indonesia
| | - David Waite
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK; Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Katie Glover
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Camila J Picco
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Anna Korelidou
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Usanee Detamornrat
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Linlin Li
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Qonita Kurnia Anjani
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK; Fakultas Farmasi, Universitas Megarezky, Jl. Antang Raya No. 43, Makassar 90234, Indonesia
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK
| | - Juan Domínguez-Robles
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK; Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain.
| | - Eneko Larrañeta
- School of Pharmacy, Queen's University Belfast, 97, Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
29
|
Glover K, Mathew E, Pitzanti G, Magee E, Lamprou DA. 3D bioprinted scaffolds for diabetic wound-healing applications. Drug Deliv Transl Res 2023; 13:2096-2109. [PMID: 35018558 PMCID: PMC10315349 DOI: 10.1007/s13346-022-01115-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 11/25/2022]
Abstract
The treatment strategy required for the effective healing of diabetic foot ulcer (DFU) is a complex process that is requiring several combined therapeutic approaches. As a result, there is a significant clinical and economic burden associated in treating DFU. Furthermore, these treatments are often unsuccessful, commonly resulting in lower-limb amputation. The use of drug-loaded scaffolds to treat DFU has previously been investigated using electrospinning and fused deposition modelling (FDM) 3D printing techniques; however, the rapidly evolving field of bioprinting is creating new opportunities for innovation within this research area. In this study, 3D-bioprinted scaffolds with different designs have been fabricated for the delivery of an antibiotic (levoflocixin) to DFU. The scaffolds were fully characterised by a variety of techniques (e.g. SEM, DSC/TGA, FTIR, and mechanical characterisation), demonstrating excellent mechanical properties and providing sustained drug release for 4 weeks. This proof of concept study demonstrates the innovative potential of bioprinting technologies in fabrication of antibiotic scaffolds for the treatment of DFU.
Collapse
Affiliation(s)
- Katie Glover
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Essyrose Mathew
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Giulia Pitzanti
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Erin Magee
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Dimitrios A Lamprou
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
30
|
Asim S, Tabish TA, Liaqat U, Ozbolat IT, Rizwan M. Advances in Gelatin Bioinks to Optimize Bioprinted Cell Functions. Adv Healthc Mater 2023; 12:e2203148. [PMID: 36802199 PMCID: PMC10330013 DOI: 10.1002/adhm.202203148] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/31/2023] [Indexed: 02/21/2023]
Abstract
Gelatin is a widely utilized bioprinting biomaterial due to its cell-adhesive and enzymatically cleavable properties, which improve cell adhesion and growth. Gelatin is often covalently cross-linked to stabilize bioprinted structures, yet the covalently cross-linked matrix is unable to recapitulate the dynamic microenvironment of the natural extracellular matrix (ECM), thereby limiting the functions of bioprinted cells. To some extent, a double network bioink can provide a more ECM-mimetic, bioprinted niche for cell growth. More recently, gelatin matrices are being designed using reversible cross-linking methods that can emulate the dynamic mechanical properties of the ECM. This review analyzes the progress in developing gelatin bioink formulations for 3D cell culture, and critically analyzes the bioprinting and cross-linking techniques, with a focus on strategies to optimize the functions of bioprinted cells. This review discusses new cross-linking chemistries that recapitulate the viscoelastic, stress-relaxing microenvironment of the ECM, and enable advanced cell functions, yet are less explored in engineering the gelatin bioink. Finally, this work presents the perspective on the areas of future research and argues that the next generation of gelatin bioinks should be designed by considering cell-matrix interactions, and bioprinted constructs should be validated against currently established 3D cell culture standards to achieve improved therapeutic outcomes.
Collapse
Affiliation(s)
- Saad Asim
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931 USA
| | - Tanveer A. Tabish
- Cardiovascular Division, Radcliff Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Usman Liaqat
- Department of Materials Engineering, School of Chemical and Materials Engineering (SCME), National University of Sciences & Technology (NUST), Pakistan
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics, Penn State, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State, University Park, PA 16802, USA
- Department of Neurosurgery, Penn State, Hershey, PA 16802, USA
- Department of Medical Oncology, Cukurova University, Adana 01330, Turkey
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931 USA
- Health Research Institute, Michigan Technological University, Houghton, MI, 49931 USA
| |
Collapse
|
31
|
Shokrani H, Shokrani A, Seidi F, Mashayekhi M, Kar S, Nedeljkovic D, Kuang T, Saeb MR, Mozafari M. Polysaccharide-based biomaterials in a journey from 3D to 4D printing. Bioeng Transl Med 2023; 8:e10503. [PMID: 37476065 PMCID: PMC10354780 DOI: 10.1002/btm2.10503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/31/2023] [Accepted: 02/18/2023] [Indexed: 07/22/2023] Open
Abstract
3D printing is a state-of-the-art technology for the fabrication of biomaterials with myriad applications in translational medicine. After stimuli-responsive properties were introduced to 3D printing (known as 4D printing), intelligent biomaterials with shape configuration time-dependent character have been developed. Polysaccharides are biodegradable polymers sensitive to several physical, chemical, and biological stimuli, suited for 3D and 4D printing. On the other hand, engineering of mechanical strength and printability of polysaccharide-based scaffolds along with their aneural, avascular, and poor metabolic characteristics need to be optimized varying printing parameters. Multiple disciplines such as biomedicine, chemistry, materials, and computer sciences should be integrated to achieve multipurpose printable biomaterials. In this work, 3D and 4D printing technologies are briefly compared, summarizing the literature on biomaterials engineering though printing techniques, and highlighting different challenges associated with 3D/4D printing, as well as the role of polysaccharides in the technological shift from 3D to 4D printing for translational medicine.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Jiangsu Co‐Innovation Center for Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and MaterialsNanjing Forestry UniversityNanjingChina
- Department of Chemical EngineeringSharif University of TechnologyTehranIran
| | | | - Farzad Seidi
- Jiangsu Co‐Innovation Center for Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and MaterialsNanjing Forestry UniversityNanjingChina
| | | | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle EastKuwait
| | - Dragutin Nedeljkovic
- College of Engineering and Technology, American University of the Middle EastKuwait
| | - Tairong Kuang
- College of Material Science and Engineering, Zhejiang University of TechnologyHangzhouChina
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
32
|
Mullis AS, Kaplan DL. Functional bioengineered tissue models of neurodegenerative diseases. Biomaterials 2023; 298:122143. [PMID: 37146365 PMCID: PMC10209845 DOI: 10.1016/j.biomaterials.2023.122143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023]
Abstract
Aging-associated neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases remain poorly understood and no disease-modifying treatments exist despite decades of investigation. Predominant in vitro (e.g., 2D cell culture, organoids) and in vivo (e.g., mouse) models of these diseases are insufficient mimics of human brain tissue structure and function and of human neurodegenerative pathobiology, and have thus contributed to this collective translational failure. This has been a longstanding challenge in the field, and new strategies are required to address both fundamental and translational needs. Bioengineered tissue culture models constitute a class of promising alternatives, as they can overcome the low cell density, poor nutrient exchange, and long term culturability limitations of existing in vitro models. Further, they can reconstruct the structural, mechanical, and biochemical cues of native brain tissue, providing a better mimic of human brain tissues for in vitro pathobiological investigation and drug development. We discuss bioengineering techniques for the generation of these neurodegenerative tissue models, including biomaterials-, organoid-, and microfluidics-based approaches, and design considerations for their construction. To aid the development of the next generation of functional neurodegenerative disease models, we discuss approaches to incorporate greater cellular diversity and simulate aging processes within bioengineered brain tissues.
Collapse
Affiliation(s)
- Adam S Mullis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA; Allen Discovery Center, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
33
|
Zennifer A, Thangadurai M, Sundaramurthi D, Sethuraman S. Additive manufacturing of peripheral nerve conduits - Fabrication methods, design considerations and clinical challenges. SLAS Technol 2023; 28:102-126. [PMID: 37028493 DOI: 10.1016/j.slast.2023.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
Tissue-engineered nerve guidance conduits (NGCs) are a viable clinical alternative to autografts and allografts and have been widely used to treat peripheral nerve injuries (PNIs). Although these NGCs are successful to some extent, they cannot aid in native regeneration by improving native-equivalent neural innervation or regrowth. Further, NGCs exhibit longer recovery period and high cost limiting their clinical applications. Additive manufacturing (AM) could be an alternative to the existing drawbacks of the conventional NGCs fabrication methods. The emergence of the AM technique has offered ease for developing personalized three-dimensional (3D) neural constructs with intricate features and higher accuracy on a larger scale, replicating the native feature of nerve tissue. This review introduces the structural organization of peripheral nerves, the classification of PNI, and limitations in clinical and conventional nerve scaffold fabrication strategies. The principles and advantages of AM-based techniques, including the combinatorial approaches utilized for manufacturing 3D nerve conduits, are briefly summarized. This review also outlines the crucial parameters, such as the choice of printable biomaterials, 3D microstructural design/model, conductivity, permeability, degradation, mechanical property, and sterilization required to fabricate large-scale additive-manufactured NGCs successfully. Finally, the challenges and future directions toward fabricating the 3D-printed/bioprinted NGCs for clinical translation are also discussed.
Collapse
Affiliation(s)
- Allen Zennifer
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Madhumithra Thangadurai
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
34
|
Ma W, Liu X, Yang M, Hong Q, Meng L, Zhang Q, Chen J, Pan C. Fabrication of CO-releasing surface to enhance the blood compatibility and endothelialization of TiO 2 nanotubes on titanium surface. BIOMATERIALS ADVANCES 2023; 149:213393. [PMID: 36966654 DOI: 10.1016/j.bioadv.2023.213393] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 05/02/2023]
Abstract
Although the construction of nanotube arrays with the micro-nano structures on the titanium surfaces has demonstrated a great promise in the field of blood-contacting materials and devices, the limited surface hemocompatibility and delayed endothelial healing should be further improved. Carbon monoxide (CO) gas signaling molecule within the physiological concentrations has excellent anticoagulation and the ability to promote endothelial growth, exhibiting the great potential for the blood-contact biomaterials, especially the cardiovascular devices. In this study, the regular titanium dioxide nanotube arrays were firstly prepared in situ on the titanium surface by anodic oxidation, followed by the immobilization of the complex of sodium alginate/carboxymethyl chitosan (SA/CS) on the self-assembled modified nanotube surface, the CO-releasing molecule (CORM-401) was finally grafted onto the surface to create a CO-releasing bioactive surface to enhance the biocompatibility. The results of scanning electron microscopy (SEM), X-ray energy dispersion spectroscopy (EDS), and X-ray photoelectron spectroscopy (XPS) revealed that the CO-releasing molecules were successfully immobilized on the surface. The modified nanotube arrays not only exhibited excellent hydrophilicity but also could slowly release CO gas molecules, and the amount of CO release increased when cysteine was added. Furthermore, the nanotube array can promote albumin adsorption while inhibit fibrinogen adsorption to some extent, demonstrating its selective albumin adsorption; although this effect was somewhat reduced by the introduction of CORM-401, it can be significantly enhanced by the catalytic release of CO. The results of hemocompatibility and endothelial cell growth behaviors showed that, as compared with the CORM-401 modified sample, although the SA/CS-modified sample had better biocompatibility, in the case of cysteine-catalyzed CO release, the released CO could not only reduce the platelet adhesion and activation as well as hemolysis rate, but also promote endothelial cell adhesion and proliferation as well as vascular endothelial growth factor (VEGF) and nitric oxide (NO) expression. As a result, the research of the present study demonstrated that the releasing CO from TiO2 nanotubes can simultaneously enhance the surface hemocompatibility and endothelialization, which could open a new route to enhance the biocompatibility of the blood-contacting materials and devices, such as the artificial heart valve and cardiovascular stents.
Collapse
Affiliation(s)
- Wenfu Ma
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Xuhui Liu
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Minhui Yang
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qingxiang Hong
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Lingjie Meng
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qiuyang Zhang
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China.
| | - Jie Chen
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Changjiang Pan
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China.
| |
Collapse
|
35
|
Modification, 3D printing process and application of sodium alginate based hydrogels in soft tissue engineering: A review. Int J Biol Macromol 2023; 232:123450. [PMID: 36709808 DOI: 10.1016/j.ijbiomac.2023.123450] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Sodium alginate (SA) is an inexpensive and biocompatible biomaterial with fast and gentle crosslinking that has been widely used in biological soft tissue repair/regeneration. Especially with the advent of 3D bioprinting technology, SA hydrogels have been applied more deeply in tissue engineering due to their excellent printability. Currently, the research on material modification, molding process and application of SA-based composite hydrogels has become a hot topic in tissue engineering, and a lot of fruitful results have been achieved. To better help readers have a comprehensive understanding of the development status of SA based hydrogels and their molding process in tissue engineering, in this review, we summarized SA modification methods, and provided a comparative analysis of the characteristics of various SA based hydrogels. Secondly, various molding methods of SA based hydrogels were introduced, the processing characteristics and the applications of different molding methods were analyzed and compared. Finally, the applications of SA based hydrogels in tissue engineering were reviewed, the challenges in their applications were also analyzed, and the future research directions were prospected. We believe this review is of great helpful for the researchers working in biomedical and tissue engineering.
Collapse
|
36
|
A Review of the Benefits 3D Printing Brings to Patients with Neurological Diseases. Pharmaceutics 2023; 15:pharmaceutics15030892. [PMID: 36986752 PMCID: PMC10051330 DOI: 10.3390/pharmaceutics15030892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
This interdisciplinary review focuses on how flexible three-dimensional printing (3DP) technology can aid patients with neurological diseases. It covers a wide variety of current and possible applications ranging from neurosurgery to customizable polypill along with a brief description of the various 3DP techniques. The article goes into detail about how 3DP technology can aid delicate neurosurgical planning and its consequent outcome for patients. It also covers areas such as how the 3DP model can be utilized in patient counseling along with designing specific implants involved in cranioplasty and customization of a specialized instrument such as 3DP optogenetic probes. Furthermore, the review includes how a 3DP nasal cast can contribute to the development of nose-to-brain drug delivery along with looking into how bioprinting could be used for regenerating nerves and how 3D-printed drugs could offer practical benefits to patients suffering from neurological diseases via polypill.
Collapse
|
37
|
Samadi A, Moammeri A, Pourmadadi M, Abbasi P, Hosseinpour Z, Farokh A, Shamsabadipour A, Heydari M, Mohammadi MR. Cell Encapsulation and 3D Bioprinting for Therapeutic Cell Transplantation. ACS Biomater Sci Eng 2023; 9:1862-1890. [PMID: 36877212 DOI: 10.1021/acsbiomaterials.2c01183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
The promise of cell therapy has been augmented by introducing biomaterials, where intricate scaffold shapes are fabricated to accommodate the cells within. In this review, we first discuss cell encapsulation and the promising potential of biomaterials to overcome challenges associated with cell therapy, particularly cellular function and longevity. More specifically, cell therapies in the context of autoimmune disorders, neurodegenerative diseases, and cancer are reviewed from the perspectives of preclinical findings as well as available clinical data. Next, techniques to fabricate cell-biomaterials constructs, focusing on emerging 3D bioprinting technologies, will be reviewed. 3D bioprinting is an advancing field that enables fabricating complex, interconnected, and consistent cell-based constructs capable of scaling up highly reproducible cell-biomaterials platforms with high precision. It is expected that 3D bioprinting devices will expand and become more precise, scalable, and appropriate for clinical manufacturing. Rather than one printer fits all, seeing more application-specific printer types, such as a bioprinter for bone tissue fabrication, which would be different from a bioprinter for skin tissue fabrication, is anticipated in the future.
Collapse
Affiliation(s)
- Amirmasoud Samadi
- Department of Chemical and Biomolecular Engineering, 6000 Interdisciplinary Science & Engineering Building (ISEB), Irvine, California 92617, United States
| | - Ali Moammeri
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Parisa Abbasi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Avenue, Tehran 1458889694, Iran
| | - Zeinab Hosseinpour
- Biotechnology Research Laboratory, Faculty of Chemical Engineering, Babol Noshirvani University of Technology, Babol 4714871167, Mazandaran Province, Iran
| | - Arian Farokh
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Amin Shamsabadipour
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Maryam Heydari
- Department of Cell and Molecular Biology, Faculty of Biological Science, University of Kharazmi, Tehran 199389373, Iran
| | - M Rezaa Mohammadi
- Dale E. and Sarah Ann Fowler School of Engineering, Chapman University, Orange, California 92866, United States
| |
Collapse
|
38
|
Chen YS, Ng HY, Chen YW, Cho DY, Ho CC, Chen CY, Chiu SC, Jhong YR, Shie MY. Additive manufacturing of Schwann cell-laden collagen/alginate nerve guidance conduits by freeform reversible embedding regulate neurogenesis via exosomes secretion towards peripheral nerve regeneration. BIOMATERIALS ADVANCES 2023; 146:213276. [PMID: 36640522 DOI: 10.1016/j.bioadv.2022.213276] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/09/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023]
Abstract
Peripheral nerve injury is a common clinical problem that could be debilitating to one's quality of life. The complex nerve guidance conduits (NGCs) with cells in order to improve nerve regeneration. Therefore, we used freeform reversible embedding of suspended hydrogels to fabricate Schwann cells (SCs)-laden collagen/alginate (Col/Alg) NGCs. First, we evaluated Col influence on the characteristics of NGCs. After which, Wharton's jelly mesenchymal stem cells (WJMSC) are seeded onto the inner channel of NGCs and evaluated neural regeneration behaviors. Results indicated the SCs-laden NGCs with 2.5 % Col found the highest proliferation and secretion of neurotrophic protein. Furthermore, co-culture of SCs promoted differentiation of WJMSC as seen from the increased neurogenic-related protein in NGCs. To determine the molecular mechanism between SCs and WJMSC, we demonstrated the neurotrophic factors secreted by SCs act on tropomyosin receptor kinase A (TrkA) receptors of WJMSC to promote nerve regeneration. In addition, our study demonstrated SCs-derived exosomes had a critical role in regulating neural differentiation of WJMSC. Taken together, this study demonstrates the fabrication of SCs-laden Col/Alg NGCs for nerve regeneration and understanding regarding the synergistic regenerative mechanisms of different cells could bring us a step closer for clinical treatment of large nerve defects.
Collapse
Affiliation(s)
- Yueh-Sheng Chen
- School of Chinese Medicine, China Medical University, Taichung 40447, Taiwan; Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan
| | - Hooi Yee Ng
- Department of Education, China Medical University Hospital, Taichung 404332, Taiwan
| | - Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Der-Yang Cho
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Che Ho
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan; High Performance Materials Institute for x-Dimensional Printing, Asia University, Taichung City 41354, Taiwan
| | - Cheng-Yu Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Rong Jhong
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Ming-You Shie
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan; x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan; School of Dentistry, China Medical University, Taichung City 406040, Taiwan.
| |
Collapse
|
39
|
Ma H, Yu K, Wang H, Liu J, Cheng YY, Kang Y, Wang H, Zhang J, Song K. Fabrication and detection of a novel hybrid conductive scaffold based on alginate/gelatin/carboxylated carbon nanotubes (Alg/Gel/mMWCNTs) for neural tissue engineering. Tissue Cell 2023; 80:101995. [PMID: 36512950 DOI: 10.1016/j.tice.2022.101995] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Carbon nanotubes (CNTs), as kinds of conductive carbon nanomaterials, were widely applied in neural tissue engineering due to their excellent electrical conductivity and good biocompatibility. In this study, the carboxyl-modified multi-walled carbon nanotubes (mMWCNTs) were introduced into sodium alginate/gelatin (Alg/Gel) scaffolds to optimize the function of the hybrid scaffolds. The Alg/Gel/mMWCNTs conductive scaffolds with mMWCNTs content of 1%, 3%, and 5% were prepared by freeze-drying, respectively. Following this, the physicochemical properties and biocompatibility of the hybrid scaffolds at different magnetic field intensities were evaluated. The conductive scaffolds were characterized by Scanning electron microscopy (SEM) and Fourier transform infrared spectroscopy (FTIR). In general, the mMWCNTs addition improved the hydrophilic, electrical conductivity and mechanical properties of the composite scaffold, and PC12 cells showed a trend of gradual increase over culture time. Particularly, the Alg/Gel-1%C scaffold exhibited the best cell proliferation behavior. Briefly, the surface contact angle decreased from 74 ± 1° to 60 ± 3°, the electrical conductivity and compressive modulus increased to 1.32 × 10-3 ± 2.1 × 10-4 S/cm and 1.40 ± 0.076 Mpa, the G1 phase from 55.67 ± 1.86% to 59.77 ± 0.94% and the G2 phase from 10.32 ± 0.35% to 13.93 ± 1.26%,respectively. In the SEM images, PC12 cells were well-shaped and densely distributed. Therefore, the Alg/Gel/mMWCNTs conductive scaffold has potential as a tissue engineering scaffold in nerve regeneration.
Collapse
Affiliation(s)
- Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Kai Yu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hao Wang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| | - Hong Wang
- Department of Orthopeadics, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian 116033, China.
| | - Jingying Zhang
- The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 Guangdong, China.
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
40
|
Dadhich P, Kumar P, Roy A, Bitar KN. Advances in 3D Printing Technology for Tissue Engineering. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
41
|
In vitro evaluation of antibacterial activity and biocompatibility of synergistically cross-linked gelatin-alginate hydrogel beads as gentamicin carriers. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2022.104078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Liu Y, Shen S, Wu Y, Wang M, Cheng Y, Xia H, Jia R, Liu C, Wang Y, Xia Y, Cheng X, Yue Y, Xie Z. Percutaneous Electroosmosis of Berberine-Loaded Ca 2+ Crosslinked Gelatin/Alginate Mixed Hydrogel. Polymers (Basel) 2022; 14:polym14235101. [PMID: 36501495 PMCID: PMC9737946 DOI: 10.3390/polym14235101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Flexible conductive hydrogel has been driven by scientific breakthroughs and offers a wide variety of applications, including sensors, electronic skins, biomedicine, energy storage, etc. Based on the mixed-ion crosslinking method, gelatin and sodium alginate (Gel-Alg) composite hydrogels were successfully prepared using Ca2+ crosslinking. The migration behavior of berberine hydrochloride (BBH) in the matrix network structure of Gel-Alg hydrogel with a certain pore size under an electric field was studied, and the transdermal effect of berberine hydrochloride under an electric field was also studied. The experimental results show that Gel-Alg has good flexibility and conductivity, and electrical stimulation can enhance the transdermal effect of drugs. Gel-Alg composite hydrogel may be a new material with potential application value in future biomedical directions.
Collapse
Affiliation(s)
- Yinyin Liu
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Si Shen
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Yifang Wu
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Mengmeng Wang
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Yongfeng Cheng
- Clinical College of Anhui Medical University, Hefei 230031, China
- School of Life Science, University of Science and Technology of China, Hefei 230027, China
- Correspondence: (Y.C.); (H.X.); Tel./Fax: +86-13965033210 (H.X.)
| | - Hongmei Xia
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230012, China
- Correspondence: (Y.C.); (H.X.); Tel./Fax: +86-13965033210 (H.X.)
| | - Ruoyang Jia
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Chang Liu
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Yu Wang
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Ying Xia
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Xiaoman Cheng
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Yan Yue
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei 230012, China
| | - Zili Xie
- Anhui Institute for Food and Drug Control, Hefei 230051, China
| |
Collapse
|
43
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
44
|
Sonaye SY, Ertugral EG, Kothapalli CR, Sikder P. Extrusion 3D (Bio)Printing of Alginate-Gelatin-Based Composite Scaffolds for Skeletal Muscle Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15227945. [PMID: 36431432 PMCID: PMC9695625 DOI: 10.3390/ma15227945] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 05/13/2023]
Abstract
Volumetric muscle loss (VML), which involves the loss of a substantial portion of muscle tissue, is one of the most serious acute skeletal muscle injuries in the military and civilian communities. The injured area in VML may be so severely affected that the body loses its innate capacity to regenerate new functional muscles. State-of-the-art biofabrication methods such as bioprinting provide the ability to develop cell-laden scaffolds that could significantly expedite tissue regeneration. Bioprinted cell-laden scaffolds can mimic the extracellular matrix and provide a bioactive environment wherein cells can spread, proliferate, and differentiate, leading to new skeletal muscle tissue regeneration at the defect site. In this study, we engineered alginate−gelatin composite inks that could be used as bioinks. Then, we used the inks in an extrusion printing method to develop design-specific scaffolds for potential VML treatment. Alginate concentration was varied between 4−12% w/v, while the gelatin concentration was maintained at 6% w/v. Rheological analysis indicated that the alginate−gelatin inks containing 12% w/v alginate and 6% w/v gelatin were most suitable for developing high-resolution scaffolds with good structural fidelity. The printing pressure and speed appeared to influence the printing accuracy of the resulting scaffolds significantly. All the hydrogel inks exhibited shear thinning properties and acceptable viscosities, though 8−12% w/v alginate inks displayed properties ideal for printing and cell proliferation. Alginate content, crosslinking concentration, and duration played significant roles (p < 0.05) in influencing the scaffolds’ stiffness. Alginate scaffolds (12% w/v) crosslinked with 300, 400, or 500 mM calcium chloride (CaCl2) for 15 min yielded stiffness values in the range of 45−50 kPa, i.e., similar to skeletal muscle. The ionic strength of the crosslinking concentration and the alginate content significantly (p < 0.05) affected the swelling and degradation behavior of the scaffolds. Higher crosslinking concentration and alginate loading enhanced the swelling capacity and decreased the degradation kinetics of the printed scaffolds. Optimal CaCl2 crosslinking concentration (500 mM) and alginate content (12% w/v) led to high swelling (70%) and low degradation rates (28%) of the scaffolds. Overall, the results indicate that 12% w/v alginate and 6% w/v gelatin hydrogel inks are suitable as bioinks, and the printed scaffolds hold good potential for treating skeletal muscle defects such as VML.
Collapse
Affiliation(s)
| | - Elif G. Ertugral
- Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH 44115, USA
| | | | - Prabaha Sikder
- Mechanical Engineering, Cleveland State University, Cleveland, OH 44115, USA
- Correspondence:
| |
Collapse
|
45
|
Investigation of Collagen-Incorporated Sodium Alginate Bioprinting Hydrogel for Tissue Engineering. JOURNAL OF COMPOSITES SCIENCE 2022. [DOI: 10.3390/jcs6080227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tissue engineering is a promising area that is aimed at tissue regeneration and wound repair. Sodium alginate (SA) has been widely used as one of the most biocompatible materials for tissue engineering. The cost-efficiency and rapid gel ability made SA attractive in would healing and regeneration area. To improve printability and elasticity, many hydrogel-based bioinks were developed by mixing SA with other natural or synthetic polymers. In this paper, composite SA/COL bioink was used for the bioprinting of artificial cartilage tissue mimicries. The results showed that the concentration of both SA and COL has significant effects on filament diameter and merging. A higher concentration of the bioink solution led to better printing fidelity and less deformation. Overall, a higher SA concentration and a lower COL concentration contributed to a lower shrinkage ratio after crosslinking. In summary, the SA/COL composite bioink has favorable rheological properties and this study provided material composition optimization for future bioprinting of engineered tissues.
Collapse
|
46
|
Fabrication of hydrogels with adjustable mechanical properties through 3D cell-laden printing technology. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Abstract
Neuroprosthetic devices that record and modulate neural activities have demonstrated immense potential for bypassing or restoring lost neurological functions due to neural injuries and disorders. However, implantable electrical devices interfacing with brain tissue are susceptible to a series of inflammatory tissue responses along with mechanical or electrical failures which can affect the device performance over time. Several biomaterial strategies have been implemented to improve device-tissue integration for high quality and stable performance. Ranging from developing smaller, softer, and more flexible electrode designs to introducing bioactive coatings and drug-eluting layers on the electrode surface, such strategies have shown different degrees of success but with limitations. With their hydrophilic properties and specific bioactivities, carbohydrates offer a potential solution for addressing some of the limitations of the existing biomolecular approaches. In this review, we summarize the role of polysaccharides in the central nervous system, with a primary focus on glycoproteins and proteoglycans, to shed light on their untapped potential as biomaterials for neural implants. Utilization of glycosaminoglycans for neural interface and tissue regeneration applications is comprehensively reviewed to provide the current state of carbohydrate-based biomaterials for neural implants. Finally, we will discuss the challenges and opportunities of applying carbohydrate-based biomaterials for neural tissue interfaces.
Collapse
Affiliation(s)
- Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
48
|
Rao Z, Lin Z, Song P, Quan D, Bai Y. Biomaterial-Based Schwann Cell Transplantation and Schwann Cell-Derived Biomaterials for Nerve Regeneration. Front Cell Neurosci 2022; 16:926222. [PMID: 35836742 PMCID: PMC9273721 DOI: 10.3389/fncel.2022.926222] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Schwann cells (SCs) dominate the regenerative behaviors after peripheral nerve injury by supporting axonal regrowth and remyelination. Previous reports also demonstrated that the existence of SCs is beneficial for nerve regeneration after traumatic injuries in central nervous system. Therefore, the transplantation of SCs/SC-like cells serves as a feasible cell therapy to reconstruct the microenvironment and promote nerve functional recovery for both peripheral and central nerve injury repair. However, direct cell transplantation often leads to low efficacy, due to injection induced cell damage and rapid loss in the circulatory system. In recent years, biomaterials have received great attention as functional carriers for effective cell transplantation. To better mimic the extracellular matrix (ECM), many biodegradable materials have been engineered with compositional and/or topological cues to maintain the biological properties of the SCs/SCs-like cells. In addition, ECM components or factors secreted by SCs also actively contribute to nerve regeneration. Such cell-free transplantation approaches may provide great promise in clinical translation. In this review, we first present the current bio-scaffolds engineered for SC transplantation and their achievement in animal models and clinical applications. To this end, we focus on the physical and biological properties of different biomaterials and highlight how these properties affect the biological behaviors of the SCs/SC-like cells. Second, the SC-derived biomaterials are also reviewed and discussed. Finally, the relationship between SCs and functional biomaterials is summarized, and the trends of their future development are predicted toward clinical applications.
Collapse
Affiliation(s)
- Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zudong Lin
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Panpan Song
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Daping Quan
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Abdelbasset WK, Jasim SA, Sharma SK, Margiana R, Bokov DO, Obaid MA, Hussein BA, Lafta HA, Jasim SF, Mustafa YF. Alginate-Based Hydrogels and Tubes, as Biological Macromolecule-Based Platforms for Peripheral Nerve Tissue Engineering: A Review. Ann Biomed Eng 2022; 50:628-653. [PMID: 35446001 DOI: 10.1007/s10439-022-02955-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/20/2022] [Indexed: 12/25/2022]
Abstract
Unlike the central nervous system, the peripheral nervous system (PNS) has an inherent capacity to regenerate following injury. However, in the case of large nerve defects where end-to-end cooptation of two nerve stumps is not tension-free, autologous nerve grafting is often utilized to bridge the nerve gaps. To address the challenges associated with autologous nerve grafting, neural guidance channels (NGCs) have been successfully translated into clinic. Furthermore, hydrogel-based drug delivery systems have been extensively studied for the repair of PNS injuries. There are numerous biomaterial options for the production of NGCs and hydrogels. Among different candidates, alginate has shown promising results in PNS tissue engineering. Alginate is a naturally occurring polysaccharide which is biocompatible, non-toxic, non-immunogenic, and possesses modifiable properties. In the current review, applications, challenges, and future perspectives of alginate-based NGCs and hydrogels in the repair of PNS injuries will be discussed.
Collapse
Affiliation(s)
- Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, P.O. Box. 173, Al-Kharj, 11942, Saudi Arabia. .,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, 12613, Egypt.
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-anbar-Ramadi, Iraq
| | - Satish Kumar Sharma
- Pharmacology Department, Glocal School of Pharmacy, The Glocal University, Saharanpur, India
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia. .,Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia. .,Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, Russian Federation, 119991.,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr, Moscow, Russian Federation, 109240
| | - Maithm A Obaid
- College of Pharmacy, National University of Science and Technology, Thi Qar, Iraq
| | | | | | - Sara Firas Jasim
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| |
Collapse
|
50
|
Liu K, Yan L, Li R, Song Z, Ding J, Liu B, Chen X. 3D Printed Personalized Nerve Guide Conduits for Precision Repair of Peripheral Nerve Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103875. [PMID: 35182046 PMCID: PMC9036027 DOI: 10.1002/advs.202103875] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/25/2021] [Indexed: 05/07/2023]
Abstract
The treatment of peripheral nerve defects has always been one of the most challenging clinical practices in neurosurgery. Currently, nerve autograft is the preferred treatment modality for peripheral nerve defects, while the therapy is constantly plagued by the limited donor, loss of donor function, formation of neuroma, nerve distortion or dislocation, and nerve diameter mismatch. To address these clinical issues, the emerged nerve guide conduits (NGCs) are expected to offer effective platforms to repair peripheral nerve defects, especially those with large or complex topological structures. Up to now, numerous technologies are developed for preparing diverse NGCs, such as solvent casting, gas foaming, phase separation, freeze-drying, melt molding, electrospinning, and three-dimensional (3D) printing. 3D printing shows great potential and advantages because it can quickly and accurately manufacture the required NGCs from various natural and synthetic materials. This review introduces the application of personalized 3D printed NGCs for the precision repair of peripheral nerve defects and predicts their future directions.
Collapse
Affiliation(s)
- Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Lesan Yan
- Biomedical Materials and Engineering Research Center of Hubei ProvinceState Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology122 Luoshi RoadWuhan430070P. R. China
| | - Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Zhiming Song
- Department of Sports MedicineThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|