1
|
Pai V, Singh BN, Singh AK. Transformative advances in modeling brain aging and longevity: Success, challenges and future directions. Ageing Res Rev 2025; 108:102753. [PMID: 40222396 DOI: 10.1016/j.arr.2025.102753] [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: 01/02/2025] [Revised: 03/30/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Research on brain aging is crucial for understanding age-related neurodegenerative disorders and developing several therapeutic interventions. Numerous models ranging from two-dimensional (2D) cell-based, invertebrate, vertebrate, and sophisticated three-dimensional (3D) models have been used to understand the process of brain aging. Invertebrate models are ideal for researching conserved aging processes because of their simplicity, short lifespans, and genetic tractability. Moreover, vertebrate models, including zebrafish and rodents, exhibit more complex nervous systems and behaviors, enabling the exploration of age-related neurodegeneration and cognitive decline. 2D cell culture models derived from primary cells or immortalized cell lines are widely used for mechanistic studies at the cellular level but lack the physiological complexity of brain tissue. Recent advancements have shifted focus to 3D models, which better recapitulate the brain's microenvironment. Organoids derived from induced pluripotent stem cells mimic human brain architecture and enable the study of cell-cell interactions and aging in a human-specific context. Brain-on-a-chip systems integrate microfluidics and 3D cultures to model blood-brain barrier dynamics and neuronal networks. Additionally, scaffold-based 3D cultures and spheroids provide intermediate complexity, allowing researchers to study extracellular matrix interactions and age-related changes in neuronal function. These 3D models bridge the gap between traditional 2D cultures and animal-based in vivo studies, offering unprecedented insights into brain aging mechanisms. By combining these diverse models, researchers can unravel the multifaceted processes of brain aging and accelerate the development of targeted therapies for age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Varsha Pai
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bhisham Narayan Singh
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
2
|
Behrooznia Z, Nourmohammadi J, Mohammadi Z, Shabani F, Mashhadi R. "Biological evaluation of 3D-Printed chitosan-based scaffolds for tissue engineering". Carbohydr Res 2025; 551:109416. [PMID: 39977977 DOI: 10.1016/j.carres.2025.109416] [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: 09/30/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Chitosan is a natural polysaccharide with excellent biocompatibility, biodegradability, and antibacterial capabilities, making it a good candidate for tissue engineering. 3D printing has revolutionized biomaterial fabrication owing to its precision, customization, and ability to create complex structures. This article aims to provide an overview of the current advances in 3D printing techniques for fabricating scaffolds based on chitosan and its derivatives. It also describes various printing methods, including extrusion bioprinting technique, inkjet bioprinting, stereolithography, digital light processing, and indirect 3D printing for controlling porosity, mechanical strength, and biological characteristics of chitosan scaffolds for a variety of tissues, like bone, vascular, cardiac, cartilage, and skin. This review also examines the biological properties of 3D-printed chitosan scaffolds. The advancements in biological performance and 3D printing technology indicate a promising future for developing flexible, customized scaffolds consisting of chitosan.
Collapse
Affiliation(s)
- Zahra Behrooznia
- College of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Jhamak Nourmohammadi
- College of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran.
| | - Zahra Mohammadi
- College of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Fatemeh Shabani
- College of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Rahele Mashhadi
- College of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| |
Collapse
|
3
|
Tsai Y, Song J, Shi R, Knöll B, Synatschke CV. A Roadmap of Peptide-Based Materials in Neural Regeneration. Adv Healthc Mater 2025; 14:e2402939. [PMID: 39540310 PMCID: PMC11730414 DOI: 10.1002/adhm.202402939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Injuries to the nervous system lead to irreversible damage and limited functional recovery. The peripheral nervous system (PNS) can self-regenerate to some extent for short nerve gaps. In contrast, the central nervous system (CNS) has an intrinsic limitation to self-repair owing to its convoluted neural microenvironment and inhibitory response. The primary phase of CNS injury, happening within 48 h, results from external impacts like mechanical stress. Afterward, the secondary phase of the injury occurs, originating from neuronal excitotoxicity, mitochondrial dysfunction, and neuroinflammation. No golden standard to treat injured neurons exists, and conventional medicine serves only as a protective approach to alleviating the symptoms of chronic injury. Synthetic peptides provide a promising approach for neural repair, either as soluble drugs or by using their intrinsic self-assembly propensity to serve as an extracellular matrix (ECM) mimic for cell adhesion and to incorporate bioactive epitopes. In this review, an overview of nerve injury models, common in vitro models, and peptide-based therapeutics such as ECM mimics is provided. Due to the complexity of treating neuronal injuries, a multidisciplinary collaboration between biologists, physicians, and material scientists is paramount. Together, scientists with complementary expertise will be required to formulate future therapeutic approaches for clinical use.
Collapse
Affiliation(s)
- Yu‐Liang Tsai
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jialei Song
- Institute of NeurobiochemistryUniversity of UlmAlbert‐Einstein‐Allee 11D‐89081UlmGermany
- Department of OrthopedicsShanghai 9th People's HospitalShanghai Jiao Tong University School of MedicineZhizaoju Road 639Shanghai200011China
| | - Rachel Shi
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Yale School of Medicine333 Cedar StNew HavenCT06510USA
| | - Bernd Knöll
- Institute of NeurobiochemistryUniversity of UlmAlbert‐Einstein‐Allee 11D‐89081UlmGermany
| | - Christopher V. Synatschke
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
4
|
Lin Z, Li Q, Han X, Luo H, Wang Z, Qin Z, Huang Y, Feng Q, Cao X. An injectable and degradable heterogeneous microgel assembly capable of forming a "micro-nest group" for cell condensation and cartilage regeneration. MATERIALS HORIZONS 2024; 11:5438-5450. [PMID: 39189308 DOI: 10.1039/d4mh00724g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Cell condensation, linking the migration and chondrogenic differentiation of MSCs, plays a crucial role in cartilage development. Current cartilage repair strategies are inadequately concerned with this process, leading to a suboptimal quality of regenerated cartilage. Inspired by the "nest flocks" structure of Social Weavers, a degradable heterogeneous microgel assembly (F/S-MA) is developed, which can release SDF-1, to form a "micro-nest group" structure and bond with HAV peptides to promote cell recruitment, condensation and chondrogenic differentiation. First, slow-degrading microgels (S-microgels) grafted with HAV peptides and fast-degrading microgels (F-microgels) loaded with SDF-1 are fabricated by an amidation reaction and Schiff base reaction, respectively. They employ sulfhydryl-modified gelatin as assembling agents to form F/S-MA through a thiol-ene reaction, exhibiting injectability, tissue adhesion, and microporosity. F-microgels undergo rapid degradation, leading to the release of SDF-1 and the formation of a "micro-nest group" in F/S-MA. Consequently, F/S-MA exhibits cell recruitment ability, meanwhile facilitating BMSC condensation through the synergistic effects of the "micro-nest group" and HAV peptides. In vitro experiments prove that F/S-MA enhances the expression of cell-condensation-related markers, ultimately upregulating the secretion of cartilage matrix. Animal experiments show that F/S-MA optimizes the quality of regenerated cartilage by improving cell recruitment and condensation. F/S-MA enhances cell condensation through structural and component design, which will provide new insights for cartilage regeneration.
Collapse
Affiliation(s)
- Zequ Lin
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
| | - Qingtao Li
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xiyuan Han
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
| | - Huitong Luo
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
| | - Zetao Wang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
| | - Zhihao Qin
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
| | - Yue Huang
- School of Stomatology, Jinan University, Guangzhou 510641, China
| | - Qi Feng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
- School of Stomatology, Jinan University, Guangzhou 510641, China
| | - Xiaodong Cao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
5
|
Rahimi Darehbagh R, Seyedoshohadaei SA, Ramezani R, Rezaei N. Stem cell therapies for neurological disorders: current progress, challenges, and future perspectives. Eur J Med Res 2024; 29:386. [PMID: 39054501 PMCID: PMC11270957 DOI: 10.1186/s40001-024-01987-1] [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: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Stem cell-based therapies have emerged as a promising approach for treating various neurological disorders by harnessing the regenerative potential of stem cells to restore damaged neural tissue and circuitry. This comprehensive review provides an in-depth analysis of the current state of stem cell applications in primary neurological conditions, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), stroke, spinal cord injury (SCI), and other related disorders. The review begins with a detailed introduction to stem cell biology, discussing the types, sources, and mechanisms of action of stem cells in neurological therapies. It then critically examines the preclinical evidence from animal models and early human trials investigating the safety, feasibility, and efficacy of different stem cell types, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and induced pluripotent stem cells (iPSCs). While ESCs have been studied extensively in preclinical models, clinical trials have primarily focused on adult stem cells such as MSCs and NSCs, as well as iPSCs and their derivatives. We critically assess the current state of research for each cell type, highlighting their potential applications and limitations in different neurological conditions. The review synthesizes key findings from recent, high-quality studies for each neurological condition, discussing cell manufacturing, delivery methods, and therapeutic outcomes. While the potential of stem cells to replace lost neurons and directly reconstruct neural circuits is highlighted, the review emphasizes the critical role of paracrine and immunomodulatory mechanisms in mediating the therapeutic effects of stem cells in most neurological disorders. The article also explores the challenges and limitations associated with translating stem cell therapies into clinical practice, including issues related to cell sourcing, scalability, safety, and regulatory considerations. Furthermore, it discusses future directions and opportunities for advancing stem cell-based treatments, such as gene editing, biomaterials, personalized iPSC-derived therapies, and novel delivery strategies. The review concludes by emphasizing the transformative potential of stem cell therapies in revolutionizing the treatment of neurological disorders while acknowledging the need for rigorous clinical trials, standardized protocols, and multidisciplinary collaboration to realize their full therapeutic promise.
Collapse
Affiliation(s)
- Ramyar Rahimi Darehbagh
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Nanoclub Elites Association, Tehran, Iran
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Universal Scientific Education and Research Network (USERN), Sanandaj, Kurdistan, Iran
| | | | - Rojin Ramezani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Wan S, Aregueta Robles U, Poole-Warren L, Esrafilzadeh D. Advances in 3D tissue models for neural engineering: self-assembled versus engineered tissue models. Biomater Sci 2024; 12:3522-3549. [PMID: 38829222 DOI: 10.1039/d4bm00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Neural tissue engineering has emerged as a promising field that aims to create functional neural tissue for therapeutic applications, drug screening, and disease modelling. It is becoming evident in the literature that this goal requires development of three-dimensional (3D) constructs that can mimic the complex microenvironment of native neural tissue, including its biochemical, mechanical, physical, and electrical properties. These 3D models can be broadly classified as self-assembled models, which include spheroids, organoids, and assembloids, and engineered models, such as those based on decellularized or polymeric scaffolds. Self-assembled models offer advantages such as the ability to recapitulate neural development and disease processes in vitro, and the capacity to study the behaviour and interactions of different cell types in a more realistic environment. However, self-assembled constructs have limitations such as lack of standardised protocols, inability to control the cellular microenvironment, difficulty in controlling structural characteristics, reproducibility, scalability, and lengthy developmental timeframes. Integrating biomimetic materials and advanced manufacturing approaches to present cells with relevant biochemical, mechanical, physical, and electrical cues in a controlled tissue architecture requires alternate engineering approaches. Engineered scaffolds, and specifically 3D hydrogel-based constructs, have desirable properties, lower cost, higher reproducibility, long-term stability, and they can be rapidly tailored to mimic the native microenvironment and structure. This review explores 3D models in neural tissue engineering, with a particular focus on analysing the benefits and limitations of self-assembled organoids compared with hydrogel-based engineered 3D models. Moreover, this paper will focus on hydrogel based engineered models and probe their biomaterial components, tuneable properties, and fabrication techniques that allow them to mimic native neural tissue structures and environment. Finally, the current challenges and future research prospects of 3D neural models for both self-assembled and engineered models in neural tissue engineering will be discussed.
Collapse
Affiliation(s)
- Shuqian Wan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Ulises Aregueta Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
- Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
7
|
Mohammad Mehdipour N, Rajeev A, Kumar H, Kim K, Shor RJ, Natale G. Anisotropic hydrogel scaffold by flow-induced stereolithography 3D printing technique. BIOMATERIALS ADVANCES 2024; 161:213885. [PMID: 38743993 DOI: 10.1016/j.bioadv.2024.213885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
Essential organs, such as the heart and liver, contain a unique porous network that allows oxygen and nutrients to be exchanged, with distinct random to ordered regions displaying varying degrees of strength. A novel technique, referred to here as flow-induced lithography, was developed. This technique generates tunable anisotropic three-dimensional (3D) structures. The ink for this bioprinting technique was made of titanium dioxide nanorods (Ti) and kaolinite nanoclay (KLT) dispersed in a GelMA/PEGDA polymeric suspension. By controlling the flow rate, aligned particle microstructures were achieved in the suspensions. The application of UV light to trigger the polymerization of the photoactive prepolymer freezes the oriented particles in the polymer network. Because the viability test was successful in shearing suspensions containing cells, the flow-induced lithography technique can be used with both acellular scaffolds and cell-laden structures. Fabricated hydrogels show outstanding mechanical properties resembling human tissues, as well as significant cell viability (> 95 %) over one week. As a result of this technique and the introduction of bio-ink, a novel approach has been pioneered for developing anisotropic tissue implants utilizing low-viscosity biomaterials.
Collapse
Affiliation(s)
- Narges Mohammad Mehdipour
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Ashna Rajeev
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453552, India
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Roman J Shor
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Giovanniantonio Natale
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
8
|
Najafi H, Farahavar G, Jafari M, Abolmaali SS, Azarpira N, Tamaddon AM. Harnessing the Potential of Self-Assembled Peptide Hydrogels for Neural Regeneration and Tissue Engineering. Macromol Biosci 2024; 24:e2300534. [PMID: 38547473 DOI: 10.1002/mabi.202300534] [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: 11/21/2023] [Revised: 03/04/2024] [Indexed: 04/11/2024]
Abstract
Spinal cord injury, traumatic brain injury, and neurosurgery procedures usually lead to neural tissue damage. Self-assembled peptide (SAP) hydrogels, a type of innovative hierarchical nanofiber-forming peptide sequences serving as hydrogelators, have emerged as a promising solution for repairing tissue defects and promoting neural tissue regeneration. SAPs possess numerous features, such as adaptable morphologies, biocompatibility, injectability, tunable mechanical stability, and mimicking of the native extracellular matrix. This review explores the capacity of neural cell regeneration and examines the critical aspects of SAPs in neuroregeneration, including their biochemical composition, topology, mechanical behavior, conductivity, and degradability. Additionally, it delves into the latest strategies involving SAPs for central or peripheral neural tissue engineering. Finally, the prospects of SAP hydrogel design and development in the realm of neuroregeneration are discussed.
Collapse
Affiliation(s)
- Haniyeh Najafi
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, 71937-11351, Iran
| | - Ali Mohammad Tamaddon
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, 71468-64685, Iran
| |
Collapse
|
9
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
10
|
Saksena J, Hamilton AE, Gilbert RJ, Zuidema JM. Nanomaterial payload delivery to central nervous system glia for neural protection and repair. Front Cell Neurosci 2023; 17:1266019. [PMID: 37941607 PMCID: PMC10628439 DOI: 10.3389/fncel.2023.1266019] [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: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Central nervous system (CNS) glia, including astrocytes, microglia, and oligodendrocytes, play prominent roles in traumatic injury and degenerative disorders. Due to their importance, active pharmaceutical ingredients (APIs) are being developed to modulate CNS glia in order to improve outcomes in traumatic injury and disease. While many of these APIs show promise in vitro, the majority of APIs that are systemically delivered show little penetration through the blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) and into the CNS, rendering them ineffective. Novel nanomaterials are being developed to deliver APIs into the CNS to modulate glial responses and improve outcomes in injury and disease. Nanomaterials are attractive options as therapies for central nervous system protection and repair in degenerative disorders and traumatic injury due to their intrinsic capabilities in API delivery. Nanomaterials can improve API accumulation in the CNS by increasing permeation through the BBB of systemically delivered APIs, extending the timeline of API release, and interacting biophysically with CNS cell populations due to their mechanical properties and nanoscale architectures. In this review, we present the recent advances in the fields of both locally implanted nanomaterials and systemically administered nanoparticles developed for the delivery of APIs to the CNS that modulate glial activity as a strategy to improve outcomes in traumatic injury and disease. We identify current research gaps and discuss potential developments in the field that will continue to translate the use of glia-targeting nanomaterials to the clinic.
Collapse
Affiliation(s)
- Jayant Saksena
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Adelle E. Hamilton
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Albany Stratton Veterans Affairs Medical Center, Albany, NY, United States
| | - Jonathan M. Zuidema
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| |
Collapse
|
11
|
Pires LS, Melo DS, Borges JP, Henriques CR. PEDOT-Coated PLA Fibers Electrospun from Solutions Incorporating Fe(III)Tosylate in Different Solvents by Vapor-Phase Polymerization for Neural Regeneration. Polymers (Basel) 2023; 15:4004. [PMID: 37836053 PMCID: PMC10575336 DOI: 10.3390/polym15194004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 10/15/2023] Open
Abstract
Therapeutic solutions for injuries in the peripheral nervous system are limited and not existing in the case of the central nervous system. The electrical stimulation of cells through a cell-supporting conductive scaffold may contribute to new therapeutic solutions for nerve regeneration. In this work, biocompatible Polylactic acid (PLA) fibrous scaffolds incorporating Fe(III)Tosylate (FeTos) were produced by electrospinning a mixture of PLA/FeTos solutions towards a rotating cylinder, inducing fiber alignment. Fibers were coated with the conductive polymer Poly(3,4 ethylenedioxythiophene) (PEDOT) formed by vapor-phase polymerization of EDOT at 70 °C for 2 h. Different solvents (ETH, DMF and THF) were used as FeTos solvents to investigate the impact on the scaffold's conductivity. Scaffold conductivity was estimated to be as high as 1.50 × 10-1 S/cm when FeTos was dissolved in DMF. In vitro tests were performed to evaluate possible scaffold cytotoxicity, following ISO 10993-5, revealing no cytotoxic effects. Differentiation and growth of cells from the neural cell line SH-SY5Y seeded on the scaffolds were also assessed, with neuritic extensions observed in cells differentiated in neurons with retinoic acid. These extensions tended to follow the preferential alignment of the scaffold fibers.
Collapse
Affiliation(s)
- Laura S. Pires
- Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Diogo S. Melo
- Department of Physics, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - João P. Borges
- Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
- i3N/CENIMAT, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Célia R. Henriques
- Department of Physics, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
- i3N/CENIMAT, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
12
|
Xiao L, Xie P, Ma J, Shi K, Dai Y, Pang M, Luo J, Tan Z, Ma Y, Wang X, Rong L, He L. A Bioinspired Injectable, Adhesive, and Self-Healing Hydrogel with Dual Hybrid Network for Neural Regeneration after Spinal Cord Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304896. [PMID: 37462613 DOI: 10.1002/adma.202304896] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/19/2023] [Accepted: 07/02/2023] [Indexed: 09/07/2023]
Abstract
Hydrogel-based regenerated scaffolds show promise as a platform for neural regeneration following spinal cord injury (SCI). Nevertheless, the persistent problem of poor mechanical strength and limited integration with the host tissue still exists. In this study, a bioinspired hydrogel with highly sophisticated features for neural regeneration after SCI is developed. The hydrogel is composed of dihydroxyphenylalanine (DOPA)-grafted chitosan and a designer peptide, offering a unique set of qualities such as being injectable, having self-healing abilities, and adhering to tissues. Compared to conventional hydrogels, this hydrogel ensures a significant promotion of immune response modulation and axon regrowth while featuring synapse formation of various neurotransmitters and myelin regeneration. Subsequently, functional recoveries are enhanced, including motor function, sensory function, and particularly bladder defect repair. These positive findings demonstrate that the hydrogel has great potential as a strategy for repairing SCI. Moreover, the versatility of this strategy goes beyond neural regeneration and holds promise for tissue regeneration in other contexts. Overall, this proposed hydrogel represents an innovative and multifaceted tool for engineering structures in the biomedical field.
Collapse
Affiliation(s)
- Longyou Xiao
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Pengfei Xie
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Junwu Ma
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Kaixi Shi
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yu Dai
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Mao Pang
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jinghua Luo
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Zan Tan
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yahao Ma
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Xiaoying Wang
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Liumin He
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| |
Collapse
|
13
|
Muangsanit P, Chailangkarn T, Tanwattana N, Wongwanakul R, Lekcharoensuk P, Kaewborisuth C. Hydrogel-based 3D human iPSC-derived neuronal culture for the study of rabies virus infection. Front Cell Infect Microbiol 2023; 13:1215205. [PMID: 37692167 PMCID: PMC10485840 DOI: 10.3389/fcimb.2023.1215205] [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: 05/01/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Background Rabies is a highly fatal infectious disease that poses a significant threat to human health in developing countries. In vitro study-based understanding of pathogenesis and tropism of different strains of rabies virus (RABV) in the central nervous system (CNS) is limited due to the lack of suitable culture models that recapitulate the complex communication pathways among host cells, extracellular matrices, and viruses. Therefore, a three-dimensional (3D) cell culture that mimics cell-matrix interactions, resembling in vivo microenvironment, is necessary to discover relevant underlying mechanisms of RABV infection and host responses. Methods The 3D collagen-Matrigel hydrogel encapsulating hiPSC-derived neurons for RABV infection was developed and characterized based on cell viability, morphology, and gene expression analysis of neuronal markers. The replication kinetics of two different strains of RABV [wild-type Thai (TH) and Challenge Virus Standard (CVS)-11 strains] in both 2D and 3D neuronal cultures were examined. Differential gene expression analysis (DEG) of the neuropathological pathway of RABV-infected 2D and 3D models was also investigated via NanoString analysis. Results The 3D hiPSC-derived neurons revealed a more physiologically interconnected neuronal network as well as more robust and prolonged maturation and differentiation than the conventional 2D monolayer model. TH and CVS-11 exhibited distinct growth kinetics in 3D neuronal model. Additionally, gene expression analysis of the neuropathological pathway observed during RABV infection demonstrated a vast number of differentially expressed genes (DEGs) in 3D model. Unlike 2D neuronal model, 3D model displayed more pronounced cellular responses upon infection with CVS-11 when compared to the TH-infected group, highlighting the influence of the cell environment on RABV-host interactions. Gene ontology (GO) enrichment of DEGs in the infected 3D neuronal culture showed alterations of genes associated with the inflammatory response, apoptotic signaling pathway, glutamatergic synapse, and trans-synaptic signaling which did not significantly change in 2D culture. Conclusion We demonstrated the use of a hydrogel-based 3D hiPSC-derived neuronal model, a highly promising technology, to study RABV infection in a more physiological environment, which will broaden our understanding of RABV-host interactions in the CNS.
Collapse
Affiliation(s)
- Papon Muangsanit
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Thanathom Chailangkarn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Nathiphat Tanwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
| | - Ratjika Wongwanakul
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Porntippa Lekcharoensuk
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
- Center for Advance Studies in Agriculture and Food, KU Institute Studies, Kasetsart University, Bangkok, Thailand
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
14
|
Liu W, Xu B, Zhao S, Han S, Quan R, Liu W, Ji C, Chen B, Xiao Z, Yin M, Yin Y, Dai J, Zhao Y. Spinal cord tissue engineering via covalent interaction between biomaterials and cells. SCIENCE ADVANCES 2023; 9:eade8829. [PMID: 36753555 PMCID: PMC9908024 DOI: 10.1126/sciadv.ade8829] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/10/2023] [Indexed: 05/29/2023]
Abstract
Noncovalent interactions between cells and environmental cues have been recognized as fundamental physiological interactions that regulate cell behavior. However, the effects of the covalent interactions between cells and biomaterials on cell behavior have not been examined. Here, we demonstrate a combined strategy based on covalent conjugation between biomaterials (collagen fibers/lipid nanoparticles) and various cells (exogenous neural progenitor cells/astrocytes/endogenous tissue-resident cells) to promote neural regeneration after spinal cord injury (SCI). We found that metabolic azido-labeled human neural progenitor cells conjugated on dibenzocyclooctyne-modified collagen fibers significantly promoted cell adhesion, spreading, and differentiation compared with noncovalent adhesion. In addition, dibenzocyclooctyne-modified lipid nanoparticles containing edaravone, a well-known ROS scavenger, could target azide-labeled spinal cord tissues or transplanted azide-modified astrocytes to improve the SCI microenvironment. The combined application of these covalent conjugation strategies in a rat SCI model boosted neural regeneration, suggesting that the covalent interactions between cells and biomaterials have great potential for tissue regeneration.
Collapse
Affiliation(s)
- Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Bai Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Wenbin Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Chunnan Ji
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| |
Collapse
|
15
|
Khajehmohammadi M, Azizi Tafti R, Nikukar H. Effect of porosity on mechanical and biological properties of bioprinted scaffolds. J Biomed Mater Res A 2023; 111:245-260. [PMID: 36205372 DOI: 10.1002/jbm.a.37455] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 01/10/2023]
Abstract
Treatment of tissue defects commonly represents a major problem in clinics due to difficulties involving a shortage of donors, inappropriate sizes, abnormal shapes, and immunological rejection. While many scaffold parameters such as pore shape, porosity percentage, and pore connectivity could be adjusted to achieve desired mechanical and biological properties. These parameters are crucial scaffold parameters that can be accurately produced by 3D bioprinting technology based on the damaged tissue. In the present research, the effect of porosity percentage (40%, 50%, and 60%) and different pore shapes (square, star, and gyroid) on the mechanical (e.g., stiffness, compressive and tensile behavior) and biological (e.g., biodegradation, and cell viability) properties of porous polycaprolactone (PCL) scaffolds coated with gelatin have been investigated. Moreover, human foreskin fibroblast cells were cultured on the scaffolds in the in-vitro procedures. MTT assay (4, 7, and 14 days) was utilized to determine the cytotoxicity of the porous scaffolds. It is revealed that the porous scaffolds produced by the bioprinter did not produce a cytotoxic effect. Among all the porous scaffolds, scaffolds with a pore size of about 500 μm and porosity of 50% showed the best cell proliferation compared to the controls after 14 days. The results demonstrated that the pore shape, porosity percentage, and pore connectivity have an important role in improving the mechanical and biological properties of porous scaffolds. These 3D bioprinted biodegradable scaffolds exhibit potential for future application as polymeric scaffolds in hard tissue engineering applications.
Collapse
Affiliation(s)
| | | | - Habib Nikukar
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
16
|
PCL-based 3D nanofibrous structure with well-designed morphology and enhanced specific surface area for tissue engineering application. Prog Biomater 2023; 12:113-122. [PMID: 36646866 PMCID: PMC10154450 DOI: 10.1007/s40204-022-00215-5] [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: 05/25/2022] [Accepted: 12/13/2022] [Indexed: 01/18/2023] Open
Abstract
Tissue engineering opens a new horizon for biological tissue replacement applications. Scaffolds, appropriate cells, and signaling induction are the main three determinant parameters in any tissue engineering applications. Designing a suitable scaffold which can mimic the cellular inherent and natural habitation is of great importance for cellular growth and proliferation. Just like a natural extracellular matrix (ECM), scaffolds provide the cells with an environment for performing biological functions. Accordingly, vast surface area and three-dimensional nanofibrous structures are among the pivotal characteristics of functional scaffolds in tissue engineering, and enhancement of their properties is the main purpose of the present research. In our previous study, a patterned structure composed of continuous nanofibers and microparticles was introduced. In this work, a new modification is applied for adjustment of the surface area of an electrospun/electrosprayed scaffold. For this purpose, at predetermined stages during electrospinning/electrospraying, the nitrogen gas is flushed through the mesh holes of the collector in the opposite direction of the jet movement. This method has led to the formation of very thin nanofibrous layers at nitrogen flush intervals by providing a cooling effect of the sweeping nitrogen. As a consequence, a straticulated structure has been fabricated which possesses extremely high surface/volume ratio. The porosity, water absorption, and morphological analysis were conducted on the obtained scaffold. In vitro cytocompatibility assessments as well as histological analysis demonstrated that the fabricated scaffold provides a proper substrate for cellular attachment, proliferation and infiltration. These findings can be advantageous in three-dimensional tissue engineering such as bone tissue engineering applications. Furthermore, according to the advanced microstructure and vast surface area of the fabricated samples, they can be applied in many other applications, such as membrane, filtration, etc.
Collapse
|
17
|
Barrejón M, Zummo F, Mikhalchan A, Vilatela JJ, Fontanini M, Scaini D, Ballerini L, Prato M. TEGylated Double-Walled Carbon Nanotubes as Platforms to Engineer Neuronal Networks. ACS APPLIED MATERIALS & INTERFACES 2023; 15:77-90. [PMID: 36270018 PMCID: PMC9837783 DOI: 10.1021/acsami.2c16808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/12/2022] [Indexed: 05/27/2023]
Abstract
In the past two decades, important results have been obtained on the application of carbon nanotubes (CNTs) as components of smart interfaces promoting neuronal growth and differentiation. Different forms of CNTs have been employed as scaffolds, including raw CNTs and functionalized CNTs, characterized by a different number of walls, mainly single-walled CNTs (SWCNTs) or multiwalled CNTs (MWCNTs). However, double-walled carbon nanotubes (DWCNTs), which present interesting electronic and transport properties, have barely been studied in the field. Apart from the electrical conductivity, the morphology, shape, porosity, and corresponding mechanical properties of the scaffold material are important parameters when dealing with neuronal cells. Thus, the presence of open porous and interconnected networks is essential for cell growth and differentiation. Here, we present an easy methodology to prepare porous self-standing and electrically conductive DWCNT-based scaffolds and study the growth of neuro/glial networks and their synaptic activity. A cross-linking approach with triethylene glycol (TEG) derivatives is applied to improve the tensile performance of the scaffolds while neuronal growth and differentiation are promoted. By testing different DWCNT-based constructs, we confirm that the manufactured structures guarantee a biocompatible scaffold, while favoring the design of artificial networks with high complexity.
Collapse
Affiliation(s)
- Myriam Barrejón
- Department
of Chemical and Pharmaceutical Sciences, INSTM, UdR Trieste, University of Trieste, Via L. Giorgieri 1, Trieste34127, Italy
- Neural
Repair and Biomaterials Laboratory, Hospital
Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, Toledo45071, Spain
| | - Francesca Zummo
- International
School for Advanced Studies (SISSA/ISAS), Trieste34136, Italy
| | | | | | - Mario Fontanini
- International
School for Advanced Studies (SISSA/ISAS), Trieste34136, Italy
| | - Denis Scaini
- International
School for Advanced Studies (SISSA/ISAS), Trieste34136, Italy
- Basque
Foundation for Science, Ikerbasque, Bilbao48013, Spain
- University
of Basque Country, Faculty of Pharmacy, Paseo de la Universidad 7, Vitoria-Gasteiz01006, Spain
| | - Laura Ballerini
- International
School for Advanced Studies (SISSA/ISAS), Trieste34136, Italy
| | - Maurizio Prato
- Department
of Chemical and Pharmaceutical Sciences, INSTM, UdR Trieste, University of Trieste, Via L. Giorgieri 1, Trieste34127, Italy
- Basque
Foundation for Science, Ikerbasque, Bilbao48013, Spain
- Center for
Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, Donostia
San Sebastián20014, Spain
| |
Collapse
|
18
|
Liu H, Chansoria P, Delrot P, Angelidakis E, Rizzo R, Rütsche D, Applegate LA, Loterie D, Zenobi-Wong M. Filamented Light (FLight) Biofabrication of Highly Aligned Tissue-Engineered Constructs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204301. [PMID: 36095325 DOI: 10.1002/adma.202204301] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/07/2022] [Indexed: 06/15/2023]
Abstract
Cell-laden hydrogels used in tissue engineering generally lack sufficient 3D topographical guidance for cells to mature into aligned tissues. A new strategy called filamented light (FLight) biofabrication rapidly creates hydrogels composed of unidirectional microfilament networks, with diameters on the length scale of single cells. Due to optical modulation instability, a light beam is divided optically into FLight beams. Local polymerization of a photoactive resin is triggered, leading to local increase in refractive index, which itself creates self-focusing waveguides and further polymerization of photoresin into long hydrogel microfilaments. Diameter and spacing of the microfilaments can be tuned from 2 to 30 µm by changing the coherence length of the light beam. Microfilaments show outstanding cell instructive properties with fibroblasts, tenocytes, endothelial cells, and myoblasts, influencing cell alignment, nuclear deformation, and extracellular matrix deposition. FLight is compatible with multiple types of photoresins and allows for biofabrication of centimeter-scale hydrogel constructs with excellent cell viability within seconds (<10 s per construct). Multidirectional microfilaments are achievable within a single hydrogel construct by changing the direction of FLight projection, and complex multimaterial/multicellular tissue-engineered constructs are possible by sequentially exchanging the cell-laden photoresin. FLight offers a transformational approach to developing anisotropic tissues using photo-crosslinkable biomaterials.
Collapse
Affiliation(s)
- Hao Liu
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Parth Chansoria
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Paul Delrot
- Readily3D SA, EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Emmanouil Angelidakis
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Riccardo Rizzo
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Dominic Rütsche
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive & Hand Surgery, Lausanne University Hospital, University of Lausanne, Epalinges, 1066, Switzerland
| | - Damien Loterie
- Readily3D SA, EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
19
|
Manganas P, Kavatzikidou P, Kordas A, Babaliari E, Stratakis E, Ranella A. The role of mechanobiology on the Schwann cell response: A tissue engineering perspective. Front Cell Neurosci 2022; 16:948454. [PMID: 36035260 PMCID: PMC9399718 DOI: 10.3389/fncel.2022.948454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schwann cells (SCs), the glial cells of the peripheral nervous system (PNS), do not only form myelin sheaths thereby insulating the electrical signal propagated by the axons, but also play an essential role in the regeneration of injured axons. SCs are inextricably connected with their extracellular environment and the mechanical stimuli that are received determine their response during development, myelination and injuries. To this end, the mechanobiological response of SCs is being actively researched, as it can determine the suitability of fabricated scaffolds for tissue engineering and regenerative medicine applications. There is growing evidence that SCs are sensitive to changes in the mechanical properties of the surrounding environment (such as the type of material, its elasticity and stiffness), different topographical features provided by the environment, as well as shear stress. In this review, we explore how different mechanical stimuli affect SC behaviour and highlight the importance of exploring many different avenues when designing scaffolds for the repair of PNS injuries.
Collapse
Affiliation(s)
- Phanee Manganas
- Tissue Engineering, Regenerative Medicine and Immunoengineering Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
| | - Paraskevi Kavatzikidou
- Tissue Engineering, Regenerative Medicine and Immunoengineering Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
- Ultrafast Laser Micro and Nano Processing Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
| | - Antonis Kordas
- Tissue Engineering, Regenerative Medicine and Immunoengineering Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece
| | - Eleftheria Babaliari
- Tissue Engineering, Regenerative Medicine and Immunoengineering Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
- Ultrafast Laser Micro and Nano Processing Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
| | - Emmanuel Stratakis
- Ultrafast Laser Micro and Nano Processing Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
| | - Anthi Ranella
- Tissue Engineering, Regenerative Medicine and Immunoengineering Laboratory, Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (IESL-FORTH), Heraklion, Greece
- *Correspondence: Anthi Ranella
| |
Collapse
|
20
|
Carvalho IC, Mansur HS. Quo vadis? Bioengineered polysaccharide-based hydrogel scaffolds for damaged central nervous system recovery and regeneration. Neural Regen Res 2022; 17:1478-1480. [PMID: 34916424 PMCID: PMC8771114 DOI: 10.4103/1673-5374.330599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/04/2021] [Accepted: 08/25/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Isadora C. Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Herman S. Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
21
|
Osouli-Bostanabad K, Masalehdan T, Kapsa RMI, Quigley A, Lalatsa A, Bruggeman KF, Franks SJ, Williams RJ, Nisbet DR. Traction of 3D and 4D Printing in the Healthcare Industry: From Drug Delivery and Analysis to Regenerative Medicine. ACS Biomater Sci Eng 2022; 8:2764-2797. [PMID: 35696306 DOI: 10.1021/acsbiomaterials.2c00094] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Three-dimensional (3D) printing and 3D bioprinting are promising technologies for a broad range of healthcare applications from frontier regenerative medicine and tissue engineering therapies to pharmaceutical advancements yet must overcome the challenges of biocompatibility and resolution. Through comparison of traditional biofabrication methods with 3D (bio)printing, this review highlights the promise of 3D printing for the production of on-demand, personalized, and complex products that enhance the accessibility, effectiveness, and safety of drug therapies and delivery systems. In addition, this review describes the capacity of 3D bioprinting to fabricate patient-specific tissues and living cell systems (e.g., vascular networks, organs, muscles, and skeletal systems) as well as its applications in the delivery of cells and genes, microfluidics, and organ-on-chip constructs. This review summarizes how tailoring selected parameters (i.e., accurately selecting the appropriate printing method, materials, and printing parameters based on the desired application and behavior) can better facilitate the development of optimized 3D-printed products and how dynamic 4D-printed strategies (printing materials designed to change with time or stimulus) may be deployed to overcome many of the inherent limitations of conventional 3D-printed technologies. Comprehensive insights into a critical perspective of the future of 4D bioprinting, crucial requirements for 4D printing including the programmability of a material, multimaterial printing methods, and precise designs for meticulous transformations or even clinical applications are also given.
Collapse
Affiliation(s)
- Karim Osouli-Bostanabad
- Biomaterials, Bio-engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular, Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Tahereh Masalehdan
- Department of Materials Engineering, Institute of Mechanical Engineering, University of Tabriz, Tabriz 51666-16444, Iran
| | - Robert M I Kapsa
- Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Anita Quigley
- Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Aikaterini Lalatsa
- Biomaterials, Bio-engineering and Nanomedicine (BioN) Lab, Institute of Biomedical and Biomolecular, Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Kiara F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.,Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Stephanie J Franks
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Richard J Williams
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, Research School of Chemistry and the John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.,The Graeme Clark Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
22
|
Decellularization of Full Heart—Optimizing the Classical Sodium-Dodecyl-Sulfate-Based Decellularization Protocol. Bioengineering (Basel) 2022; 9:bioengineering9040147. [PMID: 35447709 PMCID: PMC9032179 DOI: 10.3390/bioengineering9040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 12/05/2022] Open
Abstract
Compared to cell therapy, where cells are injected into a defect region, the treatment of heart infarction with cells seeded in a vascularized scaffold bears advantages, such as an immediate nutrient supply or a controllable and persistent localization of cells. For this purpose, decellularized native tissues are a preferable choice as they provide an in vivo-like microenvironment. However, the quality of such scaffolds strongly depends on the decellularization process. Therefore, two protocols based on sodium dodecyl sulfate or sodium deoxycholate were tailored and optimized for the decellularization of a porcine heart. The obtained scaffolds were tested for their applicability to generate vascularized cardiac patches. Decellularization with sodium dodecyl sulfate was found to be more suitable and resulted in scaffolds with a low amount of DNA, a highly preserved extracellular matrix composition, and structure shown by GAG quantification and immunohistochemistry. After seeding human endothelial cells into the vasculature, a coagulation assay demonstrated the functionality of the endothelial cells to minimize the clotting of blood. Human-induced pluripotent-stem-cell-derived cardiomyocytes in co-culture with fibroblasts and mesenchymal stem cells transferred the scaffold into a vascularized cardiac patch spontaneously contracting with a frequency of 25.61 ± 5.99 beats/min for over 16 weeks. The customized decellularization protocol based on sodium dodecyl sulfate renders a step towards a preclinical evaluation of the scaffolds.
Collapse
|
23
|
Wertheim L, Edri R, Goldshmit Y, Kagan T, Noor N, Ruban A, Shapira A, Gat‐Viks I, Assaf Y, Dvir T. Regenerating the Injured Spinal Cord at the Chronic Phase by Engineered iPSCs-Derived 3D Neuronal Networks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105694. [PMID: 35128819 PMCID: PMC9008789 DOI: 10.1002/advs.202105694] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 05/08/2023]
Abstract
Cell therapy using induced pluripotent stem cell-derived neurons is considered a promising approach to regenerate the injured spinal cord (SC). However, the scar formed at the chronic phase is not a permissive microenvironment for cell or biomaterial engraftment or for tissue assembly. Engineering of a functional human neuronal network is now reported by mimicking the embryonic development of the SC in a 3D dynamic biomaterial-based microenvironment. Throughout the in vitro cultivation stage, the system's components have a synergistic effect, providing appropriate cues for SC neurogenesis. While the initial biomaterial supported efficient cell differentiation in 3D, the cells remodeled it to provide an inductive microenvironment for the assembly of functional SC implants. The engineered tissues are characterized for morphology and function, and their therapeutic potential is investigated, revealing improved structural and functional outcomes after acute and chronic SC injuries. Such technology is envisioned to be translated to the clinic to rewire human injured SC.
Collapse
Affiliation(s)
- Lior Wertheim
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
- The Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv6997801Israel
- The Department of Materials Science and EngineeringFaculty of EngineeringTel Aviv UniversityTel Aviv6997801Israel
| | - Reuven Edri
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - Yona Goldshmit
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
- Steyer School of Health ProfessionsSackler Faculty of MedicineTel‐Aviv UniversityTel Aviv6997801Israel
| | - Tomer Kagan
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - Nadav Noor
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - Angela Ruban
- Steyer School of Health ProfessionsSackler Faculty of MedicineTel‐Aviv UniversityTel Aviv6997801Israel
| | - Assaf Shapira
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - Irit Gat‐Viks
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - Yaniv Assaf
- School of Neurobiology, Biochemistry and BiophysicsFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv6997801Israel
| | - Tal Dvir
- Shmunis School of Biomedicine and Cancer ResearchFaculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
- The Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv6997801Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv6997801Israel
- The Department of Biomedical EngineeringFaculty of EngineeringTel Aviv UniversityTel Aviv6997801Israel
- Sagol Center for Regenerative BiotechnologyTel Aviv UniversityTel Aviv6997801Israel
| |
Collapse
|
24
|
Tupone MG, Panella G, d’Angelo M, Castelli V, Caioni G, Catanesi M, Benedetti E, Cimini A. An Update on Graphene-Based Nanomaterials for Neural Growth and Central Nervous System Regeneration. Int J Mol Sci 2021; 22:13047. [PMID: 34884851 PMCID: PMC8657785 DOI: 10.3390/ijms222313047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Thanks to their reduced size, great surface area, and capacity to interact with cells and tissues, nanomaterials present some attractive biological and chemical characteristics with potential uses in the field of biomedical applications. In this context, graphene and its chemical derivatives have been extensively used in many biomedical research areas from drug delivery to bioelectronics and tissue engineering. Graphene-based nanomaterials show excellent optical, mechanical, and biological properties. They can be used as a substrate in the field of tissue engineering due to their conductivity, allowing to study, and educate neural connections, and guide neural growth and differentiation; thus, graphene-based nanomaterials represent an emerging aspect in regenerative medicine. Moreover, there is now an urgent need to develop multifunctional and functionalized nanomaterials able to arrive at neuronal cells through the blood-brain barrier, to manage a specific drug delivery system. In this review, we will focus on the recent applications of graphene-based nanomaterials in vitro and in vivo, also combining graphene with other smart materials to achieve the best benefits in the fields of nervous tissue engineering and neural regenerative medicine. We will then highlight the potential use of these graphene-based materials to construct graphene 3D scaffolds able to stimulate neural growth and regeneration in vivo for clinical applications.
Collapse
Affiliation(s)
- Maria Grazia Tupone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
- Center for Microscopy, University of L’Aquila, 67100 L’Aquila, Italy
| | - Gloria Panella
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.G.T.); (G.P.); (M.d.); (V.C.); (G.C.); (M.C.); (A.C.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
25
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
26
|
Chen T, Jiang H, Zhu Y, Chen X, Zhang D, Li X, Shen F, Xia H, Min Y, Xie K. Highly Ordered 3D Tissue Engineering Scaffolds as a Versatile Culture Platform for Nerve Cells Growth. Macromol Biosci 2021; 21:e2100047. [PMID: 33893711 DOI: 10.1002/mabi.202100047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/11/2021] [Indexed: 12/14/2022]
Abstract
Tissue engineering scaffolds provide an encouraging alternative for nerve injuries due to their biological support for nerve cell growth, which can be used for neuronal repair. Nerve cells have been reported to be mostly cultured on 2D scaffolds that cannot mimic the native extracellular matrix. Herein, highly ordered 3D scaffolds are fabricated for nerve cell culture by melt electrospinning writing, the microstructures and geometries of the scaffolds could be well modulated. An effective strategy for scaffold surface modification to promote nerve cell growth is proposed. The effects of scaffolds with different surface modifications, viz., plasma treatment, single poly-D-lysine (PDL) coating after plasma treatment, single laminin (LM) coating after plasma treatment, double PDL and LM coatings after plasma treatment, on PC12 cell growth are evaluated. Experiments show the scaffold modified with double PDL and LM coatings after plasma treatment facilitated the growth of PC12 cells most effectively, indicating the synergistic effect of PDL and LM on the growth of nerve cells. This is the first systematic and quantitative study of the effects of different scaffold surface modifications on nerve cell growth. The above results provide a versatile culture platform for growing nerve cells, and for recovery from peripheral nerve injury.
Collapse
Affiliation(s)
- Tingkuo Chen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Haiming Jiang
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yibin Zhu
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xueliu Chen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Dao Zhang
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xiang Li
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Fangcheng Shen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Hongyan Xia
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yonggang Min
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, 510006, China
| | - Kang Xie
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| |
Collapse
|