1
|
Kary AD, Noelle H, Magin CM. Tissue-Informed Biomaterial Innovations Advance Pulmonary Regenerative Engineering. ACS Macro Lett 2025; 14:434-447. [PMID: 40102038 DOI: 10.1021/acsmacrolett.5c00075] [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: 03/20/2025]
Abstract
Irreversible progressive pulmonary diseases drastically reduce the patient quality of life, while transplantation remains the only definitive cure. Research into lung regeneration pathways holds significant potential to expand and promote the discovery of new treatment options. Polymeric biomaterials designed to replicate key tissue characteristics (i.e., biochemical composition and mechanical cues) show promise for creating environments in which to study chronic lung diseases and initiate lung tissue regeneration. In this Viewpoint, we explore how naturally derived materials can be employed alone or combined with engineered polymer systems to create advanced tissue culture platforms. Pulmonary tissue models have historically leveraged natural materials, including basement membrane extracts and a decellularized extracellular matrix, as platforms for lung regeneration studies. Here, we provide an overview of the progression of pulmonary regenerative engineering, exploring how innovations in the growing field of tissue-informed biomaterials have the potential to advance lung regeneration research by bridging the gap between biological relevance and mechanical precision.
Collapse
Affiliation(s)
- Anton D Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Haley Noelle
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
2
|
Acharya JR, Kumar S, Girdhar GA, Patel S, Parekh NH, Patadiya HH, Zinjala AN, Haque M. 3D Bioprinting: Shaping the Future of Periodontal Tissue Regeneration and Disease Management. Cureus 2025; 17:e82432. [PMID: 40255528 PMCID: PMC12007905 DOI: 10.7759/cureus.82432] [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: 03/21/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
The periodontium is one of the most complex tissues in the body, consisting of a hierarchical blend of soft and hard tissues. Its complex architecture makes treating and regenerating disease-damaged periodontal tissues a persistent challenge in biomedicine. Three-dimensional (3D) bioprinting represents a transformative approach to tissue engineering, offering promising advancements in treating and regenerating periodontal disease. This innovative technology enables the precise fabrication of complex, patient-specific tissue structures, facilitating the repair and restoration of damaged periodontal tissues, including the gingiva, bone, and periodontal ligament (PDL). By utilizing biocompatible materials such as living cells, hydrogels, and growth factors, 3D bioprinting has the potential to create functional, biologically integrated constructs that can mimic the natural architecture of periodontal tissues. However, translating these advancements into clinical applications remains a challenge. Emerging technologies like bioprinting have been developed to address some limitations of traditional tissue engineering methods. This review explores the current state of 3D bioprinting technology, its application in periodontal disease treatment, and the challenges associated with scaling up this technology for clinical use. Additionally, it discusses the future implications of bioprinting for personalized medicine, offering a new frontier for regenerating periodontal tissues and improving patient outcomes in oral health. Integrating 3D bioprinting into periodontal regenerative therapies could revolutionize clinical practices, offering more effective, tailored, and sustainable solutions to address the challenges of periodontal disease.
Collapse
Affiliation(s)
- Jahnavi R Acharya
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Gaurav A Girdhar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Shirishkumar Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Nirav H Parekh
- Department of Periodontology, Smile Rite Dental Care, Southington, USA
| | | | - Anjali Narsinhbhai Zinjala
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Mainul Haque
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
- Department of Research, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
3
|
Watcharajittanont N, Jatuworapruk K, Prarokijjak W, Sangsuwan P, Meesane J. Mimicking bone remodeling scaffolds of polyvinylalcohol/silk fibroin with phytoactive compound of soy protein isolate as surgical supporting biomaterials for tissue formation at defect area in osteoporosis; characterization, morphology, and in-vitrotesting. Biomed Mater 2025; 20:025046. [PMID: 39951896 DOI: 10.1088/1748-605x/adb66f] [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: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 02/17/2025]
Abstract
Mimicking bone remodeling scaffolds were developed as supportive biomaterials to promote tissue formation at defect sites in osteoporosis. Scaffolds made of polyvinyl alcohol (PVA) were mixed with varying weight ratios of silk fibroin (SF) and a phytoactive compound-based soy protein isolate (SPI); PVA30SF, PVA20SF10SPI, PVA15SF15SPI, PVA10SF20SPI, PVA30SPI. PVA was used as control. These components were mixed into aqueous solution and crosslinking with EDC before freeze thawing and freeze drying, respectively. Then, the scaffolds were characterized at the molecular level using Fourier transform infrared spectroscopy and their morphology was observed using scanning electron microscopy. Physical properties including swelling and degradation were tested, as well as mechanical properties like stress-strain behavior and modulus. The biological performance of the scaffolds was evaluated through osteoblast cell culturing, assessing cell viability, proliferation, alkaline phosphatase (ALP) activity, calcium content, and calcium deposition. The results demonstrate that the scaffolds with both SF and SPI had greater molecular mobility of -OH, amide I, II, and III groups, compared to the scaffold with only SF or SPI. These scaffolds also displayed larger pore sizes. Scaffolds with both SF and SPI showed higher swelling and degradation rates than those with only SF or SPI. Additionally, they exhibited better cell viability and calcium deposition, along with increased cell proliferation, ALP activity, and calcium content. Notably, the scaffold with a higher amount of SPI, PVA10SF20SPI, exhibited the most suitable performance for enhancing cell response, thereby promoting bone formation. This scaffold is proposed as a supportive biomaterial to be incorporated with plates and screws for bone fixation at defect sites in osteoporosis.
Collapse
Affiliation(s)
| | - Kanon Jatuworapruk
- Department of Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Worasak Prarokijjak
- Faculty of Learning Sciences and Education, Thammasat University, Pathumthani, Thailand
| | - Prawichaya Sangsuwan
- Molecular Biology and Bioinformatics Program, Biological Science Division, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Jirut Meesane
- Institute of Biomedical Engineering, Department of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
4
|
Mainik P, Aponte‐Santamaría C, Fladung M, Curticean RE, Wacker I, Hofhaus G, Bastmeyer M, Schröder RR, Gräter F, Blasco E. Responsive 3D Printed Microstructures Based on Collagen Folding and Unfolding. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408597. [PMID: 39604251 PMCID: PMC11753499 DOI: 10.1002/smll.202408597] [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: 10/15/2024] [Indexed: 11/29/2024]
Abstract
Mimicking extracellular matrices holds great potential for tissue engineering in biological and biomedical applications. A key compound for the mechanical stability of these matrices is collagen, which also plays an important role in many intra- and intercellular processes. Two-photon 3D laser printing offers structuring of these matrices with subcellular resolution. So far, efforts on 3D microprinting of collagen have been limited to simple geometries and customized set-ups. Herein, an easily accessible approach is presented using a collagen type I methacrylamide (ColMA) ink system which can be stored at room temperature and be precisely printed using a commercial two-photon 3D laser printer. The formulation and printing parameters are carefully optimized enabling the manufacturing of defined 3D microstructures. Furthermore, these printed microstructures show a fully reversible response upon heating and cooling in multiple cycles, indicating successful collagen folding and unfolding. This experimental observation has been supported by molecular dynamics simulations. Thus, the study opens new perspectives for designing new responsive biomaterials for 4D (micro)printing.
Collapse
Affiliation(s)
- Philipp Mainik
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg University69120HeidelbergGermany
- Organic Chemistry Institute (OCI)Heidelberg University69120HeidelbergGermany
| | | | - Magdalena Fladung
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology (KIT)76131KarlsruheGermany
| | | | - Irene Wacker
- BioQuantHeidelberg University69120HeidelbergGermany
| | - Götz Hofhaus
- BioQuantHeidelberg University69120HeidelbergGermany
| | - Martin Bastmeyer
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology (KIT)76131KarlsruheGermany
- Institute for Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP)Karlsruhe Institute of Technology (KIT)76344KarlsruheGermany
| | | | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS)69118HeidelbergGermany
- Interdisciplinary Center for Scientific Computing (IWR)Heidelberg University69120HeidelbergGermany
| | - Eva Blasco
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg University69120HeidelbergGermany
- Organic Chemistry Institute (OCI)Heidelberg University69120HeidelbergGermany
| |
Collapse
|
5
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [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: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
6
|
Li S, Zhang H, Sun L, Zhang X, Guo M, Liu J, Wang W, Zhao N. 4D printing of biological macromolecules employing handheld bioprinters for in situ wound healing applications. Int J Biol Macromol 2024; 280:135999. [PMID: 39326614 DOI: 10.1016/j.ijbiomac.2024.135999] [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: 07/02/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
In situ bioprinting may be preferred over standard in vitro bioprinting in specific cases when de novo tissues are to be created directly on the appropriate anatomical region in the live organism, employing the body as a bioreactor. So far, few efforts have been made to create in situ tissues that can be safely halted and immobilized during printing in preclinical live animals. However, the technique has to be improved significantly in order to manufacture complex tissues in situ, which may be attainable in the future thanks to multidisciplinary advances in tissue engineering. Thanks to the biological macromolecules, natural and synthetic hydrogels and polymers are among the most used biomaterials in in situ bioprinting procedure. Bioprinters, which encounter multiple challenges, including cross-linking the printed structure, adjusting the rheology parameters, and printing various constructs. The introduction of handheld 3D and 4D bioprinters might potentially overcome the difficulties and problems associated with using traditional bioprinters. Studies showed that this technique could be efficient in wound healing and skin tissue regeneration. This study aims to analyze the benefits and difficulties associated with materials in situ 4D printing via handheld bioprinters.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Hongyang Zhang
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Lei Sun
- Department of Thoracic surgery, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Xinyue Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China
| | - Meiqi Guo
- China Medical University, Shenyang, 110122, Liaoning, China
| | - Jingyang Liu
- China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China.
| | - Ning Zhao
- Department of Otolaryngology, The First Hospital of China Medical University, 155 Nanjing Street, Heping, Shenyang, Liaoning 110001, China.
| |
Collapse
|
7
|
Desai N, Pande S, Vora L, Kommineni N. Correction to "Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration". ACS APPLIED BIO MATERIALS 2024; 7:6325-6331. [PMID: 39162584 PMCID: PMC11409221 DOI: 10.1021/acsabm.4c01057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 08/21/2024]
|
8
|
Abuhamad AY, Masri S, Fadilah NIM, Alamassi MN, Maarof M, Fauzi MB. Application of 3D-Printed Bioinks in Chronic Wound Healing: A Scoping Review. Polymers (Basel) 2024; 16:2456. [PMID: 39274089 PMCID: PMC11397625 DOI: 10.3390/polym16172456] [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: 07/18/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/16/2024] Open
Abstract
Chronic wounds, such as diabetic foot ulcers, pressure ulcers, and venous ulcers, pose significant clinical challenges and burden healthcare systems worldwide. The advent of 3D bioprinting technologies offers innovative solutions for enhancing chronic wound care. This scoping review evaluates the applications, methodologies, and effectiveness of 3D-printed bioinks in chronic wound healing, focusing on bioinks incorporating living cells to facilitate wound closure and tissue regeneration. Relevant studies were identified through comprehensive searches in databases, including PubMed, Scopus, and Web of Science databases, following strict inclusion criteria. These studies employ various 3D bioprinting techniques, predominantly extrusion-based, to create bioinks from natural or synthetic polymers. These bioinks are designed to support cell viability, promote angiogenesis, and provide structural integrity to the wound site. Despite these promising results, further research is necessary to optimize bioink formulations and printing parameters for clinical application. Overall, 3D-printed bioinks offer a transformative approach to chronic wound care, providing tailored and efficient solutions. Continued development and refinement of these technologies hold significant promise for improving chronic wound management and patient outcomes.
Collapse
Affiliation(s)
- Asmaa Y. Abuhamad
- Department for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (A.Y.A.); (S.M.); (N.I.M.F.); (M.M.)
| | - Syafira Masri
- Department for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (A.Y.A.); (S.M.); (N.I.M.F.); (M.M.)
| | - Nur Izzah Md Fadilah
- Department for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (A.Y.A.); (S.M.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Mohammed Numan Alamassi
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Manira Maarof
- Department for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (A.Y.A.); (S.M.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Mh Busra Fauzi
- Department for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (A.Y.A.); (S.M.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
9
|
Thonglam J, Nuntanaranont T, Kong X, Meesane J. Tissue scaffolds mimicking hierarchical bone morphology as biomaterials for oral maxillofacial surgery with augmentation: structure, properties, and performance evaluation for in vitrotesting. Biomed Mater 2024; 19:055035. [PMID: 39094618 DOI: 10.1088/1748-605x/ad6ac4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/02/2024] [Indexed: 08/04/2024]
Abstract
In this study, tissue scaffolds mimicking hierarchical morphology are constructed and proposed for bone augmentation. The scaffolds are fabricated using lyophilization, before coating them with collagen (Col). Subsequently, the Col-coated scaffolds undergo a second lyophilization, followed by silk fibroin (SF) coating, and a third lyophilization. Thereafter, the scaffolds are divided into six groups with varying ratios of Col to SF: Col/SF = 7:3, 5:5, 3:7, 10:0, and 0:10, with an SF scaffold serving as the control group. The scaffold morphology is examined using a scanning electron microscope, while molecular and structural formations are characterized by Fourier transform infrared spectrometer and differential scanning calorimeter, respectively. Physical and mechanical properties including swelling and compression are tested. Biological functions are assessed throughin vitroosteoblast cell culturing. Biomarkers indicative of bone formation-cell viability and proliferation, alkaline phosphatase activity, and calcium content-are analyzed. Results demonstrate that scaffolds coated with Col and SF exhibit sub-porous formations within the main pore. The molecular formation reveals interactions between the hydrophilic groups of Col and SF. The scaffold structure contains bound water and SF formation gets disrupted by Col. Physical and mechanical properties are influenced by the Col/SF ratio and morphology due to coating. The biological functions of scaffolds with Col and SF coating show enhanced potential for promoting bone tissue formation, particularly the Col/SF (7:3) ratio, which is most suitable for bone augmentation in small defect areas.
Collapse
Affiliation(s)
- Jutakan Thonglam
- Institute of Biomedical Engineering, Department of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Thongchai Nuntanaranont
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Jirut Meesane
- Institute of Biomedical Engineering, Department of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| |
Collapse
|
10
|
Desai N, Pande S, Vora LK, Kommineni N. Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration. ACS APPLIED BIO MATERIALS 2024; 7:4270-4292. [PMID: 38950103 PMCID: PMC11253102 DOI: 10.1021/acsabm.4c00613] [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: 05/04/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Bone, a fundamental constituent of the human body, is a vital scaffold for support, protection, and locomotion, underscoring its pivotal role in maintaining skeletal integrity and overall functionality. However, factors such as trauma, disease, or aging can compromise bone structure, necessitating effective strategies for regeneration. Traditional approaches often lack biomimetic environments conducive to efficient tissue repair. Nanofibrous microspheres (NFMS) present a promising biomimetic platform for bone regeneration by mimicking the native extracellular matrix architecture. Through optimized fabrication techniques and the incorporation of active biomolecular components, NFMS can precisely replicate the nanostructure and biochemical cues essential for osteogenesis promotion. Furthermore, NFMS exhibit versatile properties, including tunable morphology, mechanical strength, and controlled release kinetics, augmenting their suitability for tailored bone tissue engineering applications. NFMS enhance cell recruitment, attachment, and proliferation, while promoting osteogenic differentiation and mineralization, thereby accelerating bone healing. This review highlights the pivotal role of NFMS in bone tissue engineering, elucidating their design principles and key attributes. By examining recent preclinical applications, we assess their current clinical status and discuss critical considerations for potential clinical translation. This review offers crucial insights for researchers at the intersection of biomaterials and tissue engineering, highlighting developments in this expanding field.
Collapse
Affiliation(s)
- Nimeet Desai
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Shreya Pande
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Lalitkumar K. Vora
- School
of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| |
Collapse
|
11
|
Razavi ZS, Soltani M, Mahmoudvand G, Farokhi S, Karimi-Rouzbahani A, Farasati-Far B, Tahmasebi-Ghorabi S, Pazoki-Toroudi H, Afkhami H. Advancements in tissue engineering for cardiovascular health: a biomedical engineering perspective. Front Bioeng Biotechnol 2024; 12:1385124. [PMID: 38882638 PMCID: PMC11176440 DOI: 10.3389/fbioe.2024.1385124] [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: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Myocardial infarction (MI) stands as a prominent contributor to global cardiovascular disease (CVD) mortality rates. Acute MI (AMI) can result in the loss of a large number of cardiomyocytes (CMs), which the adult heart struggles to replenish due to its limited regenerative capacity. Consequently, this deficit in CMs often precipitates severe complications such as heart failure (HF), with whole heart transplantation remaining the sole definitive treatment option, albeit constrained by inherent limitations. In response to these challenges, the integration of bio-functional materials within cardiac tissue engineering has emerged as a groundbreaking approach with significant potential for cardiac tissue replacement. Bioengineering strategies entail fortifying or substituting biological tissues through the orchestrated interplay of cells, engineering methodologies, and innovative materials. Biomaterial scaffolds, crucial in this paradigm, provide the essential microenvironment conducive to the assembly of functional cardiac tissue by encapsulating contracting cells. Indeed, the field of cardiac tissue engineering has witnessed remarkable strides, largely owing to the application of biomaterial scaffolds. However, inherent complexities persist, necessitating further exploration and innovation. This review delves into the pivotal role of biomaterial scaffolds in cardiac tissue engineering, shedding light on their utilization, challenges encountered, and promising avenues for future advancement. By critically examining the current landscape, we aim to catalyze progress toward more effective solutions for cardiac tissue regeneration and ultimately, improved outcomes for patients grappling with cardiovascular ailments.
Collapse
Affiliation(s)
- Zahra-Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Sustainable Business, International Business University, Toronto, ON, Canada
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arian Karimi-Rouzbahani
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bahareh Farasati-Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Samaneh Tahmasebi-Ghorabi
- Master of Health Education, Research Expert, Clinical Research Development Unit, Emam Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
12
|
Luo C, Li YM, Jiang K, Wang K, Kuzmanović M, You XH, Zhang Y, Lei J, Huang SS, Xu JZ. ECM-inspired calcium/zinc laden cellulose scaffold for enhanced bone regeneration. Carbohydr Polym 2024; 331:121823. [PMID: 38388030 DOI: 10.1016/j.carbpol.2024.121823] [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: 10/18/2023] [Revised: 01/01/2024] [Accepted: 01/11/2024] [Indexed: 02/24/2024]
Abstract
Cellulose-based polymer scaffolds are highly diverse for designing and fabricating artificial bone substitutes. However, realizing the multi-biological functions of cellulose-based scaffolds has long been challenging. In this work, inspired by the structure and function of the extracellular matrix (ECM) of bone, we developed a novel yet feasible strategy to prepare ECM-like scaffolds with hybrid calcium/zinc mineralization. The 3D porous structure was formed via selective oxidation and freeze drying of bacterial cellulose. Following the principle of electrostatic interaction, calcium/zinc hybrid hydroxyapatite nucleated, crystallized, and precipitated on the 3D scaffold in simulated physiological conditions, which was well confirmed by morphology and composition analysis. Compared with alternative scaffold cohorts, this hybrid ion-loaded cellulose scaffold exhibited a pronounced elevation in alkaline phosphatase (ALP) activity, osteogenic gene expression, and cranial defect regeneration. Notably, the hybrid ion-loaded cellulose scaffold effectively fostered an M2 macrophage milieu and had a strong immune effect in vivo. In summary, this study developed a hybrid multifunctional cellulose-based scaffold that appropriately simulates the ECM to regulate immunomodulatory and osteogenic differentiation, setting a measure for artificial bone substitutes.
Collapse
Affiliation(s)
- Chuan Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yuan-Min Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Kai Jiang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Maja Kuzmanović
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xuan-He You
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jun Lei
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Shi-Shu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China.
| | - Jia-Zhuang Xu
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
13
|
Qiao S, Peijie T, Nan J. Crosslinking strategies of decellularized extracellular matrix in tissue regeneration. J Biomed Mater Res A 2024; 112:640-671. [PMID: 37990863 DOI: 10.1002/jbm.a.37650] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023]
Abstract
By removing the immunogenic cellular components through various decellularization methods, decellularized extracellular matrix (dECM) is considered a promising material in the field of tissue engineering and regenerative medicine with highly preserved physicochemical properties and superior biocompatibility. However, decellularization treatment can lead to some loss of structural integrity, mechanical strength, degradation stability, and biological performance of dECM biomaterials. Therefore, physical and chemical crosslinking methods are preferred to restore or even improve the biomechanical properties, stability, and bioactivity, and to achieve a delicate balance between degradation of the implanted biomaterial and regeneration of the host tissue. This review provides an overview of dECM biomaterials, and describes and compares the mechanisms and characteristics of commonly used crosslinking methods for dECM, with a focus on the potential applications of versatile dECM-based biomaterials derived from skin, cardiac tissues (pericardium, heart valves, myocardial tissue), blood vessels, liver, and kidney, modified with different chemical crosslinking reagents, in tissue and organ regeneration.
Collapse
Affiliation(s)
- Su Qiao
- State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Tan Peijie
- State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiang Nan
- State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Zhang X, Cheng F, Islam MR, Li H. The fabrication of the chitosan-based bioink for in vitro tissue repair and regeneration: A review. Int J Biol Macromol 2024; 257:128504. [PMID: 38040155 DOI: 10.1016/j.ijbiomac.2023.128504] [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: 07/30/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
The repair and regeneration of the injured tissues or organs is a major challenge for biomedicine, and the emerging 3D bioprinting technology as a class of promising techniques in biomedical research for the development of tissue engineering and regenerative medicine. Chitosan-based bioinks, as the natural biomaterials, are considered as ideal materials for 3D bioprinting to design and fabricate the various scaffold due to their unique dynamic reversibility and fantastic biological properties. Our review aims to provide an overview of chitosan-based bioinks for in vitro tissue repair and regeneration, starting from modification of chitosan that affect these bioprinting processes. In addition, we summarize the advances in chitosan-based bioinks used in the various 3D printing strategies. Moreover, the biomedical applications of chitosan-based bioinks are discussed, primarily centered on regenerative medicine and tissue modeling engineering. Finally, current challenges and future opportunities in this field are discussed. The combination of chitosan-based bioinks and 3D bioprinting will hold promise for developing novel biomedical scaffolds for tissue or organ repair and regeneration.
Collapse
Affiliation(s)
- Xiao Zhang
- School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, Harbin Institute of Technology, Harbin, Heilongjiang 150001, PR China
| | - Feng Cheng
- School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, Harbin Institute of Technology, Harbin, Heilongjiang 150001, PR China.
| | - Md Rashidul Islam
- College of Light Industry and Textile, Qiqihar University, Qiqihar, Heilongjiang 161000, PR China
| | - Hongbin Li
- School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, Harbin Institute of Technology, Harbin, Heilongjiang 150001, PR China; College of Light Industry and Textile, Qiqihar University, Qiqihar, Heilongjiang 161000, PR China.
| |
Collapse
|
15
|
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: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
16
|
Dortaj H, Azarpira N, Pakbaz S. Insight to Biofabrication of Liver Microtissues for Disease Modeling: Challenges and Opportunities. Curr Stem Cell Res Ther 2024; 19:1303-1311. [PMID: 37846577 DOI: 10.2174/011574888x257744231009071810] [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: 04/13/2023] [Revised: 08/26/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023]
Abstract
In the last decade, liver diseases with high mortality rates have become one of the most important health problems in the world. Organ transplantation is currently considered the most effective treatment for compensatory liver failure. An increasing number of patients and shortage of donors has led to the attention of reconstructive medicine methods researchers. The biggest challenge in the development of drugs effective in chronic liver disease is the lack of a suitable preclinical model that can mimic the microenvironment of liver problems. Organoid technology is a rapidly evolving field that enables researchers to reconstruct, evaluate, and manipulate intricate biological processes in vitro. These systems provide a biomimetic model for studying the intercellular interactions necessary for proper organ function and architecture in vivo. Liver organoids, formed by the self-assembly of hepatocytes, are microtissues and can exhibit specific liver characteristics for a long time in vitro. Hepatic organoids are identified as an impressive tool for evaluating potential cures and modeling liver diseases. Modeling various liver diseases, including tumors, fibrosis, non-alcoholic fatty liver, etc., allows the study of the effects of various drugs on these diseases in personalized medicine. Here, we summarize the literature relating to the hepatic stem cell microenvironment and the formation of liver Organoids.
Collapse
Affiliation(s)
- Hengameh Dortaj
- Department of Tissue Engineering and Applied Cell Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Pakbaz
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
17
|
Dobaj Štiglic A, Lackner F, Nagaraj C, Beaumont M, Bračič M, Duarte I, Kononenko V, Drobne D, Madhan B, Finšgar M, Kargl R, Stana Kleinschek K, Mohan T. 3D-Printed Collagen-Nanocellulose Hybrid Bioscaffolds with Tailored Properties for Tissue Engineering Applications. ACS APPLIED BIO MATERIALS 2023; 6:5596-5608. [PMID: 38050684 PMCID: PMC10731651 DOI: 10.1021/acsabm.3c00767] [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: 09/06/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/06/2023]
Abstract
Hybrid collagen (Coll) bioscaffolds have emerged as a promising solution for tissue engineering (TE) and regenerative medicine. These innovative bioscaffolds combine the beneficial properties of Coll, an important structural protein of the extracellular matrix, with various other biomaterials to create platforms for long-term cell growth and tissue formation. The integration or cross-linking of Coll with other biomaterials increases mechanical strength and stability and introduces tailored biochemical and physical factors that mimic the natural tissue microenvironment. This work reports on the fabrication of chemically cross-linked hybrid bioscaffolds with enhanced properties from the combination of Coll, nanofibrillated cellulose (NFC), carboxymethylcellulose (CMC), and citric acid (CA). The bioscaffolds were prepared by 3D printing ink containing Coll-NFC-CMC-CA followed by freeze-drying, dehydrothermal treatment, and neutralization. Cross-linking through the formation of ester bonds between the polymers and CA in the bioscaffolds was achieved by exposing the bioscaffolds to elevated temperatures in the dry state. The morphology, pores/porosity, chemical composition, structure, thermal behavior, swelling, degradation, and mechanical properties of the bioscaffolds in the dry and wet states were investigated as a function of Coll concentration. The bioscaffolds showed no cytotoxicity to MG-63 human bone osteosarcoma cells as tested by different assays measuring different end points. Overall, the presented hybrid Coll bioscaffolds offer a unique combination of biocompatibility, stability, and structural support, making them valuable tools for TE.
Collapse
Affiliation(s)
- Andreja Dobaj Štiglic
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Faculty
of Chemistry and Chemical Engineering, Laboratory for Analytical Chemistry
and Industrial Analysis, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Florian Lackner
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Chandran Nagaraj
- Ludwig
Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, 8010 Graz, Austria
| | - Marco Beaumont
- Department
of Chemistry, Institute of Chemistry o Renewable Resources, University of Natural Resources and Life Sciences
Vienna (BOKU), A-3430 Tulln, Austria
| | - Matej Bračič
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Isabel Duarte
- Department
of Mechanical Engineering, Centre for Mechanical Technology and Automation
(TEMA), Intelligent Systems Associate Laboratory (LASI), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Veno Kononenko
- Department
of Biology, Biotechnical Faculty, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Damjana Drobne
- Department
of Biology, Biotechnical Faculty, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Balaraman Madhan
- CSIR-Central
Leather Research Institute, Chennai 600 020, Tamil Nadu, India
| | - Matjaž Finšgar
- Faculty
of Chemistry and Chemical Engineering, Laboratory for Analytical Chemistry
and Industrial Analysis, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Rupert Kargl
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Karin Stana Kleinschek
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
- Institute
of Automation, Faculty of Electrical Engineering and Computer Science, University of Maribor, Koroska cesta 46, 2000 Maribor, Slovenia
| | - Tamilselvan Mohan
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| |
Collapse
|
18
|
Cruz-Maya I, Altobelli R, Alvarez-Perez MA, Guarino V. Mineralized Microgels via Electrohydrodynamic Atomization: Optimization and In Vitro Model for Dentin-Pulp Complex. Gels 2023; 9:846. [PMID: 37998935 PMCID: PMC10670945 DOI: 10.3390/gels9110846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
There is growing interest in the use of micro-sized hydrogels, including bioactive signals, as efficient platforms for tissue regeneration because they are able to mimic cell niche structure and selected functionalities. Herein, it is proposed to optimize bioactive composite microgels via electrohydrodynamic atomization (EHDA) to regenerate the dentin-pulp complex. The addition of disodium phosphate (Na2HPO4) salts as mineral precursors triggered an in situ reaction with divalent ions in solution, thus promoting the encapsulation of different amounts of apatite-like phases. Morphological analysis via image analysis of optical images confirmed a narrow distribution of perfectly rounded particles, with an average diameter ranging from 223 ± 18 μm to 502 ± 64 μm as a function of mineral content and process parameters used. FTIR, TEM, and EDAX analyses confirmed the formation of calcium phosphates with a characteristic Ca/P ratio close to 1.67 and a needle-like crystal shape. In vitro studies-using dental pulp stem cells (DPSCs) in crown sections of natural teeth slices-showed an increase in cell viability until 14 days, recording a decay of proliferation at 21 days, independent on the mineral amount, suggesting that differentiation is started, as confirmed by the increase of ALP activity at 14 days. In this view, mineralized microgels could be successfully used to support in vitro osteogenesis, working as an interesting model to study dental tissue regeneration.
Collapse
Affiliation(s)
- Iriczalli Cruz-Maya
- Institute of Polymers, Composites and Biomaterials (IPCB), National Research Council of Italy, Mostra d’Oltremare Pad. 20, Viale J.F. Kennedy 54, 80125 Naples, Italy
- Tissue Bioengineering Laboratory of DEPeI-FO, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Rosaria Altobelli
- Institute of Polymers, Composites and Biomaterials (IPCB), National Research Council of Italy, Mostra d’Oltremare Pad. 20, Viale J.F. Kennedy 54, 80125 Naples, Italy
| | - Marco Antonio Alvarez-Perez
- Tissue Bioengineering Laboratory of DEPeI-FO, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Vincenzo Guarino
- Institute of Polymers, Composites and Biomaterials (IPCB), National Research Council of Italy, Mostra d’Oltremare Pad. 20, Viale J.F. Kennedy 54, 80125 Naples, Italy
| |
Collapse
|
19
|
M Galindo J, San-Millán I, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Herrero MA, Merino S, Vázquez E. Mimicking the extracellular matrix by incorporating functionalized graphene into hybrid hydrogels. NANOSCALE 2023; 15:14238-14248. [PMID: 37599610 DOI: 10.1039/d3nr02689b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The efficient functionalization of graphene with sulfonic groups using a sustainable approach facilitates the interaction of biomolecules with its surface. The inclusion of these graphene sheets inside a photopolymerized acrylamide-based hydrogel provides a 3D scaffold with viscoelastic behaviour closer to that found in natural tissues. Cell-culture experiments and differentiation assays with SH-SY5Y cells showed that these hybrid hydrogels are non-cytotoxic, thus making them potentially useful as scaffold materials mimicking the extracellular environment.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | | | | | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| |
Collapse
|
20
|
Stenlund P, Enstedt L, Gilljam KM, Standoft S, Ahlinder A, Lundin Johnson M, Lund H, Millqvist Fureby A, Berglin M. Development of an All-Marine 3D Printed Bioactive Hydrogel Dressing for Treatment of Hard-to-Heal Wounds. Polymers (Basel) 2023; 15:2627. [PMID: 37376274 DOI: 10.3390/polym15122627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Current standard wound care involves dressings that provide moisture and protection; however, dressings providing active healing are still scarce and expensive. We aimed to develop an ecologically sustainable 3D printed bioactive hydrogel-based topical wound dressing targeting healing of hard-to-heal wounds, such as chronic or burn wounds, which are low on exudate. To this end, we developed a formulation composed of renewable marine components; purified extract from unfertilized salmon roe (heat-treated X, HTX), alginate from brown seaweed, and nanocellulose from tunicates. HTX is believed to facilitate the wound healing process. The components were successfully formulated into a 3D printable ink that was used to create a hydrogel lattice structure. The 3D printed hydrogel showed a HTX release profile enhancing pro-collagen I alpha 1 production in cell culture with potential of promoting wound closure rates. The dressing has recently been tested on burn wounds in Göttingen minipigs and shows accelerated wound closure and reduced inflammation. This paper describes the dressings development, mechanical properties, bioactivity, and safety.
Collapse
Affiliation(s)
- Patrik Stenlund
- Department of Methodology, Textile and Medical Technology, RISE Research Institutes of Sweden AB, Arvid Wallgrens backe 20, SE-413 46 Gothenburg, Sweden
| | - Linnea Enstedt
- Department of Chemical Process and Pharmaceutical Development, RISE Research Institutes of Sweden AB, Drottning Kristinas väg 61B, SE-114 28 Stockholm, Sweden
| | | | - Simon Standoft
- Department of Methodology, Textile and Medical Technology, RISE Research Institutes of Sweden AB, Arvid Wallgrens backe 20, SE-413 46 Gothenburg, Sweden
| | - Astrid Ahlinder
- Department of Agriculture and Food, RISE Research Institutes of Sweden AB, Frans Perssons väg 6, SE-412 76 Gothenburg, Sweden
| | - Maria Lundin Johnson
- Department of Chemical Process and Pharmaceutical Development, RISE Research Institutes of Sweden AB, Drottning Kristinas väg 61B, SE-114 28 Stockholm, Sweden
| | - Henrik Lund
- Regenics AS, Gaustadalléen 21, N-0349 Oslo, Norway
| | - Anna Millqvist Fureby
- Department of Chemical Process and Pharmaceutical Development, RISE Research Institutes of Sweden AB, Drottning Kristinas väg 61B, SE-114 28 Stockholm, Sweden
| | - Mattias Berglin
- Department of Methodology, Textile and Medical Technology, RISE Research Institutes of Sweden AB, Arvid Wallgrens backe 20, SE-413 46 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, Kemigården 4, SE-412 96 Gothenburg, Sweden
| |
Collapse
|
21
|
Łach A, Wnuk A, Wójtowicz AK. Experimental Models to Study the Functions of the Blood-Brain Barrier. Bioengineering (Basel) 2023; 10:bioengineering10050519. [PMID: 37237588 DOI: 10.3390/bioengineering10050519] [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: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
The purpose of this paper was to discuss the achievements of in vitro modeling in terms of the blood-brain barrier [BBB] and to create a clear overview of this research area, which is useful in research planning. The text was divided into three main parts. The first part describes the BBB as a functional structure, its constitution, cellular and noncellular components, mechanisms of functioning and importance for the central nervous system, in terms of both protection and nourishment. The second part is an overview of parameters important in terms of establishing and maintaining a barrier phenotype that allows for formulating criteria of evaluation of the BBB in vitro models. The third and last part discusses certain techniques for developing the BBB in vitro models. It describes subsequent research approaches and models, as they underwent change alongside technological advancement. On the one hand, we discuss possibilities and limitations of different research approaches: primary cultures vs. cell lines and monocultures vs. multicultures. On the other hand, we review advantages and disadvantages of specific models, such as models-on-a-chip, 3D models or microfluidic models. We not only attempt to state the usefulness of specific models in different kinds of research on the BBB but also emphasize the significance of this area of research for advancement of neuroscience and the pharmaceutical industry.
Collapse
Affiliation(s)
- Andrzej Łach
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| | - Agnieszka Wnuk
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Anna Katarzyna Wójtowicz
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| |
Collapse
|
22
|
Cao S, Bo R, Zhang Y. Polymeric Scaffolds for Regeneration of Central/Peripheral Nerves and Soft Connective Tissues. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Shunze Cao
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| | - Renheng Bo
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| | - Yihui Zhang
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| |
Collapse
|
23
|
Trikalitis VD, Kroese NJJ, Kaya M, Cofiño-Fabres C, Ten Den S, Khalil ISM, Misra S, Koopman BFJM, Passier R, Schwach V, Rouwkema J. Embedded 3D printing of dilute particle suspensions into dense complex tissue fibers using shear thinning xanthan baths. Biofabrication 2022; 15. [PMID: 36347040 DOI: 10.1088/1758-5090/aca124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022]
Abstract
In order to fabricate functional organoids and microtissues, a high cell density is generally required. As such, the placement of cell suspensions in molds or microwells to allow for cell concentration by sedimentation is the current standard for the production of organoids and microtissues. Even though molds offer some level of control over the shape of the resulting microtissue, this control is limited as microtissues tend to compact towards a sphere after sedimentation of the cells. 3D bioprinting on the other hand offers complete control over the shape of the resulting structure. Even though the printing of dense cell suspensions in the ink has been reported, extruding dense cellular suspensions is challenging and generally results in high shear stresses on the cells and a poor shape fidelity of the print. As such, additional materials such as hydrogels are added in the bioink to limit shear stresses, and to improve shape fidelity and resolution. The maximum cell concentration that can be incorporated in a hydrogel-based ink before the ink's rheological properties are compromised, is significantly lower than the concentration in a tissue equivalent. Additionally, the hydrogel components often interfere with cellular self-assembly processes. To circumvent these limitations, we report a simple and inexpensive xanthan bath based embedded printing method to 3D print dense functional linear tissues using dilute particle suspensions consisting of cells, spheroids, hydrogel beads, or combinations thereof. Using this method, we demonstrated the self-organization of functional cardiac tissue fibers with a layer of epicardial cells surrounding a body of cardiomyocytes.
Collapse
Affiliation(s)
- Vasileios D Trikalitis
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Niels J J Kroese
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Mert Kaya
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Simone Ten Den
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Islam S M Khalil
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands
| | - Sarthak Misra
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Bart F J M Koopman
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| |
Collapse
|
24
|
Shen Y, Cao X, Lu M, Gu H, Li M, Posner DA. Current treatments after spinal cord injury: Cell engineering, tissue engineering, and combined therapies. SMART MEDICINE 2022; 1:e20220017. [PMID: 39188731 PMCID: PMC11235943 DOI: 10.1002/smmd.20220017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/20/2022] [Indexed: 08/28/2024]
Abstract
Both traumatic and non-traumatic spinal cord injuries (SCIs) can be categorized as damages done to our central nervous system (CNS). The patients' physical and mental health may suffer greatly because of traumatic SCI. With the widespread use of motor vehicles and increasingly aged population, the occurrence of SCI is more frequent than before, creating a considerable burden to global public health. The regeneration process of the spinal cord is hampered by a series of events that occur following SCI like edema, hemorrhage, formation of cystic cavities, and ischemia. An effective strategy for the treatment of SCI and functional recovery still has not been discovered; however, recent advances have been made in bioengineering fields that therapies based on cells, biomaterials, and biomolecules have proved effective in the repair of the spinal cord. In the light of worldwide importance of treatments for SCI, this article aims to provide a review of recent advances by first introducing the physiology, etiology, epidemiology, and mechanisms of SCI. We then put emphasis on the widely used clinical treatments and bioengineering strategies (cell-based, biomaterial-based, and biomolecule-based) for the functional regeneration of the spinal cord as well as challenges faced by scientists currently. This article provides scientists and clinicians with a comprehensive outlook on the recent advances of preclinical and clinical treatments of SCI, hoping to help them find keys to the functional regeneration of SCI.
Collapse
Affiliation(s)
- Yingbo Shen
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Xinyue Cao
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Minhui Lu
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Hongcheng Gu
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Minli Li
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - David A. Posner
- Molecular Immunity UnitCambridge Institute of Therapeutic Immunology and Infectious DiseasesDepartment of MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
25
|
Naranda J, Bračič M, Vogrin M, Maver U, Trojner T. Practical Use of Quartz Crystal Microbalance Monitoring in Cartilage Tissue Engineering. J Funct Biomater 2022; 13:jfb13040159. [PMID: 36278628 PMCID: PMC9590066 DOI: 10.3390/jfb13040159] [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: 08/09/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
Quartz crystal microbalance (QCM) is a real-time, nanogram-accurate technique for analyzing various processes on biomaterial surfaces. QCM has proven to be an excellent tool in tissue engineering as it can monitor key parameters in developing cellular scaffolds. This review focuses on the use of QCM in the tissue engineering of cartilage. It begins with a brief discussion of biomaterials and the current state of the art in scaffold development for cartilage tissue engineering, followed by a summary of the potential uses of QCM in cartilage tissue engineering. This includes monitoring interactions with extracellular matrix components, adsorption of proteins onto biomaterials, and biomaterial–cell interactions. In the last part of the review, the material selection problem in tissue engineering is highlighted, emphasizing the importance of surface nanotopography, the role of nanofilms, and utilization of QCM as a “screening” tool to improve the material selection process. A step-by-step process for scaffold design is proposed, as well as the fabrication of thin nanofilms in a layer-by-layer manner using QCM. Finally, future trends of QCM application as a “screening” method for 3D printing of cellular scaffolds are envisioned.
Collapse
Affiliation(s)
- Jakob Naranda
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
- Department of Orthopaedics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
- Correspondence: (J.N.); (M.B.); Tel.: +386-2-321-1541 (J.N.); +386-2-220-7929 (M.B.)
| | - Matej Bračič
- Laboratory for Characterisation and Processing of Polymers (LCPP), Faculty of Mechanical Engineering, University of Maribor, SI-2000 Maribor, Slovenia
- Correspondence: (J.N.); (M.B.); Tel.: +386-2-321-1541 (J.N.); +386-2-220-7929 (M.B.)
| | - Matjaž Vogrin
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
- Department of Orthopaedics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
- Department of Pharmacology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| | - Teodor Trojner
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
| |
Collapse
|
26
|
Hospodiuk-Karwowski M, Chi K, Pritchard J, Catchmark JM. Vascularized pancreas-on-a-chip device produced using a printable simulated extracellular matrix. Biomed Mater 2022; 17. [PMID: 36001993 DOI: 10.1088/1748-605x/ac8c74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
The extracellular matrix (ECM) influences cellular behavior, function, and fate. The ECM surrounding Langerhans islets has not been investigated in detail to explain its role in the development and maturation of pancreatic β-cells. Herein, a complex combination of the simulated ECM (sECM) has been examined with a comprehensive analysis of cell response and a variety of controls. The most promising results were obtained from group containing fibrin, collagen type I, Matrigel®, hyaluronic acid, methylcellulose, and two compounds of functionalized, ionically crosslinking bacterial cellulose (sECMbc). Even though the cell viability was not significantly impacted, the performance of group of sECMbc showed 2 to 4x higher sprouting number and length, 2 to 4x higher insulin secretion in static conditions, and 2 to 10x higher gene expression of VEGF-A, Endothelin-1, and NOS3 than the control group of fibrin matrix (sECMf). Each material was tested in a hydrogel-based, perfusable, pancreas-on-a-chip device and the best group - sECMbc has been tested with the drug Sunitinib to show the extended possibilities of the device for both diabetes-like screening as well as PDAC chemotherapeutics screening for potential personal medicine approach. It proved its functionality in 7 days dynamic culture and is suitable as a physiological tissue model. Moreover, the device with the pancreatic-like spheroids was 3D bioprintable and perfusable.
Collapse
Affiliation(s)
- Monika Hospodiuk-Karwowski
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Kai Chi
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Justin Pritchard
- Biomedical Engineering Department, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Jeffrey M Catchmark
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| |
Collapse
|
27
|
Abstract
Vascular transplantation is an effective and common treatment for cardiovascular disease (CVD). However, the low biocompatibility of implants is a major problem that hinders its clinical application. Surface modification of implants with extracellular matrix (ECM) coatings is an effective approach to improve the biocompatibility of cardiovascular materials. The complete ECM seems to have better biocompatibility, which may give cardiovascular biomaterials a more functional surface. The use of one or several ECM proteins to construct a surface allows customization of coating composition and structure, possibly resulting in some unique functions. ECM is a complex three-dimensional structure composed of a variety of functional biological macromolecules, and changes in the composition will directly affect the function of the coating. Therefore, understanding the chemical composition of the ECM and its interaction with cells is beneficial to provide new approaches for coating surface modification. This article reviews novel ECM coatings, including coatings composed of intact ECM and biomimetic coatings tailored from several ECM proteins, and introduces new advances in coating fabrication. These ECM coatings are effective in improving the biocompatibility of vascular grafts.
Collapse
|
28
|
Zhang CY, Fu CP, Li XY, Lu XC, Hu LG, Kankala RK, Wang SB, Chen AZ. Three-Dimensional Bioprinting of Decellularized Extracellular Matrix-Based Bioinks for Tissue Engineering. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113442. [PMID: 35684380 PMCID: PMC9182049 DOI: 10.3390/molecules27113442] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 01/01/2023]
Abstract
Three-dimensional (3D) bioprinting is one of the most promising additive manufacturing technologies for fabricating various biomimetic architectures of tissues and organs. In this context, the bioink, a critical element for biofabrication, is a mixture of biomaterials and living cells used in 3D printing to create cell-laden structures. Recently, decellularized extracellular matrix (dECM)-based bioinks derived from natural tissues have garnered enormous attention from researchers due to their unique and complex biochemical properties. This review initially presents the details of the natural ECM and its role in cell growth and metabolism. Further, we briefly emphasize the commonly used decellularization treatment procedures and subsequent evaluations for the quality control of the dECM. In addition, we summarize some of the common bioink preparation strategies, the 3D bioprinting approaches, and the applicability of 3D-printed dECM bioinks to tissue engineering. Finally, we present some of the challenges in this field and the prospects for future development.
Collapse
Affiliation(s)
- Chun-Yang Zhang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Chao-Ping Fu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- Correspondence: (C.-P.F.); (A.-Z.C.)
| | - Xiong-Ya Li
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Xiao-Chang Lu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Long-Ge Hu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China; (C.-Y.Z.); (X.-Y.L.); (X.-C.L.); (L.-G.H.); (R.K.K.); (S.-B.W.)
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- Correspondence: (C.-P.F.); (A.-Z.C.)
| |
Collapse
|
29
|
Taniguchi Nagahara MH, Caiado Decarli M, Inforçatti Neto P, Lopes da Silva JV, Moraes ÂM. Crosslinked alginate‐xanthan gum blends as effective hydrogels for
3D
bioprinting of biological tissues. J Appl Polym Sci 2022. [DOI: 10.1002/app.52612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Mariana Harue Taniguchi Nagahara
- Department of Engineering of Materials and of Bioprocesses, School of Chemical Engineering University of Campinas Campinas São Paulo Brazil
| | - Monize Caiado Decarli
- Department of Engineering of Materials and of Bioprocesses, School of Chemical Engineering University of Campinas Campinas São Paulo Brazil
- Three‐Dimensional Technologies Research Group CTI Renato Archer Campinas São Paulo Brazil
| | - Paulo Inforçatti Neto
- Three‐Dimensional Technologies Research Group CTI Renato Archer Campinas São Paulo Brazil
| | | | - Ângela Maria Moraes
- Department of Engineering of Materials and of Bioprocesses, School of Chemical Engineering University of Campinas Campinas São Paulo Brazil
| |
Collapse
|
30
|
Tang S, Liu K, Chen J, Li Y, Liu M, Lu L, Zhou C, Luo B. Dual-Cross-linked Liquid Crystal Hydrogels with Controllable Viscoelasticity for Regulating Cell Behaviors. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21966-21977. [PMID: 35503918 DOI: 10.1021/acsami.2c02689] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The liquid crystal properties and viscoelasticity of the natural bone extracellular matrix (ECM) play a decisive role in guiding cell behavior, conducting cell signals, and regulating mineralization. Here, we develop a facile approach for preparing a novel polysaccharide hydrogel with liquid crystal properties and viscoelasticity similar to those of natural bone ECM. First, a series of chitin whisker/chitosan (CHW/CS) hydrogels were prepared by chemical cross-linking with genipin, in which CHW can self-assemble to form cholesteric liquid crystals under ultrasonic treatment and CS chains can enter into the gaps between the helical layers of the CHW cholesteric liquid crystal phase to endow morphological stability and good mechanical properties. Subsequently, the obtained chemically cross-linked liquid crystal hydrogels were immersed into the desired concentration of the NaCl solution to form physical cross-linking. Due to the Hofmeister effect, the as-prepared dual-cross-linked liquid crystal hydrogels showed an enhanced modulus, viscoelasticity similar to that of natural ECM with relatively fast stress relaxation behavior, and fold surface morphology. Compared to both CHW/CS hydrogels without liquid crystal properties and CHW/CS liquid crystal hydrogels without further physical cross-linking, the dual-cross-linked CHW/CS liquid crystal hydrogels are more favorable for the adhesion, proliferation, and osteogenic differentiation of bone marrow mesenchymal stem cells. This approach could inspire the design of hydrogels mimicking the liquid crystal properties and viscoelasticity of natural bone ECM for bone repair.
Collapse
Affiliation(s)
- Shengyue Tang
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Jingsheng Chen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Yizhi Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Lu Lu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, P. R. China
| |
Collapse
|
31
|
A 3D in vitro co-culture model for evaluating biomaterial-mediated modulation of foreign-body responses. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
32
|
Shokrani H, Shokrani A, Jouyandeh M, Seidi F, Gholami F, Kar S, Munir MT, Kowalkowska-Zedler D, Zarrintaj P, Rabiee N, Saeb MR. Green Polymer Nanocomposites for Skin Tissue Engineering. ACS APPLIED BIO MATERIALS 2022; 5:2107-2121. [PMID: 35504039 DOI: 10.1021/acsabm.2c00313] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fabrication of an appropriate skin scaffold needs to meet several standards related to the mechanical and biological properties. Fully natural/green scaffolds with acceptable biodegradability, biocompatibility, and physiological properties quite often suffer from poor mechanical properties. Therefore, for appropriate skin tissue engineering and to mimic the real functions, we need to use synthetic polymers and/or additives as complements to green polymers. Green nanocomposites (either nanoscale natural macromolecules or biopolymers containing nanoparticles) are a class of scaffolds with acceptable biomedical properties window (drug delivery and cardiac, nerve, bone, cartilage as well as skin tissue engineering), enabling one to achieve the required level of skin regeneration and wound healing. In this review, we have collected, summarized, screened, analyzed, and interpreted the properties of green nanocomposites used in skin tissue engineering and wound dressing. We particularly emphasize the mechanical and biological properties that skin cells need to meet when seeded on the scaffold. In this regard, the latest state of the art studies directed at fabrication of skin tissue and bionanocomposites as well as their mechanistic features are discussed, whereas some unspoken complexities and challenges for future developments are highlighted.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, 210037 Nanjing, China
| | - Amirhossein Shokrani
- Department of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran
| | - Maryam Jouyandeh
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, 11155-4563 Tehran, Iran
| | - Farzad Seidi
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, 210037 Nanjing, China
| | - Fatemeh Gholami
- New Technologies - Research Centre, University of West Bohemia, Veleslavínova 42, 301 00 Plzeň, Czech Republic
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait
| | - Muhammad Tajammal Munir
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait
| | - Daria Kowalkowska-Zedler
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Payam Zarrintaj
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, P.O. Box 11155-9161, Tehran 145888-9694, Iran.,School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| |
Collapse
|
33
|
Costa BL, Adão RMR, Maibohm C, Accardo A, Cardoso VF, Nieder JB. Cellular Interaction of Bone Marrow Mesenchymal Stem Cells with Polymer and Hydrogel 3D Microscaffold Templates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13013-13024. [PMID: 35282678 PMCID: PMC8949723 DOI: 10.1021/acsami.1c23442] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/03/2022] [Indexed: 05/05/2023]
Abstract
Biomimicking biological niches of healthy tissues or tumors can be achieved by means of artificial microenvironments, where structural and mechanical properties are crucial parameters to promote tissue formation and recreate natural conditions. In this work, three-dimensional (3D) scaffolds based on woodpile structures were fabricated by two-photon polymerization (2PP) of different photosensitive polymers (IP-S and SZ2080) and hydrogels (PEGDA 700) using two different 2PP setups, a commercial one and a customized one. The structures' properties were tuned to study the effect of scaffold dimensions (gap size) and their mechanical properties on the adhesion and proliferation of bone marrow mesenchymal stem cells (BM-MSCs), which can serve as a model for leukemic diseases, among other hematological applications. The woodpile structures feature gap sizes of 25, 50, and 100 μm and a fixed beam diameter of 25 μm, to systematically study the optimal cell colonization that promotes healthy cell growth and potential tissue formation. The characterization of the scaffolds involved scanning electron microscopy and mechanical nanoindenting, while their suitability for supporting cell growth was evaluated with live/dead cell assays and multistaining 3D confocal imaging. In the mechanical assays of the hydrogel material, we observed two different stiffness ranges depending on the indentation depth. Larger gap woodpile structures coated with fibronectin were identified as the most promising scaffolds for 3D BM-MSC cellular models, showing higher proliferation rates. The results indicate that both the design and the employed materials are suitable for further assays, where retaining the BM-MSC stemness and original features is crucial, including studies focused on BM disorders such as leukemia and others. Moreover, the combination of 3D scaffold geometry and materials holds great potential for the investigation of cellular behaviors in a co-culture setting, for example, mesenchymal and hematopoietic stem cells, to be further applied in medical research and pharmacological studies.
Collapse
Affiliation(s)
- Beatriz
N. L. Costa
- INL—International
Iberian Nanotechnology Laboratory, Ultrafast
Bio- and Nanophotonics Group, Av. Mestre José Veiga S/n, 4715-330 Braga, Portugal
- CMEMS-UMinho,
University of Minho, DEI, Campus de Azurém, Guimarães 4800-058, Portugal
- Faculty
of Mechanical, Maritime, and Materials Engineering (3mE), Department
of Precision and Microsystems Engineering (PME), Delft University of Technology, Mekelweg 2, Delft 2628 CD, The Netherlands
| | - Ricardo M. R. Adão
- INL—International
Iberian Nanotechnology Laboratory, Ultrafast
Bio- and Nanophotonics Group, Av. Mestre José Veiga S/n, 4715-330 Braga, Portugal
| | - Christian Maibohm
- INL—International
Iberian Nanotechnology Laboratory, Ultrafast
Bio- and Nanophotonics Group, Av. Mestre José Veiga S/n, 4715-330 Braga, Portugal
| | - Angelo Accardo
- Faculty
of Mechanical, Maritime, and Materials Engineering (3mE), Department
of Precision and Microsystems Engineering (PME), Delft University of Technology, Mekelweg 2, Delft 2628 CD, The Netherlands
| | - Vanessa F. Cardoso
- CMEMS-UMinho,
University of Minho, DEI, Campus de Azurém, Guimarães 4800-058, Portugal
- CF-UM-UP,
Centro de Física das Universidades do Minho e Porto, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Jana B. Nieder
- INL—International
Iberian Nanotechnology Laboratory, Ultrafast
Bio- and Nanophotonics Group, Av. Mestre José Veiga S/n, 4715-330 Braga, Portugal
| |
Collapse
|
34
|
Mahdavi MR, Enderami SE. Electrospun silk nanofibers promoted the in vitro expansion potential of CD 133 + cells derived from umbilical cord blood. Gene 2022; 809:146005. [PMID: 34673210 DOI: 10.1016/j.gene.2021.146005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 01/14/2023]
Abstract
Stem cells from umbilical cord blood (UCB) are able to proliferate and differentiate into various somatic cell types. Thereby, they are considered as one of the attractive stem cell sources in tissue engineering and regenerative medicine. However, the limited number of hematopoietic CD 133+ stem cells in UCB restricted the clinical application of such stem cells. This study was aimed to expand CD 133+ stem cells derived from UCB on a 3D silk scaffold. UCB133+ stem cells were extracted using Magnetic cell sorting (MACS) and characterized by flow cytometry. Isolated cells were seeded on a fabricated electrospun silk scaffold and cultured for 7 days. The real-time PCR, cell counting, colony-forming assay, and MTT assay were performed to evaluate the expansion and homing of stem cells. The results showed a higher expression of CXCR4 gene, the number of cultured stem cells, and colony-forming units in the 3D silk scaffold group after 7 days when compared to the tissue culture plate. Moreover, higher viability and proliferation of stem cells were seen in cells cultured on silk scaffold. It seems electrospun silk scaffold could be used as a suitable substrate for UCB CD 133+ stem cell expansion.
Collapse
Affiliation(s)
- Mohammad Reza Mahdavi
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Seyed Ehsan Enderami
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
35
|
Huang Y, Jayathilaka PB, Islam MS, Tanaka CB, Silberstein MN, Kilian KA, Kruzic JJ. Structural aspects controlling the mechanical and biological properties of tough, double network hydrogels. Acta Biomater 2022; 138:301-312. [PMID: 34757233 DOI: 10.1016/j.actbio.2021.10.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 01/05/2023]
Abstract
Anticipating an increasing demand for hybrid double network (DN) hydrogels in biomedicine and biotechnology, this study evaluated the effects of each network on the mechanical and biological properties. Polyethylene glycol (PEG) (meth)acrylate hydrogels with varied monomer molecular weights and architectures (linear vs. 4-arm) were produced with and without an added ionically bonded alginate network and their mechanical properties were characterized using compression testing. The results showed that while some mechanical properties of PEG single network (SN) hydrogels decreased or changed negligibly with increasing molecular weight, the compressive modulus, strength, strain to failure, and toughness of DN hydrogels all significantly increased with increased PEG monomer molecular weight. At a fixed molecular weight (10 kDa), 4-arm PEG SN hydrogels exhibited better overall mechanical performance; however, this benefit was diminished for the corresponding DN hydrogels with comparable strength and toughness and lower strain to failure for the 4-arm case. Regardless of the PEG monomer structure, the alginate network made a relatively larger contribution to the overall DN mechanical properties when the covalent PEG network was looser with a larger mesh size (e.g., for larger monomer molecular weight and/or linear architecture) which presumably enabled more ionic crosslinking. Considering the biological performance, adipose derived stem cell cultures demonstrated monotonically increasing cell area and Yes-associated protein related mechanosensing with increasing amounts of alginate from 0 to 2 wt.%, demonstrating the possibility for using DN hydrogels in guiding musculoskeletal differentiation. These findings will be useful to design suitable hydrogels with controllable mechanical and biological properties for mechanically demanding applications. STATEMENT OF SIGNIFICANCE: Hydrogels are widely used in commercial applications, and recently developed hybrid double network hydrogels have enhanced strength and toughness that will enable further expansion into more mechanically demanding applications (e.g., medical implants, etc.). The significance of this work is that it uncovers some key principles regarding monomer molecular weight, architecture, and concentration for developing strong and tough hybrid double network hydrogels that would not be predicted from their single network counterparts or a linear combination of the two networks. Additionally, novel insight is given into the biological performance of hybrid double network hydrogels in the presence of adipose derived stem cell cultures which suggests new scope for using double network hydrogels in guiding musculoskeletal differentiation.
Collapse
Affiliation(s)
- Yuwan Huang
- School of Mechanical and Manufacturing Engineering, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia
| | - Pavithra B Jayathilaka
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia
| | - Md Shariful Islam
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia
| | - Carina B Tanaka
- School of Mechanical and Manufacturing Engineering, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia
| | - Meredith N Silberstein
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Kristopher A Kilian
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia; School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia
| | - Jamie J Kruzic
- School of Mechanical and Manufacturing Engineering, University of New South Wales (UNSW Sydney), Sydney NSW 2052, Australia.
| |
Collapse
|
36
|
Choi YJ, Cho DW, Lee H. Development of Silk Fibroin Scaffolds by Using Indirect 3D-Bioprinting Technology. MICROMACHINES 2021; 13:mi13010043. [PMID: 35056208 PMCID: PMC8779165 DOI: 10.3390/mi13010043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/30/2022]
Abstract
Due to the excellent biocompatibility of natural polymers, a variety of natural polymers have been widely used as biomaterials for manufacturing tissue engineered scaffolds. Despite the excellent biological activity of natural polymers, there have been obstacles in using them on their own to prepare 3D scaffolds with sufficient mechanical strength. Although multiple 3D-bioprinting technologies have recently emerged as effective manufacturing tools for scaffold preparation, scaffold preparation using only natural polymers with tunable mechanical properties is still difficult. Herein, we introduce novel scaffold fabrication methods using the natural polymer silk fibroin via indirect 3D-bioprinting technology. The developed silk fibroin scaffolds showed biocompatibility and tunable mechanical strength by changing the concentration of the silk fibroin. Furthermore, controlling the flexibility of the silk fibroin scaffolds was made possible by changing the solvent for the silk fibroin solution used to fabricate the scaffold. Consequently, silk fibroin scaffolds fabricated via our method can be considered for various applications in the bioengineering of either soft or musculoskeletal tissues.
Collapse
Affiliation(s)
- Yeong-Jin Choi
- Department of Advanced Biomaterials Research, Korea Institute of Materials Science (KIMS), 797, Changwon-daero, Seongsan-gu, Changwon-si 51508, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
- Correspondence: (D.-W.C.); (H.L.)
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), 1 Gangwondaehakgil, Chuncheon-si 24341, Korea
- Interdisciplinary Program in Biohealth-Machinery Convergence Engineering, Kangwon National University (KNU), 1 Gangwondaehakgil, Chuncheon-si 24341, Korea
- Correspondence: (D.-W.C.); (H.L.)
| |
Collapse
|
37
|
Elkhoury K, Morsink M, Sanchez-Gonzalez L, Kahn C, Tamayol A, Arab-Tehrany E. Biofabrication of natural hydrogels for cardiac, neural, and bone Tissue engineering Applications. Bioact Mater 2021; 6:3904-3923. [PMID: 33997485 PMCID: PMC8080408 DOI: 10.1016/j.bioactmat.2021.03.040] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/05/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022] Open
Abstract
Natural hydrogels are one of the most promising biomaterials for tissue engineering applications, due to their biocompatibility, biodegradability, and extracellular matrix mimicking ability. To surpass the limitations of conventional fabrication techniques and to recapitulate the complex architecture of native tissue structure, natural hydrogels are being constructed using novel biofabrication strategies, such as textile techniques and three-dimensional bioprinting. These innovative techniques play an enormous role in the development of advanced scaffolds for various tissue engineering applications. The progress, advantages, and shortcomings of the emerging biofabrication techniques are highlighted in this review. Additionally, the novel applications of biofabricated natural hydrogels in cardiac, neural, and bone tissue engineering are discussed as well.
Collapse
Affiliation(s)
| | - Margaretha Morsink
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7500AE, the Netherlands
| | | | - Cyril Kahn
- LIBio, Université de Lorraine, Nancy, F-54000, France
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT, 06030, USA
| | | |
Collapse
|
38
|
Sthijns MMJPE, van Blitterswijk CA, LaPointe VLS. Synthetic Materials that Affect the Extracellular Matrix via Cellular Metabolism and Responses to a Metabolic State. Front Bioeng Biotechnol 2021; 9:742132. [PMID: 34708025 PMCID: PMC8542861 DOI: 10.3389/fbioe.2021.742132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
In regenerative medicine and tissue engineering, many materials are developed to mimic the extracellular matrix (ECM). However, these ECM-mimicking materials do not yet completely recapitulate the diversity and complexity of biological tissue-specific ECM. In this review, an alternative strategy is proposed to generate ECM, namely synthesizing a material that functions as a drug delivery system, releasing molecules that target cellular metabolic pathways and thereby stimulate the local cells to create their own ECM. This is based on the fact that ECM synthesis, modification, composition, signaling, stiffness, and degradation are modulated by cellular metabolism. Metabolism can be targeted at different levels, ranging from modulating the availability of substrates or co-factors to regulating the activity of essential transcription factors. Depending on the drug of interest, its characteristics, mechanism of action, cellular target, and application, a different drug delivery system should be designed. Metabolic drugs modulating the ECM require cellular uptake for their function, therefore reversible linkers are recommended. Preferably the metabolic modulators are only released when needed, which will be upon a specific metabolic state, a change in ECM stiffness, or ECM remodeling. Therefore, reversible linkers that respond to an environmental stimulus could be incorporated. All in all, a novel strategy is suggested to develop a tissue-specific ECM by generating a synthetic material that releases metabolic molecules modulating the ECM. Various ways to modulate the ECM properties via the metabolism are reviewed and guidelines for the development of these materials are provided.
Collapse
Affiliation(s)
- Mireille M J P E Sthijns
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Department of Food Innovation and Health at the Centre of Healthy Eating and Food Innovation, Maastricht University, Maastricht, Netherlands
| | - Clemens A van Blitterswijk
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
39
|
Da LC, Huang YZ, Xie HQ, Zheng BH, Huang YC, Du SR. Membranous Extracellular Matrix-Based Scaffolds for Skin Wound Healing. Pharmaceutics 2021; 13:1796. [PMID: 34834211 PMCID: PMC8620109 DOI: 10.3390/pharmaceutics13111796] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 02/05/2023] Open
Abstract
Membranous extracellular matrix (ECM)-based scaffolds are one of the most promising biomaterials for skin wound healing, some of which, such as acellular dermal matrix, small intestinal submucosa, and amniotic membrane, have been clinically applied to treat chronic wounds with acceptable outcomes. Nevertheless, the wide clinical applications are always hindered by the poor mechanical properties, the uncontrollable degradation, and other factors after implantation. To highlight the feasible strategies to overcome the limitations, in this review, we first outline the current clinical use of traditional membranous ECM scaffolds for skin wound healing and briefly introduce the possible repair mechanisms; then, we discuss their potential limitations and further summarize recent advances in the scaffold modification and fabrication technologies that have been applied to engineer new ECM-based membranes. With the development of scaffold modification approaches, nanotechnology and material manufacturing techniques, various types of advanced ECM-based membranes have been reported in the literature. Importantly, they possess much better properties for skin wound healing, and would become promising candidates for future clinical translation.
Collapse
Affiliation(s)
- Lin-Cui Da
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; (L.-C.D.); (B.-H.Z.)
| | - Yi-Zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China;
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China;
| | - Bei-Hong Zheng
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; (L.-C.D.); (B.-H.Z.)
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Sheng-Rong Du
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; (L.-C.D.); (B.-H.Z.)
| |
Collapse
|
40
|
Veiga A, Silva IV, Duarte MM, Oliveira AL. Current Trends on Protein Driven Bioinks for 3D Printing. Pharmaceutics 2021; 13:1444. [PMID: 34575521 PMCID: PMC8471984 DOI: 10.3390/pharmaceutics13091444] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023] Open
Abstract
In the last decade, three-dimensional (3D) extrusion bioprinting has been on the top trend for innovative technologies in the field of biomedical engineering. In particular, protein-based bioinks such as collagen, gelatin, silk fibroin, elastic, fibrin and protein complexes based on decellularized extracellular matrix (dECM) are receiving increasing attention. This current interest is the result of protein's tunable properties, biocompatibility, environmentally friendly nature and possibility to provide cells with the adequate cues, mimicking the extracellular matrix's function. In this review we describe the most relevant stages of the development of a protein-driven bioink. The most popular formulations, molecular weights and extraction methods are covered. The different crosslinking methods used in protein bioinks, the formulation with other polymeric systems or molecules of interest as well as the bioprinting settings are herein highlighted. The cell embedding procedures, the in vitro, in vivo, in situ studies and final applications are also discussed. Finally, we approach the development and optimization of bioinks from a sequential perspective, discussing the relevance of each parameter during the pre-processing, processing, and post-processing stages of technological development. Through this approach the present review expects to provide, in a sequential manner, helpful methodological guidelines for the development of novel bioinks.
Collapse
Affiliation(s)
- Anabela Veiga
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4099-002 Porto, Portugal
| | - Inês V. Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Marta M. Duarte
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| | - Ana L. Oliveira
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, 4169-005 Porto, Portugal; (A.V.); (I.V.S.); (M.M.D.)
| |
Collapse
|
41
|
Da Silva K, Kumar P, van Vuuren SF, Pillay V, Choonara YE. Three-Dimensional Printability of an ECM-Based Gelatin Methacryloyl (GelMA) Biomaterial for Potential Neuroregeneration. ACS OMEGA 2021; 6:21368-21383. [PMID: 34471741 PMCID: PMC8388000 DOI: 10.1021/acsomega.1c01903] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/30/2021] [Indexed: 05/03/2023]
Abstract
The current study introduces two novel, smart polymer three-dimensional (3D)-printable interpenetrating polymer network (IPN) hydrogel biomaterials with favorable chemical, mechanical, and morphological properties for potential applications in traumatic brain injury (TBI) such as potentially assisting in the restoration of neurological function through closure of the wound deficit and neural tissue regeneration. Additionally, removal of injury matter to allow for the appropriate scaffold grafting may assist in providing a TBI treatment. Furthermore, due to the 3D printability of the IPN biomaterials, complex structures can be designed and fabricated to mimic the native shape and structure of the injury sight, which can potentially assist with neural tissue regeneration after TBI. In this study, a peptide-only approach was employed, wherein collagen and elastin in a blend with gelatin methacryloyl were prepared and crosslinked using either Irgacure or Irgacure and Genipin to form either a semi or full IPN hydrogel 3D-printable neuromimicking platform system, respectively. The scaffolds displayed favorable thermal stability and were amorphous in nature with high full width at half-maximum values. Furthermore, no alteration to the peptide secondary structure was noted using Fourier transform infrared spectroscopy. The IPN biomaterials have a stiffness of around 600 Pa and are suitable for softer tissue engineering applications-that is, the brain. Scanning electron micrographs indicated that the IPN biomaterials had a morphological structure with a significant resemblance to the native rat cortex. Both biomaterial scaffolds were shown to support the growth of PC12 cells over a 72 h period. Furthermore, the increased nuclear eccentricity and nuclear area were shown to support the postulation that the IPN biomaterials maintain the cells in a healthy state encouraging cellular mitosis and proliferation. The Genipin component of the full IPN was further shown to exhibit antimicrobial properties and this suggests that Genipin can prevent the growth of pathogens associated with postsurgical brain infections. In addition to these findings, the study presents an anomaly, wherein the full IPN is found to be more brittle than the semi IPN, a finding that is in contradiction with the literature. This research, therefore, contributes to the collection of potential biomaterials for TBI applications coupled with 3D printing and can assist in the progression of neural treatments toward patient-specific scaffolds through the development of custom scaffolds.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South
Africa
| | - Pradeep Kumar
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South
Africa
| | - Sandy F. van Vuuren
- Department
of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty
of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South
Africa
| | - Viness Pillay
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South
Africa
| | - Yahya E. Choonara
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South
Africa
- .
Phone: +27-11-717-2052. Fax: +27-11-642-4355, +27-86-553-4733
| |
Collapse
|
42
|
Rozhin P, Charitidis C, Marchesan S. Self-Assembling Peptides and Carbon Nanomaterials Join Forces for Innovative Biomedical Applications. Molecules 2021; 26:4084. [PMID: 34279424 PMCID: PMC8271590 DOI: 10.3390/molecules26134084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 02/07/2023] Open
Abstract
Self-assembling peptides and carbon nanomaterials have attracted great interest for their respective potential to bring innovation in the biomedical field. Combination of these two types of building blocks is not trivial in light of their very different physico-chemical properties, yet great progress has been made over the years at the interface between these two research areas. This concise review will analyze the latest developments at the forefront of research that combines self-assembling peptides with carbon nanostructures for biological use. Applications span from tissue regeneration, to biosensing and imaging, and bioelectronics.
Collapse
Affiliation(s)
- Petr Rozhin
- Chemical and Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy;
| | - Costas Charitidis
- School of Chemical Engineering, National Technical University of Athens, Iroon Polytechneiou 9, Zografou, 157 80 Athens, Greece;
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy;
- INSTM, Unit of Trieste, 34127 Trieste, Italy
| |
Collapse
|
43
|
Chirivì M, Maiullari F, Milan M, Presutti D, Cordiglieri C, Crosti M, Sarnicola ML, Soluri A, Volpi M, Święszkowski W, Prati D, Rizzi M, Costantini M, Seliktar D, Parisi C, Bearzi C, Rizzi R. Tumor Extracellular Matrix Stiffness Promptly Modulates the Phenotype and Gene Expression of Infiltrating T Lymphocytes. Int J Mol Sci 2021; 22:5862. [PMID: 34070750 PMCID: PMC8198248 DOI: 10.3390/ijms22115862] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system is a fine modulator of the tumor biology supporting or inhibiting its progression, growth, invasion and conveys the pharmacological treatment effect. Tumors, on their side, have developed escaping mechanisms from the immune system action ranging from the direct secretion of biochemical signals to an indirect reaction, in which the cellular actors of the tumor microenvironment (TME) collaborate to mechanically condition the extracellular matrix (ECM) making it inhospitable to immune cells. TME is composed of several cell lines besides cancer cells, including tumor-associated macrophages, cancer-associated fibroblasts, CD4+ and CD8+ lymphocytes, and innate immunity cells. These populations interface with each other to prepare a conservative response, capable of evading the defense mechanisms implemented by the host's immune system. The presence or absence, in particular, of cytotoxic CD8+ cells in the vicinity of the main tumor mass, is able to predict, respectively, the success or failure of drug therapy. Among various mechanisms of immunescaping, in this study, we characterized the modulation of the phenotypic profile of CD4+ and CD8+ cells in resting and activated states, in response to the mechanical pressure exerted by a three-dimensional in vitro system, able to recapitulate the rheological and stiffness properties of the tumor ECM.
Collapse
Affiliation(s)
- Maila Chirivì
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.so della Repubblica 79, 04100 Latina, Italy
| | - Fabio Maiullari
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marika Milan
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
| | - Dario Presutti
- Institute of Physical Chemistry Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warszawa, Poland; (D.P.); (M.C.)
| | - Chiara Cordiglieri
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Mariacristina Crosti
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Maria Lucia Sarnicola
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Andrea Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
- Unit of Molecular Neurosciences, University Campus Bio-Medico, 00128 Roma, Italy
| | - Marina Volpi
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02-507 Warsaw, Poland; (M.V.); (W.Ś.)
| | - Wojciech Święszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02-507 Warsaw, Poland; (M.V.); (W.Ś.)
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, IRCCS Granda Hospital Maggiore Policlinico Foundation, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Marta Rizzi
- Ufficio Programmazione e Grant Office, National Research Council of Italy (UPGO-CNR), Piazzale Aldo Moro 7, 00185 Rome, Italy;
| | - Marco Costantini
- Institute of Physical Chemistry Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warszawa, Poland; (D.P.); (M.C.)
| | - Dror Seliktar
- Department of Biomedical Engineering, Technion Institute, Haifa 32000, Israel;
| | - Chiara Parisi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Genetic and Biomedical Research, UOS of Milan, National Research Council (IRGB-CNR), Via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090 Milan, Italy
| |
Collapse
|
44
|
Sugioka Y, Nakamura J, Ohtsuki C, Sugawara-Narutaki A. Thixotropic Hydrogels Composed of Self-Assembled Nanofibers of Double-Hydrophobic Elastin-Like Block Polypeptides. Int J Mol Sci 2021; 22:4104. [PMID: 33921095 PMCID: PMC8071462 DOI: 10.3390/ijms22084104] [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: 03/30/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/29/2022] Open
Abstract
Physically crosslinked hydrogels with thixotropic properties attract considerable attention in the biomedical research field because their self-healing nature is useful in cell encapsulation, as injectable gels, and as bioinks for three-dimensional (3D) bioprinting. Here, we report the formation of thixotropic hydrogels containing nanofibers of double-hydrophobic elastin-like polypeptides (ELPs). The hydrogels are obtained with the double-hydrophobic ELPs at 0.5 wt%, the concentration of which is an order of magnitude lower than those for previously reported ELP hydrogels. Although the kinetics of hydrogel formation is slower for the double-hydrophobic ELP with a cell-binding sequence, the storage moduli G' of mature hydrogels are similar regardless of the presence of a cell-binding sequence. Reversible gel-sol transitions are demonstrated in step-strain rheological measurements. The degree of recovery of the storage modulus G' after the removal of high shear stress is improved by chemical crosslinking of nanofibers when intermolecular crosslinking is successful. This work would provide deeper insight into the structure-property relationships of the self-assembling polypeptides and a better design strategy for hydrogels with desired viscoelastic properties.
Collapse
Affiliation(s)
- Yusuke Sugioka
- Department of Materials Chemistry, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; (Y.S.); (J.N.); (C.O.)
| | - Jin Nakamura
- Department of Materials Chemistry, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; (Y.S.); (J.N.); (C.O.)
| | - Chikara Ohtsuki
- Department of Materials Chemistry, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; (Y.S.); (J.N.); (C.O.)
| | - Ayae Sugawara-Narutaki
- Department of Energy Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| |
Collapse
|
45
|
Wu Y, Fortunato GM, Okesola BO, Brocchetti FLPD, Suntornnond R, Connelly J, De Maria C, Rodriguez-Cabello JC, Vozzi G, Wang W, Mata A. An interfacial self-assembling bioink for the manufacturing of capillary-like structures with tuneable and anisotropic permeability. Biofabrication 2021; 13. [PMID: 33561850 DOI: 10.1088/1758-5090/abe4c3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Self-assembling bioinks offer the possibility to biofabricate with molecular precision, hierarchical control, and biofunctionality. For this to become a reality with widespread impact, it is essential to engineer these ink systems ensuring reproducibility and providing suitable standardization. We have reported a self-assembling bioink based on disorder-to-order transitions of an elastin-like recombinamer (ELR) to co-assemble with graphene oxide (GO). Here, we establish reproducible processes, optimize printing parameters for its use as a bioink, describe new advantages that the self-assembling bioink can provide, and demonstrate how to fabricate novel structures with physiological relevance. We fabricate capillary-like structures with resolutions down to ∼10µm in diameter and ∼2µm thick tube walls and use both experimental and finite element analysis to characterize the printing conditions, underlying interfacial diffusion-reaction mechanism of assembly, printing fidelity, and material porosity and permeability. We demonstrate the capacity to modulate the pore size and tune the permeability of the resulting structures with and without human umbilical vascular endothelial cells. Finally, the potential of the ELR-GO bioink to enable supramolecular fabrication of biomimetic structures was demonstrated by printing tubes exhibiting walls with progressively different structure and permeability.
Collapse
Affiliation(s)
- Yuanhao Wu
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Gabriele Maria Fortunato
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Babatunde O Okesola
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | | | - Ratima Suntornnond
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - John Connelly
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Carmelo De Maria
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | | | - Giovanni Vozzi
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Wen Wang
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom.,Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
46
|
Mu X, Agostinacchio F, Xiang N, Pei Y, Khan Y, Guo C, Cebe P, Motta A, Kaplan DL. Recent Advances in 3D Printing with Protein-Based Inks. Prog Polym Sci 2021; 115:101375. [PMID: 33776158 PMCID: PMC7996313 DOI: 10.1016/j.progpolymsci.2021.101375] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) printing is a transformative manufacturing strategy, allowing rapid prototyping, customization, and flexible manipulation of structure-property relationships. Proteins are particularly appealing to formulate inks for 3D printing as they serve as essential structural components of living systems, provide a support presence in and around cells and for tissue functions, and also provide the basis for many essential ex vivo secreted structures in nature. Protein-based inks are beneficial in vivo due to their mechanics, chemical and physical match to the specific tissue, and full degradability, while also to promoting implant-host integration and serving as an interface between technology and biology. Exploiting the biological, chemical, and physical features of protein-based inks can provide key opportunities to meet the needs of tissue engineering and regenerative medicine. Despite these benefits, protein-based inks impose nontrivial challenges to 3D printing such as concentration and rheological features and reconstitution of the structural hierarchy observed in nature that is a source of the robust mechanics and functions of these materials. This review introduces photo-crosslinking mechanisms and rheological principles that underpins a variety of 3D printing techniques. The review also highlights recent advances in the design, development, and biomedical utility of monolithic and composite inks from a range of proteins, including collagen, silk, fibrinogen, and others. One particular focus throughout the review is to introduce unique material characteristics of proteins, including amino acid sequences, molecular assembly, and secondary conformations, which are useful for designing printing inks and for controlling the printed structures. Future perspectives of 3D printing with protein-based inks are also provided to support the promising spectrum of biomedical research accessible to these materials.
Collapse
Affiliation(s)
- Xuan Mu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Francesca Agostinacchio
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Industrial Engineering, University of Trento, via Sommarive 9, Trento 38123, Italy
| | - Ning Xiang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Ying Pei
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Yousef Khan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Chengchen Guo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Peggy Cebe
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, via Sommarive 9, Trento 38123, Italy
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
47
|
Guo QR, Zhang LL, Liu JF, Li Z, Li JJ, Zhou WM, Wang H, Li JQ, Liu DY, Yu XY, Zhang JY. Multifunctional microfluidic chip for cancer diagnosis and treatment. Nanotheranostics 2021; 5:73-89. [PMID: 33391976 PMCID: PMC7738943 DOI: 10.7150/ntno.49614] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Microfluidic chip is not a chip in the traditional sense. It is technologies that control fluids at the micro level. As a burgeoning biochip, microfluidic chips integrate multiple disciplines, including physiology, pathology, cell biology, biophysics, engineering mechanics, mechanical design, materials science, and so on. The application of microfluidic chip has shown tremendous promise in the field of cancer therapy in the past three decades. Various types of cell and tissue cultures, including 2D cell culture, 3D cell culture and tissue organoid culture could be performed on microfluidic chips. Patient-derived cancer cells and tissues can be cultured on microfluidic chips in a visible, controllable, and high-throughput manner, which greatly advances the process of personalized medicine. Moreover, the functionality of microfluidic chip is greatly expanding due to the customizable nature. In this review, we introduce its application in developing cancer preclinical models, detecting cancer biomarkers, screening anti-cancer drugs, exploring tumor heterogeneity and producing nano-drugs. We highlight the functions and recent development of microfluidic chip to provide references for advancing cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao-ru Guo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ling-ling Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ji-fang Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R.China
| | - Jia-jun Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Wen-min Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jing-quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| | - Da-yu Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R.China
| | - Xi-yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jian-ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| |
Collapse
|
48
|
Zhang X, Liu Y, Clark KL, Padget AM, Alexander PG, Dai J, Zhu W, Lin H. Mesenchymal stem cell-derived extracellular matrix (mECM): a bioactive and versatile scaffold for musculoskeletal tissue engineering. ACTA ACUST UNITED AC 2020; 16:012002. [PMID: 32906098 DOI: 10.1088/1748-605x/abb6b3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cell-derived extracellular matrix (mECM) has received increased attention in the fields of tissue engineering and scaffold-assisted regeneration. mECM exhibits many unique characteristics, such as robust bioactivity, biocompatibility, ease of use, and the potential for autologous tissue engineering. As the use of mECM has increased in musculoskeletal tissue engineering, it should be noted that mECM generated from current methods has inherited insufficiencies, such as low mechanical properties and lack of internal architecture. In this review, we first summarize the development and use of mECM as a scaffold for musculoskeletal tissue regeneration and highlight our current progress on moving this technology toward clinical application. Then we review recent methods to improve the properties of mECM that will overcome current weaknesses. Lastly, we propose future studies that will pave the road for mECM application in regenerating tissues in humans.
Collapse
Affiliation(s)
- Xiurui Zhang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America. Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, People's Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Petersen EV, Chudakova DA, Skorova EY, Anikin V, Reshetov IV, Mynbaev OA. The Extracellular Matrix-Derived Biomarkers for Diagnosis, Prognosis, and Personalized Therapy of Malignant Tumors. Front Oncol 2020; 10:575569. [PMID: 33425730 PMCID: PMC7793707 DOI: 10.3389/fonc.2020.575569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
The tumor biomarkers already have proven clinical value and have become an integral part in cancer management and modern translational oncology. The tumor tissue microenvironment (TME), which includes extracellular matrix (ECM), signaling molecules, immune and stromal cells, and adjacent non-tumorous tissue, contributes to cancer pathogenesis. Thus, TME-derived biomarkers have many clinical applications. This review is predominately based on the most recent publications (manuscripts published in a last 5 years, or seminal publications published earlier) and fills a gap in the current literature on the cancer biomarkers derived from the TME, with particular attention given to the ECM and products of its processing and degradation, ECM-associated extracellular vesicles (EVs), biomechanical characteristics of ECM, and ECM-derived biomarkers predicting response to the immunotherapy. We discuss the clinical utility of the TME-incorporating three-dimensional in vitro and ex vivo cell culture models for personalized therapy. We conclude that ECM is a critical driver of malignancies and ECM-derived biomarkers should be included in diagnostics and prognostics panels of markers in the clinic.
Collapse
Affiliation(s)
- Elena V. Petersen
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daria A. Chudakova
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Ekaterina Yu. Skorova
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Anikin
- Harefield Hospital, The Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, United Kingdom
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Igor V. Reshetov
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Ospan A. Mynbaev
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|