1
|
Ramesh G, Dommeti VK, Kumar HR, Sadasivam G. Developing and analyzing a Nanocomposites coated material for hammertoe implants. J Orthop 2025; 62:182-190. [PMID: 40241854 PMCID: PMC11999487 DOI: 10.1016/j.jor.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 04/18/2025] Open
Abstract
Hammertoe implants, frequently used for arthroplasty or proximal interphalangeal joint arthrodesis, have serious drawbacks because of bacterial colonization and material corrosion, which can result in infections and other problems. This study creates a new nanocomposite covering to improve the corrosion resistance, antibacterial qualities, and biocompatibility of 316L stainless steel (SS). 316L SS was treated with a nanocomposite made of graphene oxide (GO), zirconia (ZrO₂), and hydroxyapatite (HAp). The coating's structural and functional characteristics were examined using Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and energy-dispersive X-ray spectroscopy (EDX). Standard assays were used to evaluate the coated material's antibacterial activity, and simulated bodily fluid (SBF) was used to assess corrosion resistance in vitro. Biocompatibility was verified using assays for cell survival. Apatite layer formation during SBF immersion indicated increased bioactivity and the results showed that the HAp-ZrO₂-GO nanocomposite coating greatly increased the corrosion resistance of 316L SS. The coating showed anti-inflammatory and potent antibacterial qualities, successfully preventing bacterial colonization. Additionally, cell survival tests verified the coated material's biocompatibility, indicating that it would be safe for use in biomedical applications. This work presents a scalable and reasonably priced process for creating bioactive nanocomposite coatings for medical implants. The HAp-ZrO₂-GO coating addresses important drawbacks of conventional implant materials by improving physico-chemical interactions and providing better performance. With significant ramifications for developing biomedical engineering and enhancing patient outcomes, these results demonstrate the potential of the HAp-ZrO₂-GO nanocomposite as a workable option for long-lasting, antimicrobial, and biocompatible bioimplant coatings.
Collapse
Affiliation(s)
- Gayathri Ramesh
- Biomaterials Laboratory, Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur Campus, Chengalpattu, TamilNadu - 603 203, India
| | - Vamsi Krishna Dommeti
- Department of Mechanical Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Potheri, Kattankulathur Campus, Chengalpattu, TamilNadu - 603 203, India
| | - Hari Raj Kumar
- Biomaterials Laboratory, Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur Campus, Chengalpattu, TamilNadu - 603 203, India
| | - Gnanavel Sadasivam
- Biomaterials Laboratory, Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur Campus, Chengalpattu, TamilNadu - 603 203, India
| |
Collapse
|
2
|
Kumar R, Igwegbe CA, Khandel SK. Nanotherapeutic and Nano-Bio Interface for Regeneration and Healing. Biomedicines 2024; 12:2927. [PMID: 39767834 PMCID: PMC11673698 DOI: 10.3390/biomedicines12122927] [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: 04/14/2024] [Revised: 05/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Wound and injury healing processes are intricate and multifaceted, involving a sequence of events from coagulation to scar tissue formation. Effective wound management is crucial for achieving favorable clinical outcomes. Understanding the cellular and molecular mechanisms underlying wound healing, inflammation, and regeneration is essential for developing innovative therapeutics. This review explored the interplay of cellular and molecular processes contributing to wound healing, focusing on inflammation, innervation, angiogenesis, and the role of cell surface adhesion molecules. Additionally, it delved into the significance of calcium signaling in skeletal muscle regeneration and its implications for regenerative medicine. Furthermore, the therapeutic targeting of cellular senescence for long-term wound healing was discussed. The integration of cutting-edge technologies, such as quantitative imaging and computational modeling, has revolutionized the current approach of wound healing dynamics. The review also highlighted the role of nanotechnology in tissue engineering and regenerative medicine, particularly in the development of nanomaterials and nano-bio tools for promoting wound regeneration. Moreover, emerging nano-bio interfaces facilitate the efficient transport of biomolecules crucial for regeneration. Overall, this review provided insights into the cellular and molecular mechanisms of wound healing and regeneration, emphasizing the significance of interdisciplinary approaches and innovative technologies in advancing regenerative therapies. Through harnessing the potential of nanoparticles, bio-mimetic matrices, and scaffolds, regenerative medicine offers promising avenues for restoring damaged tissues with unparalleled precision and efficacy. This pursuit marks a significant departure from traditional approaches, offering promising avenues for addressing longstanding challenges in cellular and tissue repair, thereby significantly contributing to the advancement of regenerative medicine.
Collapse
Affiliation(s)
- Rajiv Kumar
- Faculty of Science, University of Delhi, Delhi 110007, India
| | - Chinenye Adaobi Igwegbe
- Department of Chemical Engineering, Nnamdi Azikiwe University, Awka 420218, Nigeria;
- Department of Applied Bioeconomy, Wroclaw University of Environmental and Life Sciences, 51-630 Wroclaw, Poland
| | - Shri Krishna Khandel
- Clinical Diagnosis and Investigation (Rognidan), National Institute of Ayurveda, Jaipur 302002, India;
| |
Collapse
|
3
|
Mondal S, Hazra A, Paul P, Saha B, Roy S, Bhowmick P, Bhowmick M. Formulation and evaluation of n-acetyl cysteine loaded bi-polymeric physically crosslinked hydrogel with antibacterial and antioxidant activity for diabetic wound dressing. Int J Biol Macromol 2024; 279:135418. [PMID: 39245103 DOI: 10.1016/j.ijbiomac.2024.135418] [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: 07/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Diabetic wounds have become a serious global health concern, with a growing number of patients each year. Diabetic altered wound healing physiology, as well as resulting complications, make therapy difficult. Hence, diabetic wound healing necessitates a multidisciplinary strategy. This study focused on the formulation, statistical optimization, ex vivo, and in vitro evaluation of a diabetic wound healing by n-acetyl cysteine (NAC) loaded hydrogel. The objective of the study is to formulate n-acetyl loaded hydrogel with different ratio (1:1, 1:2, 1:3, 2:1) of sodium alginate and guar gum. The antibacterial and antifungal assessment against the viability of Pseudomonas aeruginosa (P. aeruginosa), Escherichia coli (E. coli), and Staphylococcus aureus (S.aureus) and Candida albicans (C. albicans) was conducted after determining the in vitro drug release profile. The results of the experiment demonstrated that the formulation F3 was an optimal formulation on triplicate measurement with a pH of 6.2 ± 0.168, and a density of 1.026 ± 0.21. In vitro cell line study exhibited F3 has potential role in cell adhesion and proliferation might be beneficial to tissue regeneration and wound healing. The results imply that F3 may be helpful for the quick healing of diabetic wounds by promoting angiogenesis and also by scavenging free oxygen radicals.
Collapse
Affiliation(s)
- Sourav Mondal
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Ahana Hazra
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Pankaj Paul
- Eminent College of Pharmaceutical Technology Barbaria, Moshpukur, Paschim Khilkapur, Barasat, Jagannathpur, West Bengal 700126, India
| | - Bishnu Saha
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Sanjita Roy
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Pratibha Bhowmick
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Mithun Bhowmick
- Bengal College of Pharmaceutical Sciences and Research, Bidhannagar, Durgapur 713212, West Bengal, India.
| |
Collapse
|
4
|
Mondal S, Hazra A, Paul P, Saha B, Roy S, Bhowmick P, Bhowmick M. Formulation and evaluation of n-acetyl cysteine loaded bi-polymeric physically crosslinked hydrogel with antibacterial and antioxidant activity for diabetic wound dressing. Int J Biol Macromol 2024; 279:135418. [DOI: https:/doi.org/10.1016/j.ijbiomac.2024.135418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
|
5
|
Liao G, Sun E, Kana EBG, Huang H, Sanusi IA, Qu P, Jin H, Liu J, Shuai L. Renewable hemicellulose-based materials for value-added applications. Carbohydr Polym 2024; 341:122351. [PMID: 38876719 DOI: 10.1016/j.carbpol.2024.122351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024]
Abstract
The importance of renewable resources and environmentally friendly materials has grown globally in recent time. Hemicellulose is renewable lignocellulosic materials that have been the subject of substantial valorisation research. Due to its distinctive benefits, including its wide availability, low cost, renewability, biodegradability, simplicity of chemical modification, etc., it has attracted increasing interest in a number of value-added fields. In this review, a systematic summarizes of the structure, extraction method, and characterization technique for hemicellulose-based materials was carried out. Also, their most current developments in a variety of value-added adsorbents, biomedical, energy-related, 3D-printed materials, sensors, food packaging applications were discussed. Additionally, the most recent challenges and prospects of hemicellulose-based materials are emphasized and examined in-depth. It is anticipated that in the near future, persistent scientific efforts will enable the renewable hemicellulose-based products to achieve practical applications.
Collapse
Affiliation(s)
- Guangfu Liao
- National Forestry and Grassland Administration Key Laboratory of Plant Fiber Functional Materials, College of Materials Engineering, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Enhui Sun
- Key Laboratory of Saline-Alkali Soil Improvement and Utilization (Coastal Saline-Alkali Lands), Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Institute of Agricultural Resources and Environment, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville 3209, South Africa; School of Environmental and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - E B Gueguim Kana
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville 3209, South Africa
| | - Hongying Huang
- Key Laboratory of Saline-Alkali Soil Improvement and Utilization (Coastal Saline-Alkali Lands), Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Institute of Agricultural Resources and Environment, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Isaac A Sanusi
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville 3209, South Africa
| | - Ping Qu
- Key Laboratory of Saline-Alkali Soil Improvement and Utilization (Coastal Saline-Alkali Lands), Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Institute of Agricultural Resources and Environment, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Hongmei Jin
- Key Laboratory of Saline-Alkali Soil Improvement and Utilization (Coastal Saline-Alkali Lands), Ministry of Agriculture and Rural Affairs, Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Institute of Agricultural Resources and Environment, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jun Liu
- School of Environmental and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Li Shuai
- National Forestry and Grassland Administration Key Laboratory of Plant Fiber Functional Materials, College of Materials Engineering, Fujian Agriculture and Forestry University, Fuzhou 350002, China..
| |
Collapse
|
6
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
7
|
Di Muzio L, Zara S, Cataldi A, Sergi C, Carriero VC, Bigi B, Carradori S, Tirillò J, Petralito S, Casadei MA, Paolicelli P. Impact of Composition and Autoclave Sterilization on the Mechanical and Biological Properties of ECM-Mimicking Cryogels. Polymers (Basel) 2024; 16:1939. [PMID: 39000793 PMCID: PMC11244042 DOI: 10.3390/polym16131939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
Cryogels represent a valid strategy as scaffolds for tissue engineering. In order to adequately support adhesion and proliferation of anchorage-dependent cells, different polymers need to be combined within the same scaffold trying to mimic the complex features of a natural extracellular matrix (ECM). For this reason, in this work, gelatin (Gel) and chondroitin sulfate (CS), both functionalized with methacrylic groups to produce CSMA and GelMA derivatives, were selected to prepare cryogel networks. Both homopolymer and heteropolymer cryogels were produced, via radical crosslinking reactions carried out at -12 °C for 2 h. All the scaffolds were characterized for their mechanical, swelling and morphological properties, before and after autoclave sterilization. Moreover, they were evaluated for their biocompatibility and ability to support the adhesion of human gingival fibroblasts and tenocytes. GelMA-based homopolymer networks better withstood the autoclave sterilization process, compared to CSMA cryogels. Indeed, GelMA cryogels showed a decrease in stiffness of approximately 30%, whereas CSMA cryogels of approximately 80%. When GelMA and CSMA were blended in the same network, an intermediate outcome was observed. However, the hybrid scaffolds showed a general worsening of the biological performance. Indeed, despite their ability to withstand autoclave sterilization with limited modification of the mechanical and morphological properties, the hybrid cryogels exhibited poor cell adhesion and high LDH leakage. Therefore, not only do network components need to be properly selected, but also their combination and ability to withstand effective sterilization process should be carefully evaluated for the development of efficient scaffolds designed for tissue engineering purposes.
Collapse
Affiliation(s)
- Laura Di Muzio
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Susi Zara
- Department of Pharmacy, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Claudia Sergi
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, 00184 Rome, Italy
| | - Vito Cosimo Carriero
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Barbara Bigi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Simone Carradori
- Department of Pharmacy, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Jacopo Tirillò
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, 00184 Rome, Italy
| | - Stefania Petralito
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Patrizia Paolicelli
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
8
|
Anaya-Sampayo LM, García-Robayo DA, Roa NS, Rodriguez-Lorenzo LM, Martínez-Cardozo C. Platelet-rich fibrin (PRF) modified nano-hydroxyapatite/chitosan/gelatin/alginate scaffolds increase adhesion and viability of human dental pulp stem cells (DPSC) and osteoblasts derived from DPSC. Int J Biol Macromol 2024; 273:133064. [PMID: 38866288 DOI: 10.1016/j.ijbiomac.2024.133064] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Bone tissue regeneration strategies have incorporated the use of natural polymers, such as hydroxyapatite (nHA), chitosan (CH), gelatin (GEL), or alginate (ALG). Additionally, platelet concentrates, such as platelet-rich fibrin (PRF) have been suggested to improve scaffold biocompatibility. This study aimed to develop scaffolds composed of nHA, GEL, and CH, with or without ALG and lyophilized PRF, to evaluate the scaffold's properties, growth factor release, and dental pulp stem cells (DPSC), and osteoblast (OB) derived from DPSC viability. Four scaffold variations were synthesized and lyophilized. Then, degradation, swelling profiles, and morphological analysis were performed. Furthermore, PDGF-BB and FGF-B growth factors release were quantified by ELISA, and cytotoxicity and cell viability were evaluated. The swelling and degradation profiles were similar in all scaffolds, with pore sizes ranging between 100 and 250 μm. FGF-B and PDGF-BB release was evidenced after 24 h of scaffold immersion in cell culture medium. DPSC and OB-DPSC viability was notably increased in PRF-supplemented scaffolds. The nHA-CH-GEL-PRF scaffold demonstrated optimal physical-biological characteristics for stimulating DPSC and OB-DPSC cell viability. These results suggest lyophilized PRF improves scaffold biocompatibility for bone tissue regeneration purposes.
Collapse
Affiliation(s)
| | | | - Nelly S Roa
- Dental Research Center, School of Dentistry, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis Maria Rodriguez-Lorenzo
- Department of Polymeric Nanomaterials and Biomaterials, Institute Science and Technology of Polymers (ICTP-CSIC), Madrid, Spain
| | | |
Collapse
|
9
|
Sudhakar MP, Ali S, Chitra S. Scrutinizing the effect of rGO-cuttlefish bone hydroxyapatite composite infused carrageenan membrane towards wound reconstruction. Int J Biol Macromol 2024; 262:130155. [PMID: 38365153 DOI: 10.1016/j.ijbiomac.2024.130155] [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: 11/21/2023] [Revised: 02/01/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Carrageenan is an emerging biopolymer for wound healing and regenerative applications. In this study, reduced graphene oxide (rGO) and hydroxyapatite (HAp) nano-composites infused carrageenan bioactive membrane was fabricated. Here, hydroxyapatite was synthesized from cuttlefish bone (CF-HAp) and its properties were compared with that of chemically synthesized HAp. Crystalline Ca5(PO4)3(OH) and Ca3(PO4)2) phases were obtained in cuttlefish bone derived HAp. Reduced graphene oxide was synthesized and composites were prepared with chemical HAp and CF-HAp. FT-IR spectral analysis showed the imprints of hydroxyapatite on the membrane and also nano-structured particles were evident through morphological estimations that confirm the distribution of nano-particles on the carrageenan membrane. Nano-particulates infused carrageenan membrane showed the maximum tensile strength, in which graphene incorporated carrageenan bioactive membrane showed highest stability of 15.26 MPa. The contact angle of chemical HAp infused carrageenan membrane (CAR-HAp) showed more hydrophilic in nature (48.63° ± 7.47°) compared to control (61.77° ± 1.28°). Bio-compatibility features enunciate the optimal compatibility of fabricated bioactive membrane with fibroblast cell line; simultaneously, CAR-rGO-CF-HAp showed tremendous wound healing behavior with zebrafish model. Hence, fabricated bioactive membrane with the infusion of rGO- hydroxyapatite derived from cuttlefish bone was found to be a versatile biopolymer membrane for wound healing application.
Collapse
Affiliation(s)
- M P Sudhakar
- Marine Biotechnology Division, National Institute of Ocean Technology, Ministry of Earth Sciences (Govt. of India), Pallikaranai, Chennai 600 100, Tamil Nadu, India
| | - Saheb Ali
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, Tamil Nadu, India
| | - S Chitra
- Department of Biomaterials (Prosthodontics), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai 600 077, Tamil Nadu, India.
| |
Collapse
|
10
|
Amiri Z, Molavi AM, Amani A, Moqadam KH, Vatanchian M, Hashemi SA, Oroojalian F. Fabrication, Characterization and Wound-Healing Properties of Core-Shell SF@chitosan/ZnO/ Astragalus Arbusculinus Gum Nanofibers. Nanomedicine (Lond) 2024; 19:499-518. [PMID: 38293919 DOI: 10.2217/nnm-2023-0311] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
AIM Silk fibroin/chitosan/ZnO/Astragalus arbusculinus (Ast) gum fibrous scaffolds along with adipose-derived mesenchymal stem cells (ADSCs) were investigated for accelerating diabetic wound healing. METHODS Scaffolds with a core-shell structure and different compositions were synthesized using the electrospinning method. Biological in vitro investigations included antibacterial testing, cell viability analysis and cell attachment evaluation. In vivo experiments, including the chicken chorioallantoic membrane (CAM) test, were conducted to assess wound-healing efficacy and histopathological changes. RESULTS The incorporation of Ast to the silk fibroin@ chitosan/ZnO scaffold improved wound healing in diabetic mice. In addition, seeding of ADSCs on the scaffold accelerated wound healing. CONCLUSION These findings suggest that the designed scaffold can be useful for skin regeneration applications.
Collapse
Affiliation(s)
- Zahra Amiri
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
| | - Amir Mahdi Molavi
- Department of Materials Research, Iranian Academic Center for Education, Culture & Research (ACECR), Khorasan Razavi Branch, Mashhad, 9177-948974, Iran
| | - Amir Amani
- Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
| | | | - Mehran Vatanchian
- Department of Anatomical Sciences School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
| | - Seyyed Ahmad Hashemi
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
- Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran
| |
Collapse
|
11
|
Zivari-Ghader T, Valioglu F, Eftekhari A, Aliyeva I, Beylerli O, Davran S, Cho WC, Beilerli A, Khalilov R, Javadov S. Recent progresses in natural based therapeutic materials for Alzheimer's disease. Heliyon 2024; 10:e26351. [PMID: 38434059 PMCID: PMC10906329 DOI: 10.1016/j.heliyon.2024.e26351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024] Open
Abstract
Alzheimer's disease is a neurological disorder that causes increased memory loss, mood swings, behavioral disorders, and disruptions in daily activities. Polymer scaffolds for the brain have been grown under laboratory, physiological, and pathological circumstances because of the limitations of conventional treatments for patients with central nervous system diseases. The blood-brain barrier prevents medications from entering the brain, challenging AD treatment. Numerous biomaterials such as biomolecules, polymers, inorganic metals, and metal oxide nanoparticles have been used to transport therapeutic medicines into the nervous system. Incorporating biocompatible materials that support neurogenesis through a combination of topographical, pharmacological, and mechanical stimuli has also shown promise for the transfer of cells to replenish dopaminergic neurons. Components made of naturally occurring biodegradable polymers are appropriate for the regeneration of nerve tissue. The ability of natural-based materials (biomaterials) has been shown to promote endogenous cell development after implantation. Also, strategic functionalization of polymeric nanocarriers could be employed for treating AD. In particular, nanoparticles could resolve Aβ aggregation and thus help cure Alzheimer's disease. Drug moieties can be effectively directed to the brain by utilizing nano-based systems and diverse colloidal carriers, including hydrogels and biodegradable scaffolds. Notably, early investigations employing neural stem cells have yielded promising results, further emphasizing the potential advancements in this field. Few studies have fully leveraged the combination of cells with cutting-edge biomaterials. This study provides a comprehensive overview of prior research, highlighting the pivotal role of biomaterials as sophisticated drug carriers. It delves into various intelligent drug delivery systems, encompassing pH and thermo-triggered mechanisms, polymeric and lipid carriers, inorganic nanoparticles, and other vectors. The discussion synthesizes existing knowledge and underscores the transformative impact of these biomaterials in devising innovative strategies, augmenting current therapeutic methodologies, and shaping new paradigms in the realm of Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Tayebeh Zivari-Ghader
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Ferzane Valioglu
- Technology Development Zones Management CO, Sakarya University, Sakarya, Turkey
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51665118, Iran
- Department of Biochemistry, Faculty of Science, Ege University, İzmir, Turkey
| | - Immi Aliyeva
- Department of Biophysics and Biochemistry, Baku State University, Baku, Azerbaijan
- Department of Environmental Engineering, Azerbaijan Technological University, Ganja, Azerbaijan
| | - Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Republic of Bashkortostan, 3 Lenin Street, Ufa, 450008, Russia
| | - Soodabeh Davran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
- Department of Life Sciences, Khazar University, Baku, Azerbaijan
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Rovshan Khalilov
- Department of Biophysics and Biochemistry, Baku State University, Baku, Azerbaijan
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA
| |
Collapse
|
12
|
Khan R, Aslam Khan MU, Stojanović GM, Javed A, Haider S, Abd Razak SI. Fabrication of Bilayer Nanofibrous-Hydrogel Scaffold from Bacterial Cellulose, PVA, and Gelatin as Advanced Dressing for Wound Healing and Soft Tissue Engineering. ACS OMEGA 2024; 9:6527-6536. [PMID: 38371763 PMCID: PMC10870282 DOI: 10.1021/acsomega.3c06613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 02/20/2024]
Abstract
Tissue engineering is currently one of the fastest-growing areas of engineering, requiring the fabrication of advanced and multifunctional materials that can be used as scaffolds or dressings for tissue regeneration. In this work, we report a bilayer material prepared by electrospinning a hybrid material of poly(vinyl alcohol) (PVA) and bacterial cellulose (BC NFs) (top layer) over a highly interconnected porous 3D gelatin-PVA hydrogel obtained by a freeze-drying process (bottom layer). The techniques were combined to produce an advanced material with synergistic effects on the physical and biological properties of the two materials. The bilayer material was characterized using Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and a water contact measurement system (WCMS). Studies on swelling, degradability, porosity, drug release, cellular and antibacterial activities were performed using standardized procedures and assays. FTIR confirmed cross-linking of both the top and bottom layers, and SEM showed porous structure for the bottom layer, random deposition of NFs on the surface, and aligned NFs in the cross section. The water contact angle (WCA) showed a hydrophilic surface for the bilayer material. Swelling analysis showed high swelling, and degradation analysis showed good stability. The bilayer material released Ag-sulfadiazine in a sustained and controlled manner and showed good antibacterial activities against severe disease-causing gram + ive and -ive (Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa) bacterial strains. In vitro biological studies were performed on fibroblasts (3T3) and human embryonic kidneys (HEK-293), which showed desirable cell viability, proliferation, and adhesion to the bilayer. Thus, the synergistic effect of NFs and the hydrogel resulted in a potential wound dressing material for wound healing and soft tissue engineering.
Collapse
Affiliation(s)
- Rawaiz Khan
- Faculty
of Chemical and Energy Engineering, Universiti
Teknologi Malaysia (UTM), UTM Skudai, Johor Bahru, Johor 81310, Malaysia
| | - Muhammad Umar Aslam Khan
- Biomedical
Research Center, Qatar University, Doha 2713, Qatar
- Department
of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
- BioInspired
Device and Tissue Engineering Research Group, School of Biomedical
Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia, Skudai, Johor 81300, Malaysia
| | - Goran M. Stojanović
- Department
of Electronics, Faculty of Technical Sciences, University of Novi Sad, Novi Sad 21000, Serbia
| | - Aneela Javed
- Department
of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Sajjad Haider
- Chemical
Engineering Department, College of Engineering, King Saud University, Riyadh 11421, Saudi Arabia
| | - Saiful Izwan Abd Razak
- BioInspired
Device and Tissue Engineering Research Group, School of Biomedical
Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia, Skudai, Johor 81300, Malaysia
- Sports
Innovation
& Technology Centre, Institute of Human Centred Engineering, Universiti Teknologi Malaysia, Skudai, Johor 81300, Malaysia
| |
Collapse
|
13
|
Zhao J, Zhou C, Xiao Y, Zhang K, Zhang Q, Xia L, Jiang B, Jiang C, Ming W, Zhang H, Long H, Liang W. Oxygen generating biomaterials at the forefront of regenerative medicine: advances in bone regeneration. Front Bioeng Biotechnol 2024; 12:1292171. [PMID: 38282892 PMCID: PMC10811251 DOI: 10.3389/fbioe.2024.1292171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Globally, an annual count of more than two million bone transplants is conducted, with conventional treatments, including metallic implants and bone grafts, exhibiting certain limitations. In recent years, there have been significant advancements in the field of bone regeneration. Oxygen tension regulates cellular behavior, which in turn affects tissue regeneration through metabolic programming. Biomaterials with oxygen release capabilities enhance therapeutic effectiveness and reduce tissue damage from hypoxia. However, precise control over oxygen release is a significant technical challenge, despite its potential to support cellular viability and differentiation. The matrices often used to repair large-size bone defects do not supply enough oxygen to the stem cells being used in the regeneration process. Hypoxia-induced necrosis primarily occurs in the central regions of large matrices due to inadequate provision of oxygen and nutrients by the surrounding vasculature of the host tissues. Oxygen generating biomaterials (OGBs) are becoming increasingly significant in enhancing our capacity to facilitate the bone regeneration, thereby addressing the challenges posed by hypoxia or inadequate vascularization. Herein, we discussed the key role of oxygen in bone regeneration, various oxygen source materials and their mechanism of oxygen release, the fabrication techniques employed for oxygen-releasing matrices, and novel emerging approaches for oxygen delivery that hold promise for their potential application in the field of bone regeneration.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Yang Xiao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Kunyan Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Qiang Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chanyi Jiang
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenyi Ming
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengjian Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
14
|
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
|
15
|
Abaszadeh F, Ashoub MH, Khajouie G, Amiri M. Nanotechnology development in surgical applications: recent trends and developments. Eur J Med Res 2023; 28:537. [PMID: 38001554 PMCID: PMC10668503 DOI: 10.1186/s40001-023-01429-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 10/03/2023] [Indexed: 11/26/2023] Open
Abstract
This paper gives a detailed analysis of nanotechnology's rising involvement in numerous surgical fields. We investigate the use of nanotechnology in orthopedic surgery, neurosurgery, plastic surgery, surgical oncology, heart surgery, vascular surgery, ophthalmic surgery, thoracic surgery, and minimally invasive surgery. The paper details how nanotechnology helps with arthroplasty, chondrogenesis, tissue regeneration, wound healing, and more. It also discusses the employment of nanomaterials in implant surfaces, bone grafting, and breast implants, among other things. The article also explores various nanotechnology uses, including stem cell-incorporated nano scaffolds, nano-surgery, hemostasis, nerve healing, nanorobots, and diagnostic applications. The ethical and safety implications of using nanotechnology in surgery are also addressed. The future possibilities of nanotechnology are investigated, pointing to a possible route for improved patient outcomes. The essay finishes with a comment on nanotechnology's transformational influence in surgical applications and its promise for future breakthroughs.
Collapse
Affiliation(s)
- Farzad Abaszadeh
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ghazal Khajouie
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Mahnaz Amiri
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
16
|
Chu WS, Park H, Moon S. Novel Fabrication of 3-D Cell Laden Micro-Patterned Porous Structure on Cell Growth and Proliferation by Layered Manufacturing. Bioengineering (Basel) 2023; 10:1092. [PMID: 37760194 PMCID: PMC10526113 DOI: 10.3390/bioengineering10091092] [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: 08/29/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
This study focuses on developing and characterizing a novel 3-dimensional cell-laden micro-patterned porous structure from a mechanical engineering perspective. Tissue engineering holds great promise for repairing damaged organs but faces challenges related to cell viability, biocompatibility, and mechanical strength. This research aims to overcome these limitations by utilizing gelatin methacrylate hydrogel as a scaffold material and employing a photolithography technique for precise patterned fabrication. The mechanical properties of the structure are of particular interest in this study. We evaluate its ability to withstand external forces through compression tests, which provide insights into its strength and stability. Additionally, structural integrity is assessed over time to determine its performance in in vitro and potential in vivo environments. We investigate cell viability and proliferation within the micro-patterned porous structure to evaluate the biological aspects. MTT assays and immunofluorescence staining are employed to analyze the metabolic activity and distribution pattern of cells, respectively. These assessments help us understand the effectiveness of the structure in supporting cell growth and tissue regeneration. The findings of this research contribute to the field of tissue engineering and provide valuable insights for mechanical engineers working on developing scaffolds and structures for regenerative medicine. By addressing challenges related to cell viability, biocompatibility, and mechanical strength, we move closer to realizing clinically viable tissue engineering solutions. The novel micro-patterned porous structure holds promise for applications in artificial organ development and lays the foundation for future advancements in large soft tissue construction.
Collapse
Affiliation(s)
- Won-Shik Chu
- Department of Mechanical Convergence Engineering, Gyeongsang National University, Changwon 51391, Gyeongsangnam-do, Republic of Korea; (W.-S.C.); (H.P.)
| | - Hyeongryool Park
- Department of Mechanical Convergence Engineering, Gyeongsang National University, Changwon 51391, Gyeongsangnam-do, Republic of Korea; (W.-S.C.); (H.P.)
| | - Sangjun Moon
- Department of Mechanical Convergence Engineering, Gyeongsang National University, Changwon 51391, Gyeongsangnam-do, Republic of Korea; (W.-S.C.); (H.P.)
- Cyberneticsimagingsystems Co., Ltd., Changwon 51391, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
17
|
Vasella M, Arnke K, Dranseikiene D, Guzzi E, Melega F, Reid G, Klein HJ, Schweizer R, Tibbitt MW, Kim BS. Methacrylated Gelatin as a Scaffold for Mechanically Isolated Stromal Vascular Fraction for Cutaneous Wound Repair. Int J Mol Sci 2023; 24:13944. [PMID: 37762247 PMCID: PMC10530931 DOI: 10.3390/ijms241813944] [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: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a highly interesting cell source for regenerative purposes, including wound healing, and a practical alternative to enzymatically isolated SVF. In the clinical context, SVF benefits from scaffolds that facilitate viability and other cellular properties. In the present work, the feasibility of methacrylated gelatin (GelMA), a stiffness-tunable, light-inducible hydrogel with high biocompatibility is investigated as a scaffold for SVF in an in vitro setting. Lipoaspirates from elective surgical procedures were collected and processed to mSVF and mixed with GelMA precursor solutions. Non-encapsulated mSVF served as a control. Viability was measured over 21 days. Secreted basic fibroblast growth factor (bFGF) levels were measured on days 1, 7 and 21 by ELISA. IHC was performed to detect VEGF-A, perilipin-2, and CD73 expression on days 7 and 21. The impact of GelMA-mSVF on human dermal fibroblasts was measured in a co-culture assay by the same viability assay. The viability of cultured GelMA-mSVF was significantly higher after 21 days (p < 0.01) when compared to mSVF alone. Also, GelMA-mSVF secreted stable levels of bFGF over 21 days. While VEGF-A was primarily expressed on day 21, perilipin-2 and CD73-positive cells were observed on days 7 and 21. Finally, GelMA-mSVF significantly improved fibroblast viability as compared with GelMA alone (p < 0.01). GelMA may be a promising scaffold for mSVF as it maintains cell viability and proliferation with the release of growth factors while facilitating adipogenic differentiation, stromal cell marker expression and fibroblast proliferation.
Collapse
Affiliation(s)
- Mauro Vasella
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Kevin Arnke
- Center for Preclinical Development, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Dalia Dranseikiene
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Elia Guzzi
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Francesca Melega
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Gregory Reid
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Holger Jan Klein
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland;
| | - Riccardo Schweizer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Regional Hospital Lugano, 6900 Lugano, Switzerland;
| | - Mark W. Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| |
Collapse
|
18
|
Hey G, Willman M, Patel A, Goutnik M, Willman J, Lucke-Wold B. Stem Cell Scaffolds for the Treatment of Spinal Cord Injury—A Review. BIOMECHANICS 2023; 3:322-342. [PMID: 37664542 PMCID: PMC10469078 DOI: 10.3390/biomechanics3030028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Spinal cord injury (SCI) is a profoundly debilitating yet common central nervous system condition resulting in significant morbidity and mortality rates. Major causes of SCI encompass traumatic incidences such as motor vehicle accidents, falls, and sports injuries. Present treatment strategies for SCI aim to improve and enhance neurologic functionality. The ability for neural stem cells (NSCs) to differentiate into diverse neural and glial cell precursors has stimulated the investigation of stem cell scaffolds as potential therapeutics for SCI. Various scaffolding modalities including composite materials, natural polymers, synthetic polymers, and hydrogels have been explored. However, most trials remain largely in the preclinical stage, emphasizing the need to further develop and refine these treatment strategies before clinical implementation. In this review, we delve into the physiological processes that underpin NSC differentiation, including substrates and signaling pathways required for axonal regrowth post-injury, and provide an overview of current and emerging stem cell scaffolding platforms for SCI.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Matthew Willman
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Aashay Patel
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Michael Goutnik
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Jonathan Willman
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
19
|
Soleimani M, Ebrahimi Z, Ebrahimi KS, Farhadian N, Shahlaei M, Cheraqpour K, Ghasemi H, Moradi S, Chang AY, Sharifi S, Baharnoori SM, Djalilian AR. Application of biomaterials and nanotechnology in corneal tissue engineering. J Int Med Res 2023; 51:3000605231190473. [PMID: 37523589 PMCID: PMC10392709 DOI: 10.1177/03000605231190473] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
Corneal diseases are among the most common causes of blindness worldwide. Regardless of the etiology, corneal opacity- or globe integrity-threatening conditions may necessitate corneal replacement procedures. Several procedure types are currently available to address these issues, based on the complexity and extent of injury. Corneal allograft or keratoplasty is considered to be first-line treatment in many cases. However, a significant proportion of the world's population are reported to have no access to this option due to limitations in donor preparation. Thus, providing an appropriate, safe, and efficient synthetic implant (e.g., artificial cornea) may revolutionize this field. Nanotechnology, with its potential applications, has garnered a lot of recent attention in this area, however, there is seemingly a long way to go. This narrative review provides a brief overview of the therapeutic interventions for corneal pathologies, followed by a summary of current biomaterials used in corneal regeneration and a discussion of the nanotechnologies that can aid in the production of superior implants.
Collapse
Affiliation(s)
- Mohammad Soleimani
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zohreh Ebrahimi
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Sadat Ebrahimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negin Farhadian
- Substance Abuse Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Ghasemi
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arthur Y Chang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sina Sharifi
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Seyed Mahbod Baharnoori
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Stoica Oprea AE, Bîrcă AC, Gherasim O, Ficai A, Grumezescu AM, Oprea OC, Vasile BȘ, Balta C, Andronescu E, Hermenean AO. Electrospun Fibrous Silica for Bone Tissue Engineering Applications. Pharmaceutics 2023; 15:1728. [PMID: 37376176 DOI: 10.3390/pharmaceutics15061728] [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: 04/08/2023] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The production of highly porous and three-dimensional (3D) scaffolds with biomimicking abilities has gained extensive attention in recent years for tissue engineering (TE) applications. Considering the attractive and versatile biomedical functionality of silica (SiO2) nanomaterials, we propose herein the development and validation of SiO2-based 3D scaffolds for TE. This is the first report on the development of fibrous silica architectures, using tetraethyl orthosilicate (TEOS) and polyvinyl alcohol (PVA) during the self-assembly electrospinning (ES) processing (a layer of flat fibers must first be created in self-assembly electrospinning before fiber stacks can develop on the fiber mat). The compositional and microstructural characteristics of obtained fibrous materials were evaluated by complementary techniques, in both the pre-ES aging period and post-ES calcination. Then, in vivo evaluation confirmed their possible use as bioactive scaffolds in bone TE.
Collapse
Affiliation(s)
- Alexandra Elena Stoica Oprea
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania
| | - Oana Gherasim
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
| | - Ovidiu-Cristian Oprea
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
| | - Bogdan Ștefan Vasile
- National Research Center for Micro and Nanomaterials, University Politehnica of Bucharest, 060042 Bucharest, Romania
- HTP Research and Consulting, Joita, 087150 Giurgiu, Romania
- Research Center for Advanced Materials, Products and Processes, University of Bucharest, 060042 Bucharest, Romania
| | - Cornel Balta
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310025 Arad, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
| | - Anca Oana Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310025 Arad, Romania
| |
Collapse
|
21
|
Sharma A, Kokil GR, He Y, Lowe B, Salam A, Altalhi TA, Ye Q, Kumeria T. Inorganic/organic combination: Inorganic particles/polymer composites for tissue engineering applications. Bioact Mater 2023; 24:535-550. [PMID: 36714332 PMCID: PMC9860401 DOI: 10.1016/j.bioactmat.2023.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Biomaterials have ushered the field of tissue engineering and regeneration into a new era with the development of advanced composites. Among these, the composites of inorganic materials with organic polymers present unique structural and biochemical properties equivalent to naturally occurring hybrid systems such as bones, and thus are highly desired. The last decade has witnessed a steady increase in research on such systems with the focus being on mimicking the peculiar properties of inorganic/organic combination composites in nature. In this review, we discuss the recent progress on the use of inorganic particle/polymer composites for tissue engineering and regenerative medicine. We have elaborated the advantages of inorganic particle/polymer composites over their organic particle-based composite counterparts. As the inorganic particles play a crucial role in defining the features and regenerative capacity of such composites, the review puts a special emphasis on the various types of inorganic particles used in inorganic particle/polymer composites. The inorganic particles that are covered in this review are categorised into two broad types (1) solid (e.g., calcium phosphate, hydroxyapatite, etc.) and (2) porous particles (e.g., mesoporous silica, porous silicon etc.), which are elaborated in detail with recent examples. The review also covers other new types of inorganic material (e.g., 2D inorganic materials, clays, etc.) based polymer composites for tissue engineering applications. Lastly, we provide our expert analysis and opinion of the field focusing on the limitations of the currently used inorganic/organic combination composites and the immense potential of new generation of composites that are in development.
Collapse
Affiliation(s)
- Astha Sharma
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Ganesh R. Kokil
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- School of Pharmacy, University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China
| | - Baboucarr Lowe
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Arwa Salam
- Chemistry Department, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Tariq A. Altalhi
- Chemistry Department, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
- School of Pharmacy, University of Queensland, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
22
|
Heydari P, Varshosaz J, Kharaziha M, Javanmard SH. Antibacterial and pH-sensitive methacrylate poly-L-Arginine/poly (β-amino ester) polymer for soft tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:16. [PMID: 37036618 PMCID: PMC10085925 DOI: 10.1007/s10856-023-06720-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/14/2023] [Indexed: 05/03/2023]
Abstract
During the last decade, pH-sensitive biomaterials containing antibacterial agents have grown exponentially in soft tissue engineering. The aim of this study is to synthesize a biodegradable pH sensitive and antibacterial hydrogel with adjustable mechanical and physical properties for soft tissue engineering. This biodegradable copolymer hydrogel was made of Poly-L-Arginine methacrylate (Poly-L-ArgMA) and different poly (β- amino ester) (PβAE) polymers. PβAE was prepared with four different diacrylate/diamine monomers including; 1.1:1 (PβAE1), 1.5:1 (PβAE1.5), 2:1 (PβAE2), and 3:1 (PβAE3), which was UV cross-linked using dimethoxy phenyl-acetophenone agent. These PβAE were then used for preparation of Poly-L-ArgMA/PβAE polymers and revealed a tunable swelling ratio, depending on the pH conditions. Noticeably, the swelling ratio increased by 1.5 times when the pH decreased from 7.4 to 5.6 in the Poly-L-ArgMA/PβAE1.5 sample. Also, the controllable degradation rate and different mechanical properties were obtained, depending on the PβAE monomer ratio. Noticeably, the tensile strength of the PβAE hydrogel increased from 0.10 ± 0.04 MPa to 2.42 ± 0.3 MPa, when the acrylate/diamine monomer molar ratio increased from 1.1:1 to 3:1. In addition, Poly-L-ArgMA/PβAE samples significantly improved L929 cell viability, attachment and proliferation. Poly-L-ArgMA also enhanced the antibacterial activities of PβAE against both Escherichia coli (~5.1 times) and Staphylococcus aureus (~2.7 times). In summary, the antibacterial and pH-sensitive Poly-L-ArgMA/PβAE1.5 with suitable mechanical, degradation and biological properties could be an appropriate candidate for soft tissue engineering, specifically wound healing applications.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
- Applied Physiology Research Center, Isfahan, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran.
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Isfahan, Iran
- Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
23
|
Chitin whiskers enhanced methacrylated hydroxybutyl chitosan hydrogels as anti-deformation scaffold for 3D cell culture. Carbohydr Polym 2023; 304:120483. [PMID: 36641181 DOI: 10.1016/j.carbpol.2022.120483] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/09/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Hydrogel, as three-dimensional (3D) cell culture scaffold, is an effective strategy for tissue and organ regeneration due to their good biocompatibility, biodegradability and resemblance to body microenvironments in vivo. However, the inherent weak mechanical properties and strong shrinkage of hydrogels during cell culture hinder its application in clinical. In this study, a two-component thermo/photo dual-sensitive hydrogel (M/C) was prepared from methacrylated hydroxybutyl chitosan (MHBC) and chitin whisker (CHW) via physical and chemical cross-linking methods. M/C hydrogel showed a special internal structure with lamellar arrangement. The rheological properties of the hydrogels could be regulated with the change of M/C ratio. It is worth emphasizing that the mechanical properties, shrinkage resistance and cellular capacitances of the M/C hydrogel were improved with the addition of CHW. Moreover, the M/C hydrogel not only exhibited excellent degradability and antibacterial properties, but also significantly promoted the adhesion and proliferation of MC3T3-E1 cells in vitro. Therefore, the M/C hydrogel showed a wide application potential in tissue regeneration as a 3D cell culture scaffold.
Collapse
|
24
|
Perrelle JM, Boreland AJ, Gamboa JM, Gowda P, Murthy NS. Biomimetic Strategies for Peripheral Nerve Injury Repair: An Exploration of Microarchitecture and Cellularization. BIOMEDICAL MATERIALS & DEVICES (NEW YORK, N.Y.) 2023; 1:21-37. [PMID: 38343513 PMCID: PMC10857769 DOI: 10.1007/s44174-022-00039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/14/2022] [Indexed: 02/15/2024]
Abstract
Injuries to the nervous system present formidable challenges to scientists, clinicians, and patients. While regeneration within the central nervous system is minimal, peripheral nerves can regenerate, albeit with limitations. The regenerative mechanisms of the peripheral nervous system thus provide fertile ground for clinical and scientific advancement, and opportunities to learn fundamental lessons regarding nerve behavior in the context of regeneration, particularly the relationship of axons to their support cells and the extracellular matrix environment. However, few current interventions adequately address peripheral nerve injuries. This article aims to elucidate areas in which progress might be made toward developing better interventions, particularly using synthetic nerve grafts. The article first provides a thorough review of peripheral nerve anatomy, physiology, and the regenerative mechanisms that occur in response to injury. This is followed by a discussion of currently available interventions for peripheral nerve injuries. Promising biomaterial fabrication techniques which aim to recapitulate nerve architecture, along with approaches to enhancing these biomaterial scaffolds with growth factors and cellular components, are then described. The final section elucidates specific considerations when developing nerve grafts, including utilizing induced pluripotent stem cells, Schwann cells, nerve growth factors, and multilayered structures that mimic the architectures of the natural nerve.
Collapse
Affiliation(s)
- Jeremy M. Perrelle
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Andrew J. Boreland
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Graduate Program in Molecular Biosciences, Rutgers University, Piscataway, NJ, USA
| | - Jasmine M. Gamboa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Prarthana Gowda
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - N. Sanjeeva Murthy
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
25
|
Zamanifard M, Khorasani MT, Daliri M. Hybrid electrospun polyhydroxybutyrate/gelatin/laminin/polyaniline scaffold for nerve tissue engineering application: Preparation, characterization, and in vitro assay. Int J Biol Macromol 2023; 235:123738. [PMID: 36805505 DOI: 10.1016/j.ijbiomac.2023.123738] [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: 11/07/2022] [Revised: 12/26/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023]
Abstract
Despite the widespread central nervous system injuries, treatment of these disorders is still an issue of concern due to the complexities. Natural recovery in these patients is rarely observed, which calls for developing new methods that address these problems. In this study, natural polymers of polyhydroxybutyrate (PHB) and gelatin were electrospun into scaffolds and cross-linked. In order to modify the PHB-based scaffold for nerve tissue engineering, the scaffold surface was modified by exposure to the ammonium gas plasma under controlled conditions, and the laminin as a promoter for neural cells was coated on the sample surface. Then, polyaniline nanoparticles were inkjet-printed on a sample surface as parallel lines to induce the differentiation of stem cells into neural cells. Infrared spectroscopy, absorption of PBS, AFM, degradation rate, contact angle, electron microscopy and optical microscopy, thermal and mechanical behavior, and analysis of the viability of L929 cells were investigated for the scaffolds. The results showed gelatin decreased the contact angle from 106.2° to 38° and increased the residual weight after PBS incubation from 82 % to 38 %. The moduli of the scaffold increased from 8.78 MPa for pure PHB to 28.74 for the modified scaffold. In addition, performed methods increased cell viability from 69 % for PHB to 89 % for modified scaffold and also had a favorable effect on cell adhesion. Investigation of culturing P19 stem cells demonstrated that they successfully differentiated into neural cells. Results show that the scaffolds prepared in this study were promising for nerve tissue engineering.
Collapse
Affiliation(s)
- Mohammad Zamanifard
- Department of Biomaterials, Faculty of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Taghi Khorasani
- Biomaterials Department of Iran Polymer and Petrochemical Institute, P.O. BOX 14965/159, Tehran, Iran.
| | - Morteza Daliri
- Department of Animal and Marine Biotechnology, National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh Km 15, Tehran-Karaj Highway, Tehran, Iran
| |
Collapse
|
26
|
Khodaverdi E, Hadizadeh F, Hoseini N, Eisvand F, Tayebi M, Kamali H, Oroojalian F. In-vitro and in-vivo evaluation of sustained-release buprenorphine using in-situ forming lipid-liquid crystal gels. Life Sci 2023; 314:121324. [PMID: 36574944 DOI: 10.1016/j.lfs.2022.121324] [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: 08/05/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
AIMS Sustained-release systems reduce the incidence of drug side effects and the need for frequent drug consumption, thus increasing patient compliance with treatment. In this study, we aimed to produce sustained-release buprenorphine (BP) using lipid-liquid crystal gels. MAIN METHODS The three experimental groups in this study included: group I: lipid-liquid crystal formulation 5 (F5) containing BP, group II: BP-free F5, group III: BP solution in NMP, and group IV: control (no treatment). The formulations were injected subcutaneously into the rabbits' back neck. KEY FINDINGS The results showed that the time required to reach the drug's maximum concentration (Tmax) was longer in group I than in group III. The maximum BP concentration (Cmax) and the constants of the drug removal rate and drug absorption rate (Ka) were significantly higher in group III compared to group I. The half-life (t1/2) of the drug in blood circulation was significantly longer in group I than in group III. Histopathological analysis revealed no histological abnormalities in the skin and heart in group I (BP-containing F5); however, mild hyperemia was observed in interstitial vessels in group III (BP-containing NMP). The kidney and liver tissues showed normal structure in the control group, as well as groups I and II. However, in the group receiving BP-containing NMP, significant congestion, tissue damage, necrosis, and fibrosis were observed in the kidney and liver. SIGNIFICANCE The results showed that the lipid-liquid crystal system can be used to design slow-release platforms for BP, minimizing the side effects associated with the use of its conventional forms.
Collapse
Affiliation(s)
- Elham Khodaverdi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nilofarsadat Hoseini
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Eisvand
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohadeseh Tayebi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
27
|
Klabukov I, Balyasin M, Krasilnikova O, Tenchurin T, Titov A, Krasheninnikov M, Mudryak D, Sulina Y, Shepelev A, Chvalun S, Dyuzheva T, Yakimova A, Sosin D, Lyundup A, Baranovskii D, Shegay P, Kaprin A. Angiogenic Modification of Microfibrous Polycaprolactone by pCMV-VEGF165 Plasmid Promotes Local Vascular Growth after Implantation in Rats. Int J Mol Sci 2023; 24:1399. [PMID: 36674913 PMCID: PMC9865169 DOI: 10.3390/ijms24021399] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Insufficient vascular growth in the area of artificial-material implantation contributes to ischemia, fibrosis, the development of bacterial infections, and tissue necrosis around the graft. The purpose of this study was to evaluate angiogenesis after implantation of polycaprolactone microfiber scaffolds modified by a pCMV-VEGF165-plasmid in rats. Influence of vascularization on scaffold degradation was also examined. We investigated flat microfibrous scaffolds obtained by electrospinning polycaprolactone with incorporation of the pCMV-VEGF-165 plasmid into the microfibers at concentrations of 0.005 ng of plasmid per 1 mg of polycaprolactone (0.005 ng/mg) (LCGroup) and 0.05 ng/mg (HCGroup). The samples were subcutaneously implanted in the interscapular area of rats. On days 7, 16, 33, 46, and 64, the scaffolds were removed, and a histological study with a morphometric evaluation of the density and diameter of the vessels and microfiber diameter was performed. The number of vessels was increased in all groups, as well as the resorption of the scaffold. On day 33, the vascular density in the HCGroup was 42% higher compared to the control group (p = 0.0344). The dose-dependent effect of the pCMV-VEGF165-plasmid was confirmed by enhanced angiogenesis in the HCGroup compared to the LCGroup on day 33 (p-value = 0.0259). We did not find a statistically significant correlation between scaffold degradation rate and vessel growth (the Pearson correlation coefficient was ρ = 0.20, p-value = 0.6134). Functionalization of polycaprolactone by incorporation of the pCMV-VEGF165 plasmid provided improved vascularization within 33 days after implantation, however, vessel growth did not seem to correlate with scaffold degradation rate.
Collapse
Affiliation(s)
- Ilya Klabukov
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249031 Obninsk, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, 115409 Obninsk, Russia
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Maksim Balyasin
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Olga Krasilnikova
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249031 Obninsk, Russia
| | - Timur Tenchurin
- National Research Centre “Kurchatov Institute”, 1, Akademika Kurchatova pl., 123182 Moscow, Russia
| | - Alexander Titov
- City Clinical Hospital No. 67 of Moscow Health Department, 2/44, Salyama Adilya St., 123423 Moscow, Russia
| | - Mikhail Krasheninnikov
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- Lomonosov Institute of Fine Chemical Technologies, Russian Technological University MIREA, 119454 Moscow, Russia
| | - Daniil Mudryak
- City Clinical Hospital No. 67 of Moscow Health Department, 2/44, Salyama Adilya St., 123423 Moscow, Russia
- Department of Hospital Surgery, Sklifosovsky Institute of Clinical Medicine, Sechenov University, 119435 Moscow, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov University, 119435 Moscow, Russia
| | - Alexey Shepelev
- National Research Centre “Kurchatov Institute”, 1, Akademika Kurchatova pl., 123182 Moscow, Russia
| | - Sergei Chvalun
- National Research Centre “Kurchatov Institute”, 1, Akademika Kurchatova pl., 123182 Moscow, Russia
| | - Tatiana Dyuzheva
- Department of Hospital Surgery, Sklifosovsky Institute of Clinical Medicine, Sechenov University, 119435 Moscow, Russia
| | - Anna Yakimova
- A. Tsyb Medical Research Radiological Center—Branch of the National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Dmitry Sosin
- Center for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, 125371 Moscow, Russia
| | - Alexey Lyundup
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Denis Baranovskii
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- A. Tsyb Medical Research Radiological Center—Branch of the National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Peter Shegay
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249031 Obninsk, Russia
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Andrey Kaprin
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249031 Obninsk, Russia
- Department of Urology and Operative Nephrology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
28
|
Chen X, Zhu L, Wang X, Xiao J. Insight into Heart-Tailored Architectures of Hydrogel to Restore Cardiac Functions after Myocardial Infarction. Mol Pharm 2023; 20:57-81. [PMID: 36413809 DOI: 10.1021/acs.molpharmaceut.2c00650] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With permanent heart muscle injury or death, myocardial infarction (MI) is complicated by inflammatory, proliferation and remodeling phases from both the early ischemic period and subsequent infarct expansion. Though in situ re-establishment of blood flow to the infarct zone and delays of the ventricular remodeling process are current treatment options of MI, they fail to address massive loss of viable cardiomyocytes while transplanting stem cells to regenerate heart is hindered by their poor retention in the infarct bed. Equipped with heart-specific mimicry and extracellular matrix (ECM)-like functionality on the network structure, hydrogels leveraging tissue-matching biomechanics and biocompatibility can mechanically constrain the infarct and act as localized transport of bioactive ingredients to refresh the dysfunctional heart under the constant cyclic stress. Given diverse characteristics of hydrogel including conductivity, anisotropy, adhesiveness, biodegradability, self-healing and mechanical properties driving local cardiac repair, we aim to investigate and conclude the dynamic balance between ordered architectures of hydrogels and the post-MI pathological milieu. Additionally, our review summarizes advantages of heart-tailored architectures of hydrogels in cardiac repair following MI. Finally, we propose challenges and prospects in clinical translation of hydrogels to draw theoretical guidance on cardiac repair and regeneration after MI.
Collapse
Affiliation(s)
- Xuerui Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Liyun Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xu Wang
- Hangzhou Medical College, Binjiang Higher Education Park, Binwen Road 481, Hangzhou 310053, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
29
|
Augustine R, Gezek M, Seray Bostanci N, Nguyen A, Camci-Unal G. Oxygen-Generating Scaffolds: One Step Closer to the Clinical Translation of Tissue Engineered Products. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 455:140783. [PMID: 36644784 PMCID: PMC9835968 DOI: 10.1016/j.cej.2022.140783] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The lack of oxygen supply in engineered constructs has been an ongoing challenge for tissue engineering and regenerative medicine. Upon implantation of an engineered tissue, spontaneous blood vessel formation does not happen rapidly, therefore, there is typically a limited availability of oxygen in engineered biomaterials. Providing oxygen in large tissue-engineered constructs is a major challenge that hinders the development of clinically relevant engineered tissues. Similarly, maintaining adequate oxygen levels in cell-laden tissue engineered products during transportation and storage is another hurdle. There is an unmet demand for functional scaffolds that could actively produce and deliver oxygen, attainable by incorporating oxygen-generating materials. Recent approaches include encapsulation of oxygen-generating agents such as solid peroxides, liquid peroxides, and fluorinated substances in the scaffolds. Recent approaches to mitigate the adverse effects, as well as achieving a sustained and controlled release of oxygen, are discussed. Importance of oxygen-generating materials in various tissue engineering approaches such as ex vivo tissue engineering, in situ tissue engineering, and bioprinting are highlighted in detail. In addition, the existing challenges, possible solutions, and future strategies that aim to design clinically relevant multifunctional oxygen-generating biomaterials are provided in this review paper.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Angelina Nguyen
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
30
|
Kazemi Asl S, Rahimzadegan M, Ostadrahimi R. The recent advancement in the chitosan hybrid-based scaffolds for cardiac regeneration after myocardial infarction. Carbohydr Polym 2023; 300:120266. [DOI: 10.1016/j.carbpol.2022.120266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/08/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022]
|
31
|
McCorry MC, Reardon KF, Black M, Williams C, Babakhanova G, Halpern JM, Sarkar S, Swami NS, Mirica KA, Boermeester S, Underhill A. Sensor technologies for quality control in engineered tissue manufacturing. Biofabrication 2022; 15:10.1088/1758-5090/ac94a1. [PMID: 36150372 PMCID: PMC10283157 DOI: 10.1088/1758-5090/ac94a1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022]
Abstract
The use of engineered cells, tissues, and organs has the opportunity to change the way injuries and diseases are treated. Commercialization of these groundbreaking technologies has been limited in part by the complex and costly nature of their manufacture. Process-related variability and even small changes in the manufacturing process of a living product will impact its quality. Without real-time integrated detection, the magnitude and mechanism of that impact are largely unknown. Real-time and non-destructive sensor technologies are key for in-process insight and ensuring a consistent product throughout commercial scale-up and/or scale-out. The application of a measurement technology into a manufacturing process requires cell and tissue developers to understand the best way to apply a sensor to their process, and for sensor manufacturers to understand the design requirements and end-user needs. Furthermore, sensors to monitor component cells' health and phenotype need to be compatible with novel integrated and automated manufacturing equipment. This review summarizes commercially relevant sensor technologies that can detect meaningful quality attributes during the manufacturing of regenerative medicine products, the gaps within each technology, and sensor considerations for manufacturing.
Collapse
Affiliation(s)
- Mary Clare McCorry
- Advanced Regenerative Manufacturing Institute, Manchester, NH 03101, United States of America
| | - Kenneth F Reardon
- Chemical and Biological Engineering and Biomedical Engineering, Colorado State University, Fort Collins, CO 80521, United States of America
| | - Marcie Black
- Advanced Silicon Group, Lowell, MA 01854, United States of America
| | - Chrysanthi Williams
- Access Biomedical Solutions, Trinity, Florida 34655, United States of America
| | - Greta Babakhanova
- National Institute of Standards and Technology, Gaithersburg, MD 20899, United States of America
| | - Jeffrey M Halpern
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824, United States of America
- Materials Science and Engineering Program, University of New Hampshire, Durham, NH 03824, United States of America
| | - Sumona Sarkar
- National Institute of Standards and Technology, Gaithersburg, MD 20899, United States of America
| | - Nathan S Swami
- Electrical and Computer Engineering, University of Virginia, Charlottesville, VA 22904, United States of America
| | - Katherine A Mirica
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, United States of America
| | - Sarah Boermeester
- Advanced Regenerative Manufacturing Institute, Manchester, NH 03101, United States of America
| | - Abbie Underhill
- Scientific Bioprocessing Inc., Pittsburgh, PA 15238, United States of America
| |
Collapse
|
32
|
Sariogullari H, Aroguz AZ, Adiguzel Z. Fabrication of a Patterned Scaffold Using Soft Lithography Technique to be Used in Cell Growth Applications. Mol Biotechnol 2022; 65:786-793. [PMID: 36214977 DOI: 10.1007/s12033-022-00581-2] [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: 08/20/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022]
Abstract
In recent years, within tissue engineering, cell growth on patterned surfaces have gained significant attention. Growing cells in patterns is important to manufacture polymeric tissues that can be used within the medical field. For this reason, the main focus of this study was to prepare patterned scaffolds using Titanium (Ti) and polyvinyl chloride (PVC) covered on microscope lamellas and examine their liability for cell growth. A polydimethylsiloxane stamp was initially prepared which was then used to transfer a predefined pattern onto PVC- and Ti-covered surfaces. Cell growth experiments were performed on the prepared materials by seeding L929 mouse fibroblasts. The growth of cells seeded on the surface of the scaffolds were spectroscopically followed using Neutral Red uptake assay. The results showed cell proliferation on both patterned surfaces, however, it was higher on Ti-covered samples. In addition, three different alkanethiols were tested for cell adhesion on patterned surfaces. A higher number of cell proliferation was observed with undecanethiol, which has a shorter alkane group among them. The morphological properties of the samples before and after cell-seeding were analyzed via scanning electron microscope and optical microscopy. Significant amount of cell proliferation was observed on all of the prepared samples.
Collapse
Affiliation(s)
- Hidayet Sariogullari
- Department of Chemistry, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Ayse Z Aroguz
- Department of Chemistry, Engineering Faculty, Istanbul University-Cerrahpasa, Avcilar, 34320, Istanbul, Turkey.
| | - Zelal Adiguzel
- Basic Medical Sciences, Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| |
Collapse
|
33
|
Alamdari SG, Alibakhshi A, de la Guardia M, Baradaran B, Mohammadzadeh R, Amini M, Kesharwani P, Mokhtarzadeh A, Oroojalian F, Sahebkar A. Conductive and Semiconductive Nanocomposite-Based Hydrogels for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200526. [PMID: 35822350 DOI: 10.1002/adhm.202200526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/26/2022] [Indexed: 01/27/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide and the most common cause is myocardial infarction. Therefore, appropriate approaches should be used to repair damaged heart tissue. Recently, cardiac tissue engineering approaches have been extensively studied. Since the creation of the nature of cardiovascular tissue engineering, many advances have been made in cellular and scaffolding technologies. Due to the hydrated and porous structures of the hydrogel, they are used as a support matrix to deliver cells to the infarct tissue. In heart tissue regeneration, bioactive and biodegradable hydrogels are required by simulating native tissue microenvironments to support myocardial wall stress in addition to preserving cells. Recently, the use of nanostructured hydrogels has increased the use of nanocomposite hydrogels and has revolutionized the field of cardiac tissue engineering. Therefore, to overcome the limitation of the use of hydrogels due to their mechanical fragility, various nanoparticles of polymers, metal, and carbon are used in tissue engineering and create a new opportunity to provide hydrogels with excellent properties. Here, the types of synthetic and natural polymer hydrogels, nanocarbon-based hydrogels, and other nanoparticle-based materials used for cardiac tissue engineering with emphasis on conductive nanostructured hydrogels are briefly introduced.
Collapse
Affiliation(s)
- Sania Ghobadi Alamdari
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, Burjassot, Valencia, 46100, Spain
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Reza Mohammadzadeh
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran.,Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran
| |
Collapse
|
34
|
Ramachandran V, Mohanasundaram T, Tiwari R, Tiwari G, Vijayakumar P, Bhongiri B, Xavier RM. Nrf2 Mediated Heme Oxygenase-1 Activation Contributes to Diabetic Wound Healing - an Overview. Drug Res (Stuttg) 2022; 72:487-495. [PMID: 35931068 DOI: 10.1055/a-1899-8233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Diabetic wound healing is a complicated procedure because hyperglycemia changes the various stages of wound healing. In type 2 diabetes mellitus (T2DM), oxidative stress is proven to be a critical factor in causing non-healing wounds and aggravating the inflammatory phase, resulting in the amputation of lower limbs in T2DM patients. This makes scientists figure out how to control oxidative stress and chronic inflammation at the molecular level. Nuclear factor erythroid 2- related factor 2 (Nrf2) releases antioxidant proteins to suppress reactive oxygen species (ROS) activation and inflammation. The current review discusses the role of Nrf2 in improving diabetic wound healing by reducing the production of ROS and thus reducing oxidative stress, as well as inhibiting nuclear factor kappa B (NF-kB) dissociation and nuclear translocation, which prevents the release of inflammatory mediators and increases antioxidant protein levels, thereby improving diabetic wound healing. As a result, the researcher will be able to find a more effective diabetic wound healing therapy.
Collapse
Affiliation(s)
- Vadivelan Ramachandran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamilnadu, India
| | - Tharani Mohanasundaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamilnadu, India
| | - Ruchi Tiwari
- Pranveer Singh institute of Technology (Pharmacy), Kanpur - Agra - Delhi, NH2, Bhauti, Kanpur, Uttar Pradesh, India
| | - Gaurav Tiwari
- Pranveer Singh institute of Technology (Pharmacy), Kanpur - Agra - Delhi, NH2, Bhauti, Kanpur, Uttar Pradesh, India
| | - Putta Vijayakumar
- Department of Pharmacy Practice, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamilnadu, India
| | - Bhargav Bhongiri
- Department of Pharmacy Practice, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamilnadu, India
| | - Rinu Mary Xavier
- Department of Pharmacy Practice, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamilnadu, India
| |
Collapse
|
35
|
Zhang Y, van den Beucken JJJP. Special Issue: Immunomodulatory Methods Toward Tissue Regeneration. Tissue Eng Part C Methods 2022; 28:375-376. [PMID: 35920866 DOI: 10.1089/ten.tec.2022.29034.editorial] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, China.,School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jeroen J J P van den Beucken
- Department of Dentistry-Regenerative Biomaterials, Radboudumc, Nijmegen, the Netherlands.,Radboud Institute for Molecular Life Sciences (RIMLS)-Theme Reconstructive & Regenerative Medicine, Nijmegen, the Netherlands
| |
Collapse
|
36
|
Chen M, Jiang R, Deng N, Zhao X, Li X, Guo C. Natural polymer-based scaffolds for soft tissue repair. Front Bioeng Biotechnol 2022; 10:954699. [PMID: 35928962 PMCID: PMC9343850 DOI: 10.3389/fbioe.2022.954699] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Soft tissues such as skin, muscle, and tendon are easily damaged due to injury from physical activity and pathological lesions. For soft tissue repair and regeneration, biomaterials are often used to build scaffolds with appropriate structures and tailored functionalities that can support cell growth and new tissue formation. Among all types of scaffolds, natural polymer-based scaffolds attract much attention due to their excellent biocompatibility and tunable mechanical properties. In this comprehensive mini-review, we summarize recent progress on natural polymer-based scaffolds for soft tissue repair, focusing on clinical translations and materials design. Furthermore, the limitations and challenges, such as unsatisfied mechanical properties and unfavorable biological responses, are discussed to advance the development of novel scaffolds for soft tissue repair and regeneration toward clinical translation.
Collapse
Affiliation(s)
- Meiwen Chen
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
| | - Rui Jiang
- School of Engineering, Westlake University, Hangzhou, Zhejiang
| | - Niping Deng
- School of Engineering, Westlake University, Hangzhou, Zhejiang
| | - Xiumin Zhao
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
| | - Xiangjuan Li
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
- *Correspondence: Xiangjuan Li, ; Chengchen Guo,
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, Zhejiang
- *Correspondence: Xiangjuan Li, ; Chengchen Guo,
| |
Collapse
|
37
|
Yu YH, Shen SJ, Hsu YH, Chou YC, Yu PC, Liu SJ. Tri-Layered Doxycycline-, Collagen- and Bupivacaine-Loaded Poly(lactic-co-glycolic acid) Nanofibrous Scaffolds for Tendon Rupture Repair. Polymers (Basel) 2022; 14:polym14132659. [PMID: 35808704 PMCID: PMC9269609 DOI: 10.3390/polym14132659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 01/05/2023] Open
Abstract
Achilles tendon rupture is a severe injury, and its optimal therapy remains controversial. Tissue engineering scaffolds play a significant role in tendon healing and tissue regeneration. In this study, we developed tri-layered doxycycline/collagen/bupivacaine (DCB)-composite nanofibrous scaffolds to repair injured Achilles tendons. Doxycycline, collagen, and bupivacaine were integrated into poly(lactic-co-glycolic acid) (PLGA) nanofibrous membranes, layer by layer, using an electrospinning technique as healing promoters, a 3D scaffold, and painkillers, respectively. After spinning, the properties of the nanofibrous scaffolds were characterized. In vitro drug discharge behavior was also evaluated. Furthermore, the effectiveness of the DCB–PLGA-composite nanofibers in repairing ruptured Achilles tendons was investigated in an animal tendon model with histological analyses. The experimental results show that, compared to the pristine PLGA nanofibers, the biomolecule-loaded nanofibers exhibited smaller fiber size distribution and an enhanced hydrophilicity. The DCB-composite nanofibers provided a sustained release of doxycycline and bupivacaine for over 28 days in vivo. Additionally, Achilles tendons repaired using DCB-composite nanofibers exhibited a significantly higher maximum load-to-failure than normal tendons, suggesting that the biomolecule-incorporated nanofibers are promising scaffolds for repairing Achilles tendons.
Collapse
Affiliation(s)
- Yi-Hsun Yu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.); (Y.-H.H.); (Y.-C.C.)
| | - Shih-Jyun Shen
- Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan;
| | - Yung-Heng Hsu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.); (Y.-H.H.); (Y.-C.C.)
| | - Ying-Chao Chou
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.); (Y.-H.H.); (Y.-C.C.)
| | - Ping-Chun Yu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Shih-Jung Liu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.); (Y.-H.H.); (Y.-C.C.)
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
- Correspondence: ; Tel.: +886-3-2118166; Fax: +886-3-2118558
| |
Collapse
|
38
|
Khosravi N, Pishavar E, Baradaran B, Oroojalian F, Mokhtarzadeh A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: A promising biomimetic nanoplatforms for cancer theranostics. J Control Release 2022; 348:706-722. [PMID: 35732250 DOI: 10.1016/j.jconrel.2022.06.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Nanomedicine research has advanced dramatically in recent decades. Nonetheless, traditional nanomedicine faces significant obstacles such as the low concentration of the drug at target sites and accelerated removal of the drug from blood circulation. Various techniques of nanotechnology, including cell membrane coating, have been developed to address these challenges and to improve targeted distribution and redcue cell membrane-mediated immunogenicity. Recently, stem cell (SC) membranes, owing to their immunosuppressive and regenerative properties, have grabbed attention as attractive therapeutic carriers for targeting specific tissues or organs. Bioengineering strategies that combine synthetic nanoparticles (NPs) with SC membranes, because of their homing potential and tumor tropism, have recently received a lot of publicity. Several laboratory experiments and clinical trials have indicated that the benefits of SC-based technologies are mostly related to the effects of SC-derived exosomes (SC-Exos). Exosomes are known as nano-sized extracellular vehicles (EVs) that deliver particular bioactive molecules for cell-to-cell communication. In this regard, SC-derived exosome membranes have recently been employed to improve the therapeutic capability of engineered drug delivery vehicles. Most recently, for further enhancing NPs' functionality, a new coating approach has been offered that combines membranes from two separate cells. These hybrid membrane delivery vehicles have paved the way for the development of biocompatible, high-efficiency, biomimetic NPs with varying hybrid capabilities that can overcome the drawbacks of present NP-based treatment techniques. This review explores stem cell membranes, SC-Exos, and hybrid SC-camouflaged NPs preparation methods and their importance in cancer therapy.
Collapse
Affiliation(s)
- Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Pishavar
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
39
|
Rogozinski N, Yanez A, Bhoi R, Lee MY, Yang H. Current methods for fabricating 3D cardiac engineered constructs. iScience 2022; 25:104330. [PMID: 35602954 PMCID: PMC9118671 DOI: 10.1016/j.isci.2022.104330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
3D cardiac engineered constructs have yielded not only the next generation of cardiac regenerative medicine but also have allowed for more accurate modeling of both healthy and diseased cardiac tissues. This is critical as current cardiac treatments are rudimentary and often default to eventual heart transplants. This review serves to highlight the various cell types found in cardiac tissues and how they correspond with current advanced fabrication methods for creating cardiac engineered constructs capable of shedding light on various pathologies and providing the therapeutic potential for damaged myocardium. In addition, insight is given toward the future direction of the field with an emphasis on the creation of specialized and personalized constructs that model the region-specific microtopography and function of native cardiac tissues.
Collapse
Affiliation(s)
- Nicholas Rogozinski
- Department of Biomedical Engineering, University of North Texas, 3940 N. Elm Street K240B, Denton, TX 76207-7102, USA
| | - Apuleyo Yanez
- Department of Biomedical Engineering, University of North Texas, 3940 N. Elm Street K240B, Denton, TX 76207-7102, USA
| | - Rahulkumar Bhoi
- Department of Biomedical Engineering, University of North Texas, 3940 N. Elm Street K240B, Denton, TX 76207-7102, USA
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, 3940 N. Elm Street K240B, Denton, TX 76207-7102, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, 3940 N. Elm Street K240B, Denton, TX 76207-7102, USA
| |
Collapse
|
40
|
Li X, Chen S, Peng Y, Zheng Z, Li J, Zhong F. Materials, Preparation Strategies, and Wearable Sensor Applications of Conductive Fibers: A Review. SENSORS (BASEL, SWITZERLAND) 2022; 22:3028. [PMID: 35459012 PMCID: PMC9032468 DOI: 10.3390/s22083028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 05/07/2023]
Abstract
The recent advances in wearable sensors and intelligent human-machine interfaces have sparked a great many interests in conductive fibers owing to their high conductivity, light weight, good flexibility, and durability. As one of the most impressive materials for wearable sensors, conductive fibers can be made from a variety of raw sources via diverse preparation strategies. Herein, to offer a comprehensive understanding of conductive fibers, we present an overview of the recent progress in the materials, the preparation strategies, and the wearable sensor applications related. Firstly, the three types of conductive fibers, including metal-based, carbon-based, and polymer-based, are summarized in terms of their principal material composition. Then, various preparation strategies of conductive fibers are established. Next, the primary wearable sensors made of conductive fibers are illustrated in detail. Finally, a robust outlook on conductive fibers and their wearable sensor applications are addressed.
Collapse
Affiliation(s)
| | | | | | | | | | - Fei Zhong
- School of Mechanical Engineering, Hubei University of Technology, Wuhan 430068, China; (X.L.); (S.C.); (Y.P.); (Z.Z.); (J.L.)
| |
Collapse
|
41
|
Guo M, Wang X, Liu Y, Yu H, Dong J, Cui Z, Bai Z, Li K, Li Q. Hierarchical Shish-Kebab Structures Functionalizing Nanofibers for Controlled Drug Release and Improved Antithrombogenicity. Biomacromolecules 2022; 23:1337-1349. [PMID: 35235295 DOI: 10.1021/acs.biomac.1c01572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The functionalization of the fibrous scaffolds including drug loading and release is of significance in tissue engineering and regenerative medicine. Our previous results have shown that the shish-kebab structure-modified fibrous scaffold shows a completely different microenvironment that mimics the topography of the collagen fibers, which interestingly facilitates the cell adhesion and migration. However, the functionalization of the unique structure needs to be further investigated. In this study, we modified the heparin-loaded fiber with a shish-kebab structure and tuned the kebab structure as the barrier for the sustained release of heparin. The introduction of the kebab structure increases the diffusion energy barrier by extending the diffusion distance. Moreover, the discontinued surface topography of the shish-kebab structure altered the surface chemistry from hydrophobic for the original poly(ε-caprolactone) (PCL) nanofibers to hydrophilic for the PCL nanofibers with the shish-kebab structure, which might have inhibited the activation of fibrinogen and thus improved the anticoagulant ability. This synergistic effect of heparin and the kebab structure significantly promotes the endothelial cell affinity and antithrombogenicity. This method might be a viable and versatile drug delivery strategy in vascular tissue engineering.
Collapse
Affiliation(s)
- Meng Guo
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yajing Liu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Haichang Yu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Jiahui Dong
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zhixiang Cui
- Department of Materials Science and Engineering, Fujian University of Technology, Fuzhou 350118, China
| | - Zhiyuan Bai
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China.,National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
42
|
Features and Methods of Making Nanofibers by Electrospinning, Phase Separation and Self-assembly. JORJANI BIOMEDICINE JOURNAL 2022. [DOI: 10.52547/jorjanibiomedj.10.1.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
43
|
Zhu X, Kazemi A, Dong Y, Pan Q, Jin P, Cheng B, Yang Y. Effectiveness of Nano Bioactive Glass Fiber Loaded with Platelet-Rich Plasma on Thermal Wound Healing Process in Rats. J Biomed Nanotechnol 2022; 18:535-545. [PMID: 35484761 DOI: 10.1166/jbn.2022.3249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In this study we evaluated the impact of topical application of bioactive glass fibers loaded PRP on a deep seconddegree thermal wound and its healing process sub-streaming molecular pathway of re-epithelialization. Wistar rats were randomly divided into four groups: normal control group, model group (deep second-degree thermal wound), PRP group, and PRP+nanobioactive glass fiber group. After treatment, the changes of wounds were observed daily. H&E staining was used to evaluate the pathological changes and also, qRT-PCR was used to detect the mRNA expression of KGF, IL-1, IL-6, IL-10, TGF-β, EGF, VEGF, HIF-1α, integrin α3 and integrin β1 in wound tissues. In the current study, we observed that PRP group and the PRP group basically re-epithelized on the 21st day. The wound healing rates of the PRP+nanobioactive glass fiber group and PRP group at each time point were higher than those in the model group, while there was no significant difference in wound healing rate between the PRP+nanobioactive glass fiber group and PRP group at each time point. H&E staining showed that the pathological scores of skin wound repairing in the PRP+nanobioactive glass fiber group on the 7th, 14th and 21st day were higher than that of in the model group. The qPCR results suggested the mRNA expression of IL-1, IL-6 and IL-10 in the PRP+nanobioactive glass fiber group and the PRP group were lower than those in the untreated group on the 14th day; the expression of VEGF and EGF mRNA were higher on the 3rd day; the mRNA expression of TGF-β, HIF-1α showed a tendency of increasing first and decreasing then; integrin β1 mRNA expression increased significantly, which was highest; integrin α3 mRNA expression was higher on day 3rd and 21th, respectively. The PRP+nanobioactive glass fibers and PRP can shorten the wound healing time and improve the healing quality mainly by promoting the wound epithelization through increasing the expression of EGF, VEGF, TGF-β, HIF-1α, Integrin α3, and meanwhile increasing the release of Integrin β1 and other mechanisms.
Collapse
Affiliation(s)
- Xuanru Zhu
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, China
| | - Aida Kazemi
- Clinical Research Development Unit, Shafa Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Yunqing Dong
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, China
| | - Qiao Pan
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, China
| | - Panshi Jin
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, China
| | - Biao Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, 510000, China
| | - Yangog Yang
- Rehabilation Unit, CHU Gabriel Touré, Bamako, 267, Mali
| |
Collapse
|
44
|
Scrapie-Responsive Gene 1 Promotes Chondrogenic Differentiation of Umbilical Cord Mesenchymal Stem Cells via Wnt5a. Stem Cells Int 2022; 2022:9124277. [PMID: 35126528 PMCID: PMC8813292 DOI: 10.1155/2022/9124277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/25/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022] Open
Abstract
Objective Repair of cartilage defects, a common condition resulting from many factors, is still a great challenge. Based on their chondrogenic differentiation ability, mesenchymal stem cell- (MSC-) based cartilage regeneration is a promising approach for cartilage defect repair. However, MSC differentiation into chondroblasts or related cell lineages is elaborately controlled by stem cell differentiation stage factors and affected by an array of bioactive elements, which may impede the efficient production of target cells. Thus, identifying a single transcription factor to promote chondrogenic differentiation is critical. Herein, we explored the mechanism by which scrapie-responsive gene 1 (SCRG1), a candidate gene for cartilage regeneration promotion, regulates chondrogenic differentiation of MSCs. Methods Expression of SCRG1 was detected in umbilical cord-derived MSCs (UCMSCs) by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemical analysis during chondrogenic differentiation. The function of SCRG1 in chondrogenic potential was evaluated after gene knockdown or overexpression by lentiviral vectors. Finally, a rabbit cartilage defect model was established to evaluate the effect of SCRG1 on cartilage repair in vivo. Results Expression of SCRG1 was upregulated during in vitro chondrogenic differentiation of UCMSCs. SCRG1 knockdown inhibited chondrogenic differentiation of UCMSCs, while SCRG1 overexpression promoted chondrogenic differentiation of UCMSCs in vitro. In addition, UCMSC overexpressing SCRG1 promoted cartilage repair in vivo. Mechanistically, SCRG1 promoted chondrogenic differentiation via upregulation of Wnt5a expression and subsequent inhibition of β-catenin. Conclusion Our results showed that SCRG1 promotes chondrogenic differentiation of UCMSCs by inhibiting canonical Wnt/β-catenin signaling through Wnt5a. Our findings provide a future target for chondrogenic differentiation and cartilage regeneration.
Collapse
|
45
|
Minardi S. Editorial: Nano-Biomaterials for the Delivery of Therapeutic and Biological Cues for Regenerative Medicine. Front Bioeng Biotechnol 2022; 9:832487. [PMID: 35096803 PMCID: PMC8792963 DOI: 10.3389/fbioe.2021.832487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Silvia Minardi
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute of BioNanotechnology, Northwestern University, Chicago, IL, United States
- *Correspondence: Silvia Minardi,
| |
Collapse
|
46
|
Fu L, Li P, Zhu J, Liao Z, Gao C, Li H, Yang Z, Zhao T, Chen W, Peng Y, Cao F, Ning C, Sui X, Guo Q, Lin Y, Liu S. Tetrahedral framework nucleic acids promote the biological functions and related mechanism of synovium-derived mesenchymal stem cells and show improved articular cartilage regeneration activity in situ. Bioact Mater 2021; 9:411-427. [PMID: 34820580 PMCID: PMC8586787 DOI: 10.1016/j.bioactmat.2021.07.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/25/2021] [Accepted: 07/22/2021] [Indexed: 02/08/2023] Open
Abstract
Many recent studies have shown that joint-resident mesenchymal stem cells (MSCs) play a vital role in articular cartilage (AC) in situ regeneration. Specifically, synovium-derived MSCs (SMSCs), which have strong chondrogenic differentiation potential, may be the main driver of cartilage repair. However, both the insufficient number of MSCs and the lack of an ideal regenerative microenvironment in the defect area will seriously affect the regeneration of AC. Tetrahedral framework nucleic acids (tFNAs), notable novel nanomaterials, are considered prospective biological regulators in biomedical engineering. Here, we aimed to explore whether tFNAs have positive effects on AC in situ regeneration and to investigate the related mechanism. The results of in vitro experiments showed that the proliferation and migration of SMSCs were significantly enhanced by tFNAs. In addition, tFNAs, which were added to chondrogenic induction medium, were shown to promote the chondrogenic capacity of SMSCs by increasing the phosphorylation of Smad2/3. In animal models, the injection of tFNAs improved the therapeutic outcome of cartilage defects compared with that of the control treatments without tFNAs. In conclusion, this is the first report to demonstrate that tFNAs can promote the chondrogenic differentiation of SMSCs in vitro and enhance AC regeneration in vivo, indicating that tFNAs may become a promising therapeutic for AC regeneration. Tetrahedral framework nucleic acids (tFNAs) can promote SMSCs proliferation by activating the Wnt/β-catenin pathway. tFNAs can promote SMSCs migration in vitro and vivo. tFNAs can promote SMSCs chondrogenic differentiation by regulating the TGF/Smad2/3 signaling pathway. tFNAs show improved articular cartilage in situ regeneration activity in vivo.
Collapse
Affiliation(s)
- Liwei Fu
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Pinxue Li
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Junyao Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.,Stomatology Department, The Fifth Hospital of Sichuan Province, Chengdu, 610031, People's Republic of China
| | - Zhiyao Liao
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Cangjian Gao
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Hao Li
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Zhen Yang
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Tianyuan Zhao
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Wei Chen
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Yu Peng
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Fuyang Cao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Chao Ning
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Xiang Sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| |
Collapse
|
47
|
Characterization and Topical Study of Aloe Vera Hydrogel on Wound-Healing Process. Polymers (Basel) 2021; 13:polym13223958. [PMID: 34833257 PMCID: PMC8623201 DOI: 10.3390/polym13223958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022] Open
Abstract
Wound healing is fundamental to restore the tissue integrity. A topical study of the influence of Aloe vera hydrogel, formulated with 1,2-propanediol (propanediol) and triethanolamine (TEA), on the skin wound-healing process was investigated in female Wistar rats. FTIR spectroscopy confirms the presence of carboxylic acid and methyl ester carboxylate groups related with important compounds that confer the hydrogel a good interaction with proteins and growth factors. SEM images show a microstructure and micro-roughness that promote a good adhesion to the wound. Therefore, the swelling kinetics and the contact angle response contribute to the understanding of the in vivo results of the animal test. The results indicated that the Aloe vera hydrogel, prepared with propanediol and TEA, together with its superficial characteristics, improve its rapid penetration without drying out the treated tissue. This produced a positive influence on inflammation, angiogenesis, and wound contraction, reducing 29% the total healing time, reaching the total closure of the wound in 15 days.
Collapse
|
48
|
Liu Z, Wang J, Chen H, Zhang G, Lv Z, Li Y, Zhao S, Li W. Coaxial Electrospun PLLA Fibers Modified with Water-Soluble Materials for Oligodendrocyte Myelination. Polymers (Basel) 2021; 13:polym13203595. [PMID: 34685353 PMCID: PMC8537353 DOI: 10.3390/polym13203595] [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: 09/02/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Myelin sheaths are essential in maintaining the integrity of axons. Development of the platform for in vitro myelination would be especially useful for demyelinating disease modeling and drug screening. In this study, a fiber scaffold with a core-shell structure was prepared in one step by the coaxial electrospinning method. A high-molecular-weight polymer poly-L-lactic acid (PLLA) was used as the core, while the shell was a natural polymer material such as hyaluronic acid (HA), sodium alginate (SA), or chitosan (CS). The morphology, differential scanning calorimetry (DSC), Fourier transform infrared spectra (FTIR), contact angle, viability assay, and in vitro myelination by oligodendrocytes were characterized. The results showed that such fibers are bead-free and continuous, with an average size from 294 ± 53 to 390 ± 54 nm. The DSC and FTIR curves indicated no changes in the phase state of coaxial brackets. Hyaluronic acid/PLLA coaxial fibers had the minimum contact angle (53.1° ± 0.24°). Myelin sheaths were wrapped around a coaxial electrospun scaffold modified with water-soluble materials after a 14-day incubation. All results suggest that such a scaffold prepared by coaxial electrospinning potentially provides a novel platform for oligodendrocyte myelination.
Collapse
Affiliation(s)
- Zhepeng Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.W.); (H.C.); (Y.L.); (S.Z.)
- Correspondence: (Z.L.); (W.L.)
| | - Jing Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.W.); (H.C.); (Y.L.); (S.Z.)
| | - Haini Chen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.W.); (H.C.); (Y.L.); (S.Z.)
| | - Guanyu Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; (G.Z.); (Z.L.)
| | - Zhuman Lv
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; (G.Z.); (Z.L.)
| | - Yijun Li
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.W.); (H.C.); (Y.L.); (S.Z.)
| | - Shoujin Zhao
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.W.); (H.C.); (Y.L.); (S.Z.)
| | - Wenlin Li
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; (G.Z.); (Z.L.)
- Correspondence: (Z.L.); (W.L.)
| |
Collapse
|
49
|
Rapid Magneto-Sonoporation of Adipose-Derived Cells. MATERIALS 2021; 14:ma14174877. [PMID: 34500968 PMCID: PMC8432646 DOI: 10.3390/ma14174877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023]
Abstract
By permeabilizing the cell membrane with ultrasound and facilitating the uptake of iron oxide nanoparticles, the magneto-sonoporation (MSP) technique can be used to instantaneously label transplantable cells (like stem cells) to be visualized via magnetic resonance imaging in vivo. However, the effects of MSP on cells are still largely unexplored. Here, we applied MSP to the widely applicable adipose-derived stem cells (ASCs) for the first time and investigated its effects on the biology of those cells. Upon optimization, MSP allowed us to achieve a consistent nanoparticle uptake (in the range of 10 pg/cell) and a complete membrane resealing in few minutes. Surprisingly, this treatment altered the metabolic activity of cells and induced their differentiation towards an osteoblastic profile, as demonstrated by an increased expression of osteogenic genes and morphological changes. Histological evidence of osteogenic tissue development was collected also in 3D hydrogel constructs. These results point to a novel role of MSP in remote biophysical stimulation of cells with focus application in bone tissue repair.
Collapse
|
50
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|