1
|
Rana I, Deepa, Aslam M, Ranjan KR, Singh P, Kumari K. A review on the use of composites of a natural protein, silk fibroin with Mxene/carbonaceous materials in biomedical science. Int J Biol Macromol 2024; 278:135101. [PMID: 39227275 DOI: 10.1016/j.ijbiomac.2024.135101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/19/2024] [Accepted: 08/25/2024] [Indexed: 09/05/2024]
Abstract
Silk fibroin (SF), a natural biodegradable and biocompatible protein, has garnered significant attention in biomedical applications due to its impressive properties, including excellent biocompatibility, biodegradability, and mechanical resilience. Nevertheless, its broader usage faces obstacles by its insufficient mechanical strength and electrical conductivity. In order to address these constraints, recent studies have concentrated on combining SF with cutting-edge nanomaterials like MXene and carbon-based materials. This review comprehensively examines the applications and potential of silk fibroin-MXene/carbon-based nanocomposites in biomedical fields. The unique properties of SF, MXene, and carbon-based materials are explored, emphasizing how their combination enhances mechanical strength, conductivity, and biocompatibility. These composites show substantial enhancements in performance for several biomedical applications by utilising the excellent conductivity and mechanical capabilities of MXene and carbonaceous elements. The innovative potential of these nanocomposites is highlighted by critically discussing key applications such as tissue engineering, drug delivery, and biosensing. In addition, the work discusses the latest research progress, difficulties, and future prospects in the sector, providing valuable insights into possible breakthroughs and uses. This review seeks to comprehensively analyse the existing information on silk fibroin-MXene/carbon based nanocomposites in healthcare.
Collapse
Affiliation(s)
- Ishika Rana
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida 201303, India
| | - Deepa
- Department of Zoology, University of Delhi, Delhi 110007, India
| | - Mohd Aslam
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, New Delhi, India
| | - Kumar Rakesh Ranjan
- Department of Chemistry, Amity Institute of Applied Sciences, Amity University Uttar Pradesh, Noida 201303, India
| | - Prashant Singh
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, New Delhi, India.
| | - Kamlesh Kumari
- Department of Zoology, University of Delhi, Delhi 110007, India.
| |
Collapse
|
2
|
Waidi YO, Debnath S, Datta S, Chatterjee K. 3D-Printed Silk Proteins for Bone Tissue Regeneration and Associated Immunomodulation. Biomacromolecules 2024; 25:5512-5540. [PMID: 39133748 DOI: 10.1021/acs.biomac.4c00540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Current bone repair methods have limitations, prompting the exploration of innovative approaches. Tissue engineering emerges as a promising solution, leveraging biomaterials to craft scaffolds replicating the natural bone environment, facilitating cell growth and differentiation. Among fabrication techniques, three-dimensional (3D) printing stands out for its ability to tailor intricate scaffolds. Silk proteins (SPs), known for their mechanical strength and biocompatibility, are an excellent choice for engineering 3D-printed bone tissue engineering (BTE) scaffolds. This article comprehensively reviews bone biology, 3D printing, and the unique attributes of SPs, specifically detailing criteria for scaffold fabrication such as composition, structure, mechanics, and cellular responses. It examines the structural, mechanical, and biological attributes of SPs, emphasizing their suitability for BTE. Recent studies on diverse 3D printing approaches using SPs-based for BTE are highlighted, alongside advancements in their 3D and four-dimensional (4D) printing and their role in osteo-immunomodulation. Future directions in the use of SPs for 3D printing in BTE are outlined.
Collapse
Affiliation(s)
- Yusuf Olatunji Waidi
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Souvik Debnath
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Sudipto Datta
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Bioengineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
3
|
Sun Y, Ma L, Wei T, Zheng M, Mao C, Yang M, Shuai Y. Green, Low-carbon Silk-based Materials in Water Treatment: Current State and Future Trends. CHEMSUSCHEM 2024; 17:e202301549. [PMID: 38298106 DOI: 10.1002/cssc.202301549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
The improper and inadequate treatment of industrial, agricultural, and household wastewater exerts substantial pressure on the existing ecosystem and poses a serious threat to the health of both humans and animals. To address these issues, different types of materials have been employed to eradicate detrimental pollutants from wastewater and facilitate the reuse of water resources. Nevertheless, owing to the challenges associated with the degradation of these traditional materials post-use and their incompatibility with the environment, natural biopolymers have garnered considerable interest. Silk protein, as a biomacromolecule, exhibits advantageous characteristics including environmental friendliness, low carbon emissions, biodegradability, sustainability, and biocompatibility. Considering recent research findings, this comprehensive review outlines the structure and properties of silk proteins and offers a detailed overview of the manufacturing techniques employed in the production of silk-based materials (SBMs) spanning different forms. Furthermore, it conducts an in-depth analysis of the state-of-the-art SBMs for water treatment purposes, encompassing adsorption, catalysis, water disinfection, desalination, and biosensing. The review highlights the potential of SBMs in addressing the challenges of wastewater treatment and provides valuable insights into prospective avenues for further research.
Collapse
Affiliation(s)
- Yuxu Sun
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| | - Lantian Ma
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| | - Tiancheng Wei
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| | - Meidan Zheng
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, 310027, Hangzhou, Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, 999077, Hong Kong SAR, P. R.China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| | - Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 310058, Hangzhou, China
| |
Collapse
|
4
|
Spessot E, Passuello S, Shah LV, Maniglio D, Motta A. Nanocomposite Methacrylated Silk Fibroin-Based Scaffolds for Bone Tissue Engineering. Biomimetics (Basel) 2024; 9:218. [PMID: 38667229 PMCID: PMC11048339 DOI: 10.3390/biomimetics9040218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The treatment of bone defects is a clinical challenge. Bone tissue engineering is gaining interest as an alternative to current treatments, with the development of 3D porous structures (scaffolds) helpful in promoting bone regeneration by ensuring temporary functional support. In this work, methacrylated silk fibroin (SilMA) sponges were investigated as scaffolds for bone tissue engineering by exploiting the combination of physical (induced by NaCl salt during particulate leaching) and chemical crosslinking (induced by UV-light exposure) techniques. A biomimetic approach was adopted to better simulate the extracellular matrix of the bone by introducing either natural (mussel shell-derived) or synthetic-origin hydroxyapatite nanoparticles into the SilMA sponges. The obtained materials were characterized in terms of pore size, water absorption capability and mechanical properties to understand both the effect of the inclusion of the two different types of nanoparticles and the effect of the photocrosslinking. Moreover, the SilMA sponges were tested for their bioactivity and suitability for bone tissue engineering purposes by using osteosarcoma cells, studying their metabolism by an AlamarBlue assay and their morphology by scanning electron microscopy. Results indicate that photocrosslinking helps in obtaining more regular structures with bimodal pore size distributions and in enhancing the stability of the constructs in water. Moreover, the addition of naturally derived hydroxyapatite was observed to be more effective at activating osteosarcoma cell metabolism than synthetic hydroxyapatite, showing a statistically significant difference in the AlamarBlue measurement on day 7 after seeding. The methacrylated silk fibroin/hydroxyapatite nanocomposite sponges developed in this work were found to be promising tools for targeting bone regeneration with a sustainable approach.
Collapse
Affiliation(s)
- Eugenia Spessot
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Serena Passuello
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
| | - Lekha Vinod Shah
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Devid Maniglio
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Antonella Motta
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [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
|
6
|
Liao S, Qiu G, Hu Y, Guo B, Qiu Y. Separable and Inseparable Silk Fibroin Microneedles for the Transdermal Delivery of Colchicine: Development, Characterization, and Comparisons. AAPS PharmSciTech 2023; 25:3. [PMID: 38114734 DOI: 10.1208/s12249-023-02716-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
Colchicine is the first-line option for both the treatment and prophylaxis of gout flares. However, due to potentially severe side effects, the clinical use of colchicine is limited. A well-tolerated and safe delivery system for colchicine is widely desired. For this purpose, colchicine-loaded inseparable microneedles were fabricated using silk fibroin. Additionally, separable microneedles made of silk fibroin as the needle tips and PVP K30 as the base material were developed. Both types of microneedles were evaluated for their mechanical strength, swelling and dissolution characteristics, insertion abilities, degradation properties, in vitro penetration, skin irritation, and in vivo anti-gout effects. The results demonstrated that separable microneedles had greater mechanical strength and insertion ability. Moreover, the separable microneedles separated quickly and caused little skin irritation. In the pharmacodynamic test, mice with acute gouty arthritis responded significantly to treatment with separable microneedles. In conclusion, the separable silk fibroin-based microneedles provide a promising route for colchicine delivery.
Collapse
Affiliation(s)
- Shiji Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Guirong Qiu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yanping Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Bohong Guo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Yuqin Qiu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
7
|
Fuest S, Smeets R, Gosau M, Aavani F, Knipfer C, Grust ALC, Kopp A, Becerikli M, Behr B, Matthies L. Layer-by-Layer Deposition of Regenerated Silk Fibroin─An Approach to the Surface Coating of Biomedical Implant Materials. ACS Biomater Sci Eng 2023; 9:6644-6657. [PMID: 37983947 DOI: 10.1021/acsbiomaterials.3c00852] [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: 11/22/2023]
Abstract
Biomaterials and coating techniques unlock major benefits for advanced medical therapies. Here, we explored layer-by-layer (LbL) deposition of silk fibroin (SF) by dip coating to deploy homogeneous films on different materials (titanium, magnesium, and polymers) frequently used for orthopedic and other bone-related implants. Titanium and magnesium specimens underwent preceding plasma electrolytic oxidation (PEO) to increase hydrophilicity. This was determined as surface properties were visualized by scanning electron microscopy and contact angle measurements as well as Fourier transform infrared spectroscopy (FTIR) analysis. Finally, biological in vitro evaluations of hemocompatibility, THP-1 cell culture, and TNF-α assays were conducted. A more hydrophilic surface could be achieved using the PEO surface, and the contact angle for magnesium and titanium showed a reduction from 73 to 18° and from 58 to 17°, respectively. Coating with SF proved successful on all three surfaces, and coating thicknesses of up to 5.14 μm (±SD 0.22 μm) were achieved. Using FTIR analysis, it was shown that the insolubility of the material was achieved by post-treatment with water vapor annealing, although the random coil peak (1640-1649 cm-1) and the α-helix peak (at 1650 cm-1) were still evident. SF did not change hemocompatibility, regardless of the substrate, whereas the PEO-coated materials showed improved hemocompatibility. THP-1 cell culture showed that cells adhered excellently to all of the tested material surfaces. Interestingly, SF coatings induced a significantly higher amount of TNF-α for all materials, indicating an inflammatory response, which plays an important role in a variety of physiological processes, including osteogenesis. LbL coatings of SF are shown to be promising candidates to modulate the body's immune response to implants manufactured from titanium, magnesium, and polymers. They may therefore facilitate future applications for bioactive implant coatings. However, further in vivo studies are needed to confirm the proposed effects on osteogenesis in a physiological environment.
Collapse
Affiliation(s)
- Sandra Fuest
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Farzaneh Aavani
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Christian Knipfer
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Audrey Laure Céline Grust
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | | | - Mustafa Becerikli
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, D-44789 Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, D-44789 Bochum, Germany
| | - Levi Matthies
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| |
Collapse
|
8
|
Tan L, Ye Z, Zhuang W, Mao B, Li H, Li X, Wu J, Sang H. 3D printed PLGA/MgO/PDA composite scaffold by low-temperature deposition manufacturing for bone tissue engineering applications. Regen Ther 2023; 24:617-629. [PMID: 38034857 PMCID: PMC10681881 DOI: 10.1016/j.reth.2023.09.015] [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: 05/31/2023] [Revised: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Bones are easily damaged. Biomimetic scaffolds are involved in tissue engineering. This study explored polydopamine (PDA)-coated poly lactic-co-glycolic acid (PLGA)-magnesium oxide (MgO) scaffold properties and its effects on bone marrow mesenchymal stem cells (BMSCs) osteogenic differentiation. Methods PLGA/MgO scaffolds were prepared by low-temperature 3D printing technology and PDA coatings were prepared by immersion method. Scaffold structure was observed by scanning electron microscopy with an energy dispersive spectrometer (SEM-EDS), fourier transform infrared spectrometer (FTIR). Scaffold hydrophilicity, compressive/elastic modulus, and degradation rates were analyzed by water contact angle measurement, mechanical tests, and simulated-body fluid immersion. Rat BMSCs were cultured in scaffold extract. Cell activity on days 1, 3, and 7 was detected by MTT. Cells were induced by osteogenic differentiation, followed by evaluation of alkaline phosphatase (ALP) activity on days 3, 7, and 14 of induction and Osteocalcin, Osteocalcin, and Collagen I expressions. Results The prepared PLGA/MgO scaffolds had dense microparticles. With the increase of MgO contents, the hydrophilicity was enhanced, scaffold degradation rate was accelerated, magnesium ion release rate and scaffold extract pH value were increased, and cytotoxicity was less when magnesium mass ratio was less than 10%. Compared with other scaffolds, compressive and elastic modulus of PLGA/MgO (10%) scaffolds were increased; BMSCs incubated with PLGA/MgO (10%) scaffold extract had higher ALP activity and Osteocalcin, Osteopontin, and Collagen I expressions. PDA coating was prepared in PLGA/MgO (10%) scaffolds and the mechanical properties were not affected. PLGA/MgO (10%)/PDA scaffolds had better hydrophilicity and biocompatibility and promoted BMSC osteogenic differentiation. Conclusion Low-temperature 3D printing PLGA/MgO (10%)/PDA scaffolds had good hydrophilicity and biocompatibility, and were conducive to BMSC osteogenic differentiation.
Collapse
Affiliation(s)
- Liang Tan
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhuofeng Ye
- Department of Orthopedics, Jiangmen Central Hospital, Jiangmen, China
| | - Weida Zhuang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Beini Mao
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
| | - Hetong Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
| | - Xiuwang Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiachang Wu
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen, Guangdong, 518000, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Chimedtseren I, Yamahara S, Akiyama Y, Ito M, Arai Y, Gantugs AE, Nastume N, Wakita T, Hiratsuka T, Honda M, Montenegro Raudales JL. Collagen type I-based recombinant peptide promotes bone regeneration in rat critical-size calvarial defects by enhancing osteoclast activity at late stages of healing. Regen Ther 2023; 24:515-527. [PMID: 37841660 PMCID: PMC10570703 DOI: 10.1016/j.reth.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction We recently demonstrated the bone-forming potential of medium-cross-linked recombinant collagen peptide (mRCP) in animal models of bone defects. However, these studies were limited to a 4-week observation period; therefore, in the present study, we aimed to further evaluate mRCP as a suitable bone graft material for the alveolar cleft by analyzing its bone-forming potential, osteogenic-inducing ability, and biodegradation over an extended period of 12 weeks, using a rat critical-size calvarial defect model. Methods Using Sprague-Dawley rats, we created critical-size calvarial defects through a surgical procedure. The defects were then filled with 3 mg of mRCP (mRCP group) or 18 mg of Cytrans® (CA) granules, which has a carbonate apatite-based composition resembling natural bone, was used as a reference material (CA group). For negative control, the defects were left untreated. Bone volume, total bone volume (bone volume including CA granules), and bone mineral density (BMD) in the defect were assessed using micro-computed tomography (μ-CT) at 0, 4, 8, and 12 weeks after implantation. Using histomorphometric analyses of hematoxylin and eosin (H&E)-stained sections, we measured the amount of newly formed bone and total newly formed bone (new bone including CA granules) in the entire defect site, as well as the amount of newly formed bone in the central side, two peripheral sides (left and right), periosteal (top) side, and dura mater (bottom) side. In addition, we measured the amount of residual bone graft material in the defect. Osteoclasts and osteoblasts in the newly formed bone were detected using tartrate-resistant acid phosphatase (TRAP) and alkaline phosphatase (ALP) staining, respectively. Results Bone volume in the mRCP group increased over time and was significantly larger at 8 and 12 weeks after surgery than at 4 weeks. The bone volume in the mRCP group was greater than that of the CA and control groups at 4, 8, and 12 weeks after implantation, and while the total bone volume was greater in the CA group after 4 and 8 weeks, the mRCP group had comparable levels of total bone volume to that of the CA group at 12 weeks after implantation. The BMD of the mRCP group reached similar levels to native calvaria bone at the same time point. H&E-stained sections revealed a larger amount of newly formed bone 12 weeks after implantation in the mRCP group compared to that of the CA and control groups. The total newly formed bone at 12 weeks after implantation was on par with that in the CA group. Furthermore, at the defect site, the area of newly formed bone was larger on the peripheral and dura mater sides. Notably, the number of osteoclasts in the mRCP group was higher than in the CA and control groups and peaked 8 weeks after implantation, which coincided with the timing of the greatest resorption of mRCP. Although the ALP-positive area was greater in the mRCP group compared to other groups, we did not detect any significant changes in the number of osteoblasts over time. Conclusion This study demonstrated the bone-forming potential of mRCP over an extended period of 12 weeks, suggesting that mRCP sufficiently resists resorption to promote bone formation through induction of osteoclast activation in the late stages of the healing period.
Collapse
Affiliation(s)
- Ichinnorov Chimedtseren
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Shoji Yamahara
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
| | - Yasunori Akiyama
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Masaaki Ito
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Yoshinori Arai
- Department of Oral and Maxillofacial Radiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Anar Erdene Gantugs
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
| | - Nagato Nastume
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
| | - Taku Wakita
- Bio Science & Engineering Laboratory, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa 258-8577, Japan
| | - Takahiro Hiratsuka
- Bio Science & Engineering Laboratory, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa 258-8577, Japan
| | - Masaki Honda
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Jorge Luis Montenegro Raudales
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| |
Collapse
|
10
|
Ahmed TA, Eldaly B, Eldosuky S, Elkhenany H, El-Derby AM, Elshazly MF, El-Badri N. The interplay of cells, polymers, and vascularization in three-dimensional lung models and their applications in COVID-19 research and therapy. Stem Cell Res Ther 2023; 14:114. [PMID: 37118810 PMCID: PMC10144893 DOI: 10.1186/s13287-023-03341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Millions of people have been affected ever since the emergence of the corona virus disease of 2019 (COVID-19) outbreak, leading to an urgent need for antiviral drug and vaccine development. Current experimentation on traditional two-dimensional culture (2D) fails to accurately mimic the in vivo microenvironment for the disease, while in vivo animal model testing does not faithfully replicate human COVID-19 infection. Human-based three-dimensional (3D) cell culture models such as spheroids, organoids, and organ-on-a-chip present a promising solution to these challenges. In this report, we review the recent 3D in vitro lung models used in COVID-19 infection and drug screening studies and highlight the most common types of natural and synthetic polymers used to generate 3D lung models.
Collapse
Affiliation(s)
- Toka A Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Bassant Eldaly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Shadwa Eldosuky
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Muhamed F Elshazly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
11
|
Zhang G, Zhen A, Chen J, Du B, Luo F, Li J, Tan H. In Vitro Effects of Waterborne Polyurethane 3D Scaffolds Containing Poly(lactic-co-glycolic acid)s of Different Lactic Acid/Glycolic Acid Ratios on the Inflammatory Response. Polymers (Basel) 2023; 15:polym15071786. [PMID: 37050400 PMCID: PMC10097270 DOI: 10.3390/polym15071786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
The physical and chemical properties of tissue engineering scaffolds have considerable effects on the inflammatory response at the implant site in soft tissue repair. The development of inflammation-modulating polymer scaffolds for soft tissue repair is attracting increasing attention. In this study, in order to regulate the inflammatory response at the implant site, a series of waterborne polyurethane (WPU) scaffolds with different properties were synthesized using polyethylene glycol (PEG), polycaprolactone (PCL) and poly (lactic acid)–glycolic acid copolymers (PLGAs) with three lactic acid/glycolic acid (LA/GA) ratios as the soft segments. Then, scaffolds were obtained using freeze-drying. The WPU scaffolds exhibited a porous cellular structure, high porosity, proper mechanical properties for repairing nerve tissue and an adjustable degradation rate. In vitro cellular experiments showed that the degradation solution possessed high biocompatibility. The in vitro inflammatory response of C57BL/6 mouse brain microglia (immortalized) (BV2) cells demonstrated that the LA/GA ratio of the PLGA in WPU scaffolds can regulate the external inflammatory response by altering the secretion of IL-10 and TNF-α. Even the IL-10/TNF-α of PU5050 (3.64) reached 69 times that of the control group (0.053). The results of the PC12 culture on the scaffolds showed that the scaffolds had positive effects on the growth, proliferation and differentiation of nerve cells and could even promote the formation of synapses. Overall, these scaffolds, particularly the PU5050, indeed prevent BV2 cells from differentiating into a pro-inflammatory M1 phenotype, which makes them promising candidates for reducing the inflammatory response and repairing nerve tissue. Furthermore, PU5050 had the best effect on preventing the transformation of BV2 cells into the pro-inflammatory M1 phenotype.
Collapse
Affiliation(s)
- Guanyu Zhang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Ao Zhen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jinlin Chen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Bohong Du
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Feng Luo
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jiehua Li
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Hong Tan
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
12
|
Yan S, He L, Hai AM, Hu Z, You R, Zhang Q, Kaplan DL. Controllable Production of Natural Silk Nanofibrils for Reinforcing Silk-Based Orthopedic Screws. Polymers (Basel) 2023; 15:polym15071645. [PMID: 37050259 PMCID: PMC10096991 DOI: 10.3390/polym15071645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
As a natural high-performance material with a unique hierarchical structure, silk is endowed with superior mechanical properties. However, the current approaches towards producing regenerated silk fibroin (SF) for the preparation of biomedical devices fail to fully exploit the mechanical potential of native silk materials. In this study, using a top-down approach, we exfoliated natural silk fibers into silk nanofibrils (SNFs), through the disintegration of interfibrillar binding forces. The as-prepared SNFs were employed to reinforce the regenerated SF solution to fabricate orthopedic screws with outstanding mechanical properties (compression modulus > 1.1 GPa in a hydrated state). Remarkably, these screws exhibited tunable biodegradation and high cytocompatibility. After 28 days of degradation in protease XIV solution, the weight loss of the screw was ~20% of the original weight. The screws offered a favorable microenvironment to human bone marrow mesenchymal stem cell growth and spread as determined by live/dead staining, F-action staining, and Alamar blue staining. The synergy between native structural components (SNFs) and regenerated SF solutions to form bionanocomposites provides a promising design strategy for the fabrication of biomedical devices with improved performance.
Collapse
|
13
|
Milazzo M, Fitzpatrick V, Owens CE, Carraretto IM, McKinley GH, Kaplan DL, Buehler MJ. 3D Printability of Silk/Hydroxyapatite Composites for Microprosthetic Applications. ACS Biomater Sci Eng 2023; 9:1285-1295. [PMID: 36857509 DOI: 10.1021/acsbiomaterials.2c01357] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Micro-prosthetics requires the fabrication of mechanically robust and personalized components with sub-millimetric feature accuracy. Three-dimensional (3D) printing technologies have had a major impact on manufacturing such miniaturized devices for biomedical applications; however, biocompatibility requirements greatly constrain the choice of usable materials. Hydroxyapatite (HA) and its composites have been widely employed to fabricate bone-like structures, especially at the macroscale. In this work, we investigate the rheology, printability, and prosthetic mechanical properties of HA and HA-silk protein composites, focusing on the roles of composition and water content. We correlate key linear and nonlinear shear rheological parameters to geometric outcomes of printing and explain how silk compensates for the inherent brittleness of printed HA components. By increasing ink ductility, the inclusion of silk improves the quality of printed items through two mechanisms: (1) reducing underextrusion by lowering the required elastic modulus and, (2) reducing slumping by increasing the ink yield stress proportional to the modulus. We demonstrate that the elastic modulus and compressive strength of parts fabricated from silk-HA inks are higher than those for rheologically comparable pure-HA inks. We construct a printing map to guide the manufacturing of HA-based inks with excellent final properties, especially for use in biomedical applications for which sub-millimetric features are required.
Collapse
Affiliation(s)
- Mario Milazzo
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology (MIT), Massachusetts Avenue 77, Cambridge, Massachusetts 02139, United States
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56122 Pisa, Italy
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Crystal E Owens
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Igor M Carraretto
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Energy, Politecnico di Milano, via Lambruschini 4a, 20156 Milano, MI, Italy
| | - Gareth H McKinley
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Markus J Buehler
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology (MIT), Massachusetts Avenue 77, Cambridge, Massachusetts 02139, United States
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
14
|
Zuluaga-Vélez A, Toro-Acevedo CA, Quintero-Martinez A, Melchor-Moncada JJ, Pedraza-Ordoñez F, Aguilar-Fernández E, Sepúlveda-Arias JC. Performance of Colombian Silk Fibroin Hydrogels for Hyaline Cartilage Tissue Engineering. J Funct Biomater 2022; 13:297. [PMID: 36547557 PMCID: PMC9788426 DOI: 10.3390/jfb13040297] [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: 11/18/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
The development and evaluation of scaffolds play a crucial role in the engineering of hyaline cartilage tissue. This work aims to evaluate the performance of silk fibroin hydrogels fabricated from the cocoons of the Colombian hybrid in the in vitro regeneration of hyaline cartilage. The scaffolds were physicochemically characterized, and their performance was evaluated in a cellular model. The results showed that the scaffolds were rich in random coils and β-sheets in their structure and susceptible to various serine proteases with different degradation profiles. Furthermore, they showed a significant increase in ACAN, COL10A1, and COL2A1 expression compared to pellet culture alone and allowed GAG deposition. The soluble portion of the scaffold did not affect chondrogenesis. Furthermore, they promoted the increase in COL1A2, showing a slight tendency to differentiate towards fibrous cartilage. The results also showed that Colombian silk could be used as a source of biomedical devices, paving the way for sericulture to become a more diverse economic activity in emerging countries.
Collapse
Affiliation(s)
- Augusto Zuluaga-Vélez
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - Carlos Andrés Toro-Acevedo
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - Adrián Quintero-Martinez
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, México City 04510, Mexico
| | - Jhon Jairo Melchor-Moncada
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | | | - Enrique Aguilar-Fernández
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - Juan Carlos Sepúlveda-Arias
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| |
Collapse
|
15
|
Yamahara S, Montenegro Raudales JL, Akiyama Y, Ito M, Chimedtseren I, Arai Y, Wakita T, Hiratsuka T, Miyazawa K, Goto S, Honda M. Appropriate pore size for bone formation potential of porous collagen type I-based recombinant peptide. Regen Ther 2022; 21:294-306. [PMID: 36110974 PMCID: PMC9445290 DOI: 10.1016/j.reth.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/04/2022] [Indexed: 12/01/2022] Open
Abstract
Introduction In this study, we developed porous medium cross-linked recombinant collagen peptide (mRCP) with two different ranges of interconnected pore sizes, Small-mRCP (S-mRCP) with a range of 100–300 μm and Large-mRCP (L-mRCP) with a range of 200–500 μm, to compare the effect of pore size on bone regeneration in a calvarial bone defect. Methods Calvarial bone defects were created in Sprague–Dawley rats through a surgical procedure. The rats were divided into 2 groups: S-mRCP implanted group and L-mRCP implanted group. The newly formed bone volume and bone mineral density (BMD) was evaluated by micro-computed tomography (micro-CT) immediately after implantation and at 1, 2, 3, and 4 weeks after implantation. In addition, histological analyses were carried out with hematoxylin and eosin (H&E) staining at 4 weeks after implantation to measure the newly formed bone area between each group in the entire defect, as well as the central side, the two peripheral sides (right and left), the periosteal (top) side and the dura matter (bottom) side of the defect. Results Micro-CT analysis showed no significant differences in the amount of bone volume between the S-mRCP and L-mRCP implanted groups at 1, 2, 3 and 4 weeks after implantation. BMD was equivalent to that of the adjacent native calvaria bone at 4 weeks after implantation. H&E images showed that the newly formed bone area in the entire defect was significantly larger in the S-mRCP implanted group than in the L-mRCP implanted group. Furthermore, the amount of newly formed bone area in all sides of the defect was significantly more in the S-mRCP implanted group than in the L-mRCP implanted group. Conclusion These results indicate that the smaller pore size range of 100–300 μm is appropriate for mRCP in bone regeneration. This study confirmed the regenerative potential of mRCP as novel bone substitute. mRCP with 2 different interconnected pores sizes have been developed. The smaller pore size range of 100–300 μm was optimal for calvarial bone regeneration. The slower absorption rate of smaller pore size mRCP influenced its effectiveness.
Collapse
Affiliation(s)
- Shoji Yamahara
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Jorge Luis Montenegro Raudales
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Yasunori Akiyama
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Masaaki Ito
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Ichinnorov Chimedtseren
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Yoshinori Arai
- Department of Oral and Maxillofacial Radiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Taku Wakita
- Bio Science & Engineering Laboratory, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa 258-8577, Japan
| | - Takahiro Hiratsuka
- Bio Science & Engineering Laboratory, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa 258-8577, Japan
| | - Ken Miyazawa
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
| | - Shigemi Goto
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi, 464-8651, Japan
| | - Masaki Honda
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
- Corresponding author. Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, 100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan. Tel.: +81-52-751-2561; Fax.: +81-52-752-5988
| |
Collapse
|
16
|
Composite silk fibroin hydrogel scaffolds for cartilage tissue regeneration. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Liu X, Li Y, Sun Y, Chen B, Du W, Li Y, Gu N. Construction of functional magnetic scaffold with temperature control switch for long-distance vascular injury. Biomaterials 2022; 290:121862. [DOI: 10.1016/j.biomaterials.2022.121862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/01/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022]
|
18
|
Caldiroli A, Pederzani E, Pezzotta M, Azzollini N, Fiori S, Tironi M, Rizzo P, Sangalli F, Figliuzzi M, Fiore GB, Remuzzi A, Riboldi SA, Soncini M, Redaelli A. Hybrid fibroin/polyurethane small-diameter vascular grafts: from fabrication to in vivo preliminary assessment. Biomed Mater 2022; 17. [PMID: 35944550 DOI: 10.1088/1748-605x/ac885a] [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: 11/09/2021] [Accepted: 08/08/2022] [Indexed: 11/12/2022]
Abstract
To address the need of alternatives to autologous vessels for small-calibre vascular applications (e.g. cardiac surgery), a hybrid semi-degradable material composed of silk fibroin and polyurethane (Silkothane®) was herein used to fabricate very small-calibre grafts (innner diameter = 1.5 mm) via electrospinning. Hybrid grafts were in vitro characterized in terms of morphology and mechanical behaviour, and compared to similar grafts of pure silk fibroin. Similarly, two native vessels from a rodent model (abdominal aorta and vena cava) were harvested and characterized. Preliminary implants were performed on Lewis rats to confirm the suitability of Silkothane® grafts for small-calibre applications, specifically as aortic insertion and femoral shunt. The manufacturing process generated pliable grafts consisting of a randomized fibrous mesh and exhibiting similar geometrical features to rat aortas. Both Silkothane® and pure silk fibroin grafts showed radial compliances in the range from 1.37 ± 0.86 to 1.88 ± 1.01 % 10-2 mmHg-1, lower than that of native vessels. The Silkothane® small-calibre devices were also implanted in rats demonstrating to be adequate for vascular applications; all the treated rats survived the surgery for 3 months after implantation, and 16 rats out of 17 (94%) still showed blood flow inside the graft at sacrifice. The obtained results lay the basis for a deeper investigation of the interaction between the Silktohane® graft and the implant site, which may deal with further analysis on the potentialities in terms of degradability and tissue formation, on longer time-points.
Collapse
Affiliation(s)
- Alice Caldiroli
- Bioengineering Laboratories Srl, Via Vivaldi 32/A, Cantù, 22063, ITALY
| | - Elia Pederzani
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Giuseppe Ponzio 34, Milano, Lombardia, 20133, ITALY
| | - Marco Pezzotta
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Giuseppe Ponzio 34, Milano, Lombardia, 20133, ITALY
| | - Nadia Azzollini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS , Via Stezzano 87, Bergamo, 24126, ITALY
| | - Sonia Fiori
- , Istituto di ricerche farmacologiche mario negri IRCCS, via Stezzano 87, Bergamo, Lombardia, 24126, ITALY
| | - Matteo Tironi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS , Via Stezzano 87, Bergamo, 24126, ITALY
| | - Paola Rizzo
- IRCCS Istituto di Ricerche Farmacologiche Mario Negri Centro Anna Maria Astori, via Stezzano 87, Bergamo, 24126, ITALY
| | - Fabio Sangalli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Stezzano 87, Bergamo, 24126, ITALY
| | - Marina Figliuzzi
- Bioengineering, Istituto di ricerche farmacologiche mario negri IRCCS, via Stezzano 87, Bergamo, Bergamo, 24126, ITALY
| | - Gianfranco Beniamino Fiore
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Giuseppe Ponzio, 34, Milano, Lombardia, 20133, ITALY
| | - Andrea Remuzzi
- Università degli Studi di Bergamo, Via Pignolo 123, Bergamo, Lombardia, 24121, ITALY
| | | | - Monica Soncini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza L Da Vinci ,32, Milano, 20133, ITALY
| | - Alberto Redaelli
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Giuseppe Ponzio 34, Milano, 20133, ITALY
| |
Collapse
|
19
|
Liu X, Wang N, Liu X, Deng R, Kang R, Xie L. Vascular Repair by Grafting Based on Magnetic Nanoparticles. Pharmaceutics 2022; 14:pharmaceutics14071433. [PMID: 35890328 PMCID: PMC9320478 DOI: 10.3390/pharmaceutics14071433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Magnetic nanoparticles (MNPs) have attracted much attention in the past few decades because of their unique magnetic responsiveness. Especially in the diagnosis and treatment of diseases, they are mostly involved in non-invasive ways and have achieved good results. The magnetic responsiveness of MNPs is strictly controlled by the size, crystallinity, uniformity, and surface properties of the synthesized particles. In this review, we summarized the classification of MNPs and their application in vascular repair. MNPs mainly use their unique magnetic properties to participate in vascular repair, including magnetic stimulation, magnetic drive, magnetic resonance imaging, magnetic hyperthermia, magnetic assembly scaffolds, and magnetic targeted drug delivery, which can significantly affect scaffold performance, cell behavior, factor secretion, drug release, etc. Although there are still challenges in the large-scale clinical application of MNPs, its good non-invasive way to participate in vascular repair and the establishment of a continuous detection process is still the future development direction.
Collapse
Affiliation(s)
| | | | | | | | | | - Lin Xie
- Correspondence: (R.K.); (L.X.)
| |
Collapse
|
20
|
Ganjali F, Eivazzadeh-Keihan R, Aghamirza Moghim Aliabadi H, Maleki A, Pouri S, Ahangari Cohan R, Hashemi SM, Mahdavi M. Biocompatibility and Antimicrobial Investigation of Agar-Tannic Acid Hydrogel Reinforced with Silk Fibroin and Zinc Manganese Oxide Magnetic Microparticles. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-022-02410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Gomes JM, Silva SS, Fernandes EM, Lobo FC, Martín-Pastor M, Taboada P, Reis RL. Silk fibroin/cholinium gallate-based architectures as therapeutic tools. Acta Biomater 2022; 147:168-184. [PMID: 35580828 DOI: 10.1016/j.actbio.2022.05.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
The combination of natural resources with biologically active biocompatible ionic liquids (Bio-IL) is presented as a combinatorial approach for developing tools to manage inflammatory diseases. Innovative biomedical solutions were constructed combining silk fibroin (SF) and Ch[Gallate], a Bio-IL with antioxidant and anti-inflammatory features, as freeze-dried 3D-based sponges. An evaluation of the effect of the Ch[Gallate] concentration (≤3% w/v) on the SF/Ch[Gallate] sponges was studied. Structural changes observed on the sponges revealed that the Ch[Gallate] presence positively affected the β-sheet formation while not influencing the silk native structure, which was suggested by the FTIR and solid-state NMR results, respectively. Also, it was possible to modulate their mechanical properties, antioxidant activity and stability/degradation in an aqueous environment, by changing the Ch[Gallate] concentration. The architectures showed high water uptake ability and a weight loss that follows the controlled Ch[Gallate] release rate studied for 7 days. Furthermore, the sponges supported human adipose stem cells growth and proliferation, up to 7 days. TNF-α, IL-6 (pro-inflammatory) and IL-10 (anti-inflammatory) release quantification from a human monocyte cell line revealed a decrease in the pro-inflammatory cytokines concentrations in samples containing Ch[Gallate]. These outcomes encourage the use of the developed architectures as tissue engineering solutions, potentially targeting inflammation processes. STATEMENT OF SIGNIFICANCE: Combining natural resources with active biocompatible ionic liquids (Bio-IL) is herein presented as a combinatorial approach for the development of tools to manage inflammatory diseases. We propose using silk fibroin (SF), a natural protein, with cholinium gallate, a Bio-IL, with antioxidant and anti-inflammatory properties, to construct 3D-porous sponges through a sustainable methodology. The morphological features, swelling, and stability of the architectures were controlled by Bio-IL content in the matrices. The sponges were able to support human adipose stem cells growth and proliferation, and their therapeutic effect was proved by the blockage of TNF-α from activated and differentiated THP-1 monocytes. We believe that these bio-friendly and bioactive SF/Bio-IL-based sponges are effective for targeting pathologies with associated inflammatory processes.
Collapse
|
22
|
Man K, Joukhdar H, Manz XD, Brunet MY, Jiang LH, Rnjak-Kovacina J, Yang XB. Bone tissue engineering using 3D silk scaffolds and human dental pulp stromal cells epigenetic reprogrammed with the selective histone deacetylase inhibitor MI192. Cell Tissue Res 2022; 388:565-581. [PMID: 35362831 PMCID: PMC9110470 DOI: 10.1007/s00441-022-03613-0] [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: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022]
Abstract
Epigenetics plays a critical role in regulating mesenchymal stem cells’ (MSCs) fate for tissue repair and regeneration. There is increasing evidence that the inhibition of histone deacetylase (HDAC) isoform 3 can enhance MSC osteogenesis. This study investigated the potential of using a selective HDAC2 and 3 inhibitor, MI192, to promote human dental pulp stromal cells (hDPSCs) bone-like tissue formation in vitro and in vivo within porous Bombyx Mori silk scaffolds. Both 2 and 5 wt% silk scaffolds were fabricated and characterised. The 5 wt% scaffolds possess thicker internal lamellae, reduced scaffold swelling and degradation rates, whilst increased compressive modulus in comparison to the 2 wt% silk scaffold. MI192 pre-treatment of hDPSCs on 5 wt% silk scaffold significantly enhanced hDPSCs alkaline phosphatase activity (ALP). The expression of osteoblast-related genes (RUNX2, ALP, Col1a, OCN) was significantly upregulated in the MI192 pre-treated cells. Histological analysis confirmed that the MI192 pre-treated hDPSCs-silk scaffold constructs promoted bone extracellular matrix (ALP, Col1a, OCN) deposition and mineralisation compared to the untreated group. Following 6 weeks of subcutaneous implantation in nude mice, the MI192 pre-treated hDPSCs-silk scaffold constructs enhanced the vascularisation and extracellular matrix mineralisation compared to untreated control. In conclusion, these findings demonstrate the potential of using epigenetic reprogramming and silk scaffolds to promote hDPSCs bone formation efficacy, which provides evidence for clinical translation of this technology for bone augmentation.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.,School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Habib Joukhdar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xue D Manz
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia.,Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xuebin B Yang
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.
| |
Collapse
|
23
|
Lujerdean C, Baci GM, Cucu AA, Dezmirean DS. The Contribution of Silk Fibroin in Biomedical Engineering. INSECTS 2022; 13:286. [PMID: 35323584 PMCID: PMC8950689 DOI: 10.3390/insects13030286] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023]
Abstract
Silk fibroin (SF) is a natural protein (biopolymer) extracted from the cocoons of Bombyx mori L. (silkworm). It has many properties of interest in the field of biotechnology, the most important being biodegradability, biocompatibility and robust mechanical strength with high tensile strength. SF is usually dissolved in water-based solvents and can be easily reconstructed into a variety of material formats, including films, mats, hydrogels, and sponges, by various fabrication techniques (spin coating, electrospinning, freeze-drying, and physical or chemical crosslinking). Furthermore, SF is a feasible material used in many biomedical applications, including tissue engineering (3D scaffolds, wounds dressing), cancer therapy (mimicking the tumor microenvironment), controlled drug delivery (SF-based complexes), and bone, eye and skin regeneration. In this review, we describe the structure, composition, general properties, and structure-properties relationship of SF. In addition, the main methods used for ecological extraction and processing of SF that make it a green material are discussed. Lastly, technological advances in the use of SF-based materials are addressed, especially in healthcare applications such as tissue engineering and cancer therapeutics.
Collapse
Affiliation(s)
- Cristian Lujerdean
- Faculty of Animal Science and Biotechnology, University of Animal Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania; (A.-A.C.); (D.S.D.)
| | - Gabriela-Maria Baci
- Faculty of Animal Science and Biotechnology, University of Animal Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania; (A.-A.C.); (D.S.D.)
| | | | | |
Collapse
|
24
|
Jing T, Yi Liu, Xu L, Chen C, Liu F. The incorporation of β-tricalcium phosphate nanoparticles within silk fibroin composite scaffolds for enhanced bone regeneration: An in vitro and in vivo study. J Biomater Appl 2022; 36:1567-1578. [PMID: 35135370 DOI: 10.1177/08853282211065621] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To investigate the osteogenesis of β-tricalcium phosphate nanoparticles-incorporated silk fibroin (SF/β-TCP) composite scaffolds, SF-based scaffolds with different β-TCP proportion (2/1, 1/1, and 1/2) were fabricated by freeze-drying technology in the present study. Structural and physicochemical properties of SF-based scaffolds were evaluated by using scanning electron microscope, X-ray diffraction, Fourier transformed infrared spectroscopy (ATR-FTIR) and transmission electron microscope. Biocompatibility and osteogenesis of SF/β-TCP scaffolds were investigated by using bone marrow mesenchymal stem cells (BMSCs). Eight New Zealand rabbits were selected, while four 8-mm-diameter calvarial defects were created in each rabbit to place SF/β-TCP scaffolds. The harvested specimens at 4 and 12 weeks were used to evaluate the bone forming ability by micro-CT and histological examination. The results suggested incorporation of β-TCP displayed flake-like pore morphology with proper pore sizes. With the increasing proportion of β-TCP, composite scaffolds exhibited higher compressive strength, lower swelling ratio and degradation rate, as well as enhanced biomineralization capacity. Alkaline phosphatase activity and collagen type I expression levels of BMSCs were significantly increased in the presence of β-TCP nanoparticles. All composite scaffolds with different β-TCP proportion had good bone formation ability at 12 weeks. Among them, SF/β-TCP (1/2) scaffold exhibited the favorable osteogenesis capability which had great potential for applications in bone regeneration.
Collapse
Affiliation(s)
- Tienan Jing
- Department of Oral Mucosa Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yi Liu
- Department of Orthodontics, School and Hospital of Stomatology, 162778China Medical University, Shenyang, China
| | - Li Xu
- Department of Laboratory Medicine, First Hospital of China Medical University, Shenyang, China
| | - Cen Chen
- College of Life College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou China
| | - Fan Liu
- Department of Orthodontics, School and Hospital of Stomatology, 162778China Medical University, Shenyang, China
| |
Collapse
|
25
|
Biodegradable polymeric conduits: Platform materials for guided nerve regeneration and vascular tissue engineering. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
26
|
Fitzpatrick V, Martín-Moldes Z, Deck A, Torres-Sanchez R, Valat A, Cairns D, Li C, Kaplan DL. Functionalized 3D-printed silk-hydroxyapatite scaffolds for enhanced bone regeneration with innervation and vascularization. Biomaterials 2021; 276:120995. [PMID: 34256231 PMCID: PMC8408341 DOI: 10.1016/j.biomaterials.2021.120995] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/20/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Our goal was to generate functionalized 3D-printed scaffolds for bone regeneration using silk-hydroxyapatite bone cements and osteoinductive, proangiogenic and neurotrophic growth factors or morphogens for accelerated bone formation. 3D printing was utilized to generate macroporous scaffolds with controlled geometries and architectures that promote osseointegration. We build on the knowledge that the osteoinductive factor Bone Morphogenetic Protein-2 (BMP2) can also positively impact vascularization, Vascular Endothelial Growth Factor (VEGF) can impact osteoblastic differentiation, and that Neural Growth Factor (NGF)-mediated signaling can influence bone regeneration. We assessed functions on the 3D printed construct via the osteogenic differentiation of human mesenchymal stem cells; migration and proliferation of human umbilical vein endothelial cells; and proliferation of human induced neural stem cells. The scaffolds provided mechanical properties suitable for bone and the materials were cytocompatible, osteoconductive and maintained the activity of the morphogens and cytokines. Synergistic outcomes between BMP-2, VEGF and NGF in terms of osteoblastic differentiation in vitro were identified, based on the upregulation of genes associated with osteoblastic differentiation (Runt-related transcription factor-2, Osteopontin, Bone Sialoprotein). Additional studies will be required to assess these scaffold designs in vivo. These results are expected to have a strong impact in bone regeneration in dental, oral and maxillofacial surgery.
Collapse
Affiliation(s)
- Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zaira Martín-Moldes
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Anna Deck
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | - Anne Valat
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dana Cairns
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Chunmei Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
27
|
Kambe Y, Yamaoka T. Initial immune response to a FRET-based MMP sensor-immobilized silk fibroin hydrogel in vivo. Acta Biomater 2021; 130:199-210. [PMID: 34087439 DOI: 10.1016/j.actbio.2021.05.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/26/2022]
Abstract
To investigate the initial immune response to biodegradable silk fibroin (SF) hydrogels in vivo, a Förster/fluorescence resonance energy transfer (FRET)-based sensor was developed to detect matrix metalloproteinase (MMP) activity (FRET-MMPS) and immobilized to SF hydrogel. FRET-MMPS immobilized to SF hydrogel in vitro displayed intra-molecular FRET more than inter-molecular FRET, and MMP activity was detected through a decrease in FRET signal intensity. Then, the SF hydrogel modified with FRET-MMPS was implanted into mice subcutaneously, and it was observed that the FRET signal intensity decreased significantly soon (< 3 h) after implantation. Although the intensity exhibited a sharp decrease toward 24 h post-implantation, histological evaluation proved that bulk-level hydrogel degradation, such as breakdown, was mainly caused by macrophages and foreign body giant cells on a timescale of weeks. These results indicated that, immediately upon implantation, active MMPs reached the SF hydrogel and began cleaving SF networks, which might result in the loosening of the networks and then enabled immune cells, such as macrophages, to start the bulk-level hydrogel degradation. The sensor clarified the initial immune response to SF hydrogels and will provide clues for designing the biodegradation behaviors of scaffolds for regenerative medicine. STATEMENT OF SIGNIFICANCE: Silk fibroin (SF) materials are degraded gradually by the immune response. Immune cells, such as macrophages, break down implanted SF materials on a timescale of weeks or months, but the initial (< 24 h) immune response to SF materials remains unclear. In this study, SF hydrogels modified with Förster/fluorescence resonance energy transfer (FRET)-based matrix metalloproteinase (MMP) sensors were implanted in mice and within 3 h post-implantation, the SF hydrogels were degraded by MMPs. Although this molecular-level biodegradation was not correlated with the hydrogel breakdown, the MMPs were likely to loosen the SF networks to enable immune cells to infiltrate and degrade the hydrogel. This is the first study to unveil the initial stage of immune response to biomaterials.
Collapse
|
28
|
Jameson JF, Pacheco MO, Butler JE, Stoppel WL. Estimating Kinetic Rate Parameters for Enzymatic Degradation of Lyophilized Silk Fibroin Sponges. Front Bioeng Biotechnol 2021; 9:664306. [PMID: 34295878 PMCID: PMC8290342 DOI: 10.3389/fbioe.2021.664306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/31/2021] [Indexed: 11/25/2022] Open
Abstract
Sponge-like biomaterials formed from silk fibroin are promising as degradable materials in clinical applications due to their controllable breakdown into simple amino acids or small peptides in vivo. Silk fibroin, isolated from Bombyx mori silkworm cocoons, can be used to form sponge-like materials with a variety of tunable parameters including the elastic modulus, porosity and pore size, and level of nanocrystalline domains. These parameters can be independently tuned during formulation resulting in a wide parameter space and set of final materials. Determining the mechanism and rate constants for biomaterial degradation of these tunable silk materials would allow scientists to evaluate and predict the biomaterial performance for the large array of tissue engineering applications and patient ailments a priori. We first measured in vitro degradation rates of silk sponges using common protein-degrading enzymes such as Proteinase K and Protease XIV. The concentration of the enzyme in solution was varied (1, 0.1, 0.01 U/mL) along with one silk sponge formulation parameter: the level of crystallinity within the sponge. Additionally, two experimental degradation methods were evaluated, termed continuous and discrete degradation methods. Silk concentration, polymer chain length and scaffold pore size were held constant during experimentation and kinetic parameter estimation. Experimentally, we observed that the enzyme itself, enzyme concentration within the bulk solution, and the sponge fabrication water annealing time were the major experimental parameters dictating silk sponge degradation in our experimental design. We fit the experimental data to two models, a Michaelis-Menten kinetic model and a modified first order kinetic model. Weighted, non-linear least squares analysis was used to determine the parameters from the data sets and Monte-Carlo simulations were utilized to obtain estimates of the error. We found that modified first order reaction kinetics fit the time-dependent degradation of lyophilized silk sponges and we obtained first order-like rate constants. These results represent the first investigations into determining kinetic parameters to predict lyophilized silk sponge degradation rates and can be a tool for future mathematical representations of silk biomaterial degradation.
Collapse
Affiliation(s)
- Julie F Jameson
- Department of Chemical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States
| | - Marisa O Pacheco
- Department of Chemical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States
| | - Jason E Butler
- Department of Chemical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States
| | - Whitney L Stoppel
- Department of Chemical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
29
|
Tissue Engineering 3D Porous Scaffolds Prepared from Electrospun Recombinant Human Collagen (RHC) Polypeptides/Chitosan Nanofibers. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11115096] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Electrospinning, the only method that can continuously produce nanofibers, has been widely used to prepare nanofibers for tissue engineering applications. However, electrospinning is not suitable for preparing clinically relevant three-dimensional (3D) nanofibrous scaffolds with hierarchical pore structures. In this study, recombinant human collagen (RHC)/chitosan nanofibers prepared by electrospinning were combined with porous scaffolds produced by freeze drying to fabricate 3D nanofibrous scaffolds. These scaffolds exhibited high porosity (over 80%) and an interconnected porous structure (ranging from sub-micrometers to 200 μm) covered with nanofibers. As confirmed by the characterization results, these scaffolds showed good swelling ability, stability, and adequate mechanical strength, making it possible to use the 3D nanofibrous scaffolds in various tissue engineering applications. In addition, after seven days of cell culturing, NIH 3T3 was infiltrated into the scaffolds while maintaining its morphology and with superior proliferation and viability. These results indicated that the 3D nanofibrous scaffolds hold great promise for tissue engineering applications.
Collapse
|
30
|
Watchararot T, Prasongchean W, Thongnuek P. Angiogenic property of silk fibroin scaffolds with adipose-derived stem cells on chick chorioallantoic membrane. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201618. [PMID: 33959331 PMCID: PMC8074929 DOI: 10.1098/rsos.201618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
Angiogenesis is a crucial step in tissue regeneration and repair. Biomaterials that allow or promote angiogenesis are thus beneficial. In this study, angiogenic properties of salt-leached silk fibroin (SF) scaffolds seeded with human adipose stem cells (hADSCs) were studied using chick chorioallantoic membrane (CAM) as a model. The hADSC-seeded SF scaffolds (SF-hADSC) with the porosity of 77.34 ± 6.96% and the pore diameter of 513.95 ± 4.99 µm were implanted on the CAM of chick embryos that were on an embryonic day 8 (E8) of development. The SF-hADSC scaffolds induced a spoke-wheel pattern of capillary network indicative of angiogenesis, which was evident since E11. Moreover, the ingrowth of blood vessels into the scaffolds was seen in histological sections. The unseeded scaffolds induced the same extent of angiogenesis later on E14. By contrast, the control group could not induce the same extent of angiogenesis. In vitro cytotoxicity tests and in vivo angioirritative study reaffirmed the biocompatibility of the scaffolds. This work highlighted that the biocompatible SF-hADSC scaffolds accelerate angiogenesis, and hence they can be a promising biomaterial for the regeneration of tissues that require angiogenesis.
Collapse
Affiliation(s)
- Tanapong Watchararot
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Weerapong Prasongchean
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Peerapat Thongnuek
- Biomedical Engineering Program, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
- Biomaterial Engineering for Medical and Health Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
- Biomedical Engineering Research Center, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
31
|
Wang HY, Zhang YQ, Wei ZG. Dissolution and processing of silk fibroin for materials science. Crit Rev Biotechnol 2021; 41:406-424. [PMID: 33749463 DOI: 10.1080/07388551.2020.1853030] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In recent decades, silk fibroin (SF) from silkworm Bombyx mori has been extensively researched and applied in several fields, including: cosmetics, biomedicine and biomaterials. The dissolution and regeneration of SF fibers is the key and prerequisite step for the application of silk protein-based materials. Various solvents and dissolving systems have been reported to dissolve SF fibers. However, the dissolution process directly affects the characteristics of SF and particularly impacts the mechanical properties of the resulting silk biomaterials in subsequent processing. The purpose of this review is to summarize the common solvents, the dissolution methods for silk protein, the properties of the resulting SF protein. The suitable use of SF dissolved in the corresponding solvent was also briefly introduced. Recent applications of SF in various biomaterials are also discussed.
Collapse
Affiliation(s)
- Hai-Yan Wang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yu-Qing Zhang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Zheng-Guo Wei
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
32
|
Biggi S, Bassani GA, Vincoli V, Peroni D, Bonaldo V, Biagiotti M, Belli R, Alessandrino A, Biasini E, Freddi G. Characterization of Physical, Mechanical, and Biological Properties of SilkBridge Nerve Conduit after Enzymatic Hydrolysis. ACS APPLIED BIO MATERIALS 2020; 3:8361-8374. [PMID: 35019608 DOI: 10.1021/acsabm.0c00613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The in vitro degradation profile and the cytotoxicity of the degradation products of a silk fibroin (SF)-based nerve conduit (SilkBridge), with a complex three-layered wall architecture comprising both native and regenerated (electrospun) fibers, are reported. The bacterial protease type XIV from Streptomyces griseus was used as a hydrolytic agent at three different enzyme/substrate ratios (1:8, 1:80, and 1:800 w/w) to account for the different susceptibility to degradation of the native and regenerated components. The incubation time was extended up to 91 days. At fixed time points, the remaining device, the insoluble debris, and the incubation buffers containing soluble degradation products were separated and analyzed. The electrospun fibers forming the inner and outer layers of the conduit wall were almost completely degraded within 10 days of incubation at an enzyme/substrate ratio of 1:80 w/w. The progression of degradation was highlighted by the emergence of zones of erosion and discontinuity along the electrospun fibers, weakening of the electrospun layers, and decrease in resistance to compressive stress. Native SF microfibers forming the middle layer of the conduit wall displayed a higher resistance to enzymatic degradation. When incubated at an enzyme/substrate ratio of 1:8 w/w, the weight decreased gradually over the incubation time as a consequence of fiber erosion and fragmentation. Analogously, the tensile properties markedly decreased. Both spectroscopic and thermal analyses confirmed the gradual increase in the crystalline character of the fibers. The incubation buffers containing the soluble degradation products were subjected to cytotoxicity testing with human HEK293 cells and mouse neuroblastoma N2a cells. No detrimental effects on cell viability were observed, suggesting that the degradation products do not retain any toxic property. Finally, the mass spectrometry analysis of degradation products showed that the SF polypeptides recovered in the incubation buffers were representative of the aminoacidic sequence of the fibroin light chain and of the highly repetitive fibroin heavy chain, indicating that virtually the entire sequence of the fibroin protein constituent of SilkBridge was degraded.
Collapse
Affiliation(s)
- Silvia Biggi
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | | | | | - Daniele Peroni
- Mass Spectrometry (MS) Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | - Valerio Bonaldo
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | - Marco Biagiotti
- Silk Biomaterials Srl, Via Cavour 2, 22074 Lomazzo, Co, Italy
| | - Romina Belli
- Mass Spectrometry (MS) Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | | | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | - Giuliano Freddi
- Silk Biomaterials Srl, Via Cavour 2, 22074 Lomazzo, Co, Italy
| |
Collapse
|
33
|
Liu X, Chen B, Li Y, Kong Y, Gao M, Zhang LZ, Gu N. Development of an electrospun polycaprolactone/silk scaffold for potential vascular tissue engineering applications. J BIOACT COMPAT POL 2020. [DOI: 10.1177/0883911520973244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Long-distance (⩾10 mm) arterial vascular defect injury was a massive challenge affecting human health. Compared with autologous transplantation, tissue-engineered scaffolds such as biocompatible silk fibroin (SF) scaffolds have been developed because they exhibit equivalent functional repair effects without adverse reactions. However, its mechanical strength and structural stability needed to be further improved to match the longer repair cycle of blood vessels while maintaining the original biological safety. Hence, we designed and prepared SF and hydrophobic polycaprolactone (PCL) composite microfibers by an improving electrospinning method. It was found that when the weight ratio of PCL to SF was 1: 1, a microfiber scaffold with high strength (6.16 N) and minimum degradability can be obtained. More importantly, compared with natural silk fibroin, the novel composite microfiber scaffolds can slightly inhibit cell infiltration and inflammation through co-culture with HUVECs in vitro and rabbit back transplantation in vivo. Furthermore, the fabricated scaffolds also demonstrated excellent structural stability in vivo because of the well-organized PCL doping in the structure. All these results indicated that the novel PCL/SF composite microfiber scaffolds were promising candidates for vascular tissue engineering applications.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Bioeletronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, P. R. China
| | - Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, Suzhou, Jiangsu, P. R. China
| | - Yan Li
- State Key Laboratory of Bioeletronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, P. R. China
| | - Yan Kong
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, P. R. China
| | - Ming Gao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, P. R. China
| | - Lu Zhong Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, P. R. China
| | - Ning Gu
- State Key Laboratory of Bioeletronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, P. R. China
| |
Collapse
|
34
|
Ghalei S, Mondal A, Hopkins S, Singha P, Devine R, Handa H. Silk Nanoparticles: A Natural Polymeric Platform for Nitric Oxide Delivery in Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53615-53623. [PMID: 33205962 DOI: 10.1021/acsami.0c13813] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, the preparation and characterization of nitric oxide (NO) releasing silk fibroin nanoparticles (SF NPs) are described for the first time. S-Nitroso-N-acetylpenicillamine (SNAP)-loaded SF NPs (SNAP-SF NPs) were prepared via an antisolvent/self-assembling method by adding a SNAP/ethanol solution to an aqueous SF solution and freeze-thawing. The prepared SNAP-SF NPs had a diameter ranging from 300 to 400 nm and an overall negative charge of -28.76 ± 0.73 mV. Among the different SNAP/SF ratios tested, the highest encapsulation efficiency (18.3 ± 1.3%) and loading capacity (9.1 ± 0.6%) values were attributed to the 1:1 ratio. The deconvolution of the amide I band in the FTIR spectra of SF NPs and SNAP-SF NPs showed an increase in the β-sheet content for SNAP-SF NPs, confirming the hydrophobic interactions between SNAP and silk macromolecules. SNAP-SF NPs released up to 1.31 ± 0.02 × 10-10 mol min-1 mg-1 NO over a 24 h period. Moreover, SNAP-SF NPs showed concentration-dependent antibacterial effects against methicillin-resistant Staphylococcus aureus and Escherichia coli. Furthermore, they did not elicit any marked cytotoxicity against 3T3 mouse fibroblast cells at concentrations equal to or below 2 mg/mL. Overall, these results demonstrated that SNAP-SF NPs have great potential to be used as a NO delivery platform for biomedical applications such as tissue engineering and wound healing, where synergistic properties of SF and NO are desired.
Collapse
Affiliation(s)
- Sama Ghalei
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Arnab Mondal
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Sean Hopkins
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Priyadarshini Singha
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Ryan Devine
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hitesh Handa
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
35
|
Belda Marín C, Fitzpatrick V, Kaplan DL, Landoulsi J, Guénin E, Egles C. Silk Polymers and Nanoparticles: A Powerful Combination for the Design of Versatile Biomaterials. Front Chem 2020; 8:604398. [PMID: 33335889 PMCID: PMC7736416 DOI: 10.3389/fchem.2020.604398] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/09/2020] [Indexed: 12/30/2022] Open
Abstract
Silk fibroin (SF) is a natural protein largely used in the textile industry but also in biomedicine, catalysis, and other materials applications. SF is biocompatible, biodegradable, and possesses high tensile strength. Moreover, it is a versatile compound that can be formed into different materials at the macro, micro- and nano-scales, such as nanofibers, nanoparticles, hydrogels, microspheres, and other formats. Silk can be further integrated into emerging and promising additive manufacturing techniques like bioprinting, stereolithography or digital light processing 3D printing. As such, the development of methodologies for the functionalization of silk materials provide added value. Inorganic nanoparticles (INPs) have interesting and unexpected properties differing from bulk materials. These properties include better catalysis efficiency (better surface/volume ratio and consequently decreased quantify of catalyst), antibacterial activity, fluorescence properties, and UV-radiation protection or superparamagnetic behavior depending on the metal used. Given the promising results and performance of INPs, their use in many different procedures has been growing. Therefore, combining the useful properties of silk fibroin materials with those from INPs is increasingly relevant in many applications. Two main methodologies have been used in the literature to form silk-based bionanocomposites: in situ synthesis of INPs in silk materials, or the addition of preformed INPs to silk materials. This work presents an overview of current silk nanocomposites developed by these two main methodologies. An evaluation of overall INP characteristics and their distribution within the material is presented for each approach. Finally, an outlook is provided about the potential applications of these resultant nanocomposite materials.
Collapse
Affiliation(s)
- Cristina Belda Marín
- Laboratory of Integrated Transformations of Renewable Matter (TIMR), Université de Technologie de Compiègne, ESCOM, Compiègne, France
- Laboratoire de réactivité de surface (UMR CNRS 7197), Sorbonne Université, Paris, France
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Jessem Landoulsi
- Laboratoire de réactivité de surface (UMR CNRS 7197), Sorbonne Université, Paris, France
| | - Erwann Guénin
- Laboratory of Integrated Transformations of Renewable Matter (TIMR), Université de Technologie de Compiègne, ESCOM, Compiègne, France
| | - Christophe Egles
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Compiègne, France
| |
Collapse
|
36
|
Yang L, Li X, Wu Y, Du P, Sun L, Yu Z, Song S, Yin J, Ma X, Jing C, Zhao J, Chen H, Dong Y, Zhang Q, Zhao L. Preparation of PU/Fibrin Vascular Scaffold with Good Biomechanical Properties and Evaluation of Its Performance in vitro and in vivo. Int J Nanomedicine 2020; 15:8697-8715. [PMID: 33192062 PMCID: PMC7656973 DOI: 10.2147/ijn.s274459] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/25/2020] [Indexed: 01/22/2023] Open
Abstract
PURPOSE The development of tissue-engineered blood vessels provides a new source of donors for coronary artery bypass grafting and peripheral blood vessel transplantation. Fibrin fiber has good biocompatibility and is an ideal tissue engineering vascular scaffold, but its mechanical property needs improvement. METHODS We mixed polyurethane (PU) and fibrin to prepare the PU/fibrin vascular scaffolds by using electrospinning technology in order to enhance the mechanical properties of fibrin scaffold. We investigated the morphological, mechanical strength, hydrophilicity, degradation, blood and cell compatibility of PU/fibrin (0:100), PU/fibrin (5:95), PU/fibrin (15:85) and PU/fibrin (25:75) vascular scaffolds. Based on the results in vitro, PU/fibrin (15:85) was selected for transplantation in vivo to repair vascular defects, and the extracellular matrix formation, vascular remodeling, and immune response were evaluated. RESULTS The results indicated that the fiber diameter of the PU/fibrin (15:85) scaffold was about 712nm. With the increase of PU content, the mechanical strength of the composite scaffolds increased, however, the degradation rate decreased gradually. The PU/fibrin scaffold showed good hydrophilicity and hemocompatibility. PU/fibrin (15:85) vascular scaffold could promote the adhesion and proliferation of mesenchymal stromal cells (MSCs). Quantitative RT-PCR experimental results showed that the expression of collagen, survivin and vimentin genes in PU/fibrin (15:85) was higher than that in PU/fibrin (25:75). The results in vivo indicated the mechanical properties and compliance of PU/fibrin grafts could meet clinical requirements and the proportion of thrombosis or occlusion was significantly lower. The graft showed strong vasomotor response, and the smooth muscle cells, endothelial cells, and ECM deposition of the neoartery were comparable to that of native artery after 3 months. At 3 months, the amount of macrophages in PU/fibrin grafts was significantly lower, and the secretion of pro-inflammatory and anti-inflammatory cytokines decreased. CONCLUSION PU/fibrin (15:85) vascular scaffolds had great potential to be used as small-diameter tissue engineering blood vessels.
Collapse
Affiliation(s)
- Lei Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Xiafei Li
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yiting Wu
- Xiacun Community Health Service Center, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, People’s Republic of China
| | - Pengchong Du
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Department of Cardio-Thoracic Surgery, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Lulu Sun
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhenyang Yu
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Shuang Song
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Jianshen Yin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Xianfen Ma
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Changqin Jing
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Junqiang Zhao
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Hongli Chen
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yuzhen Dong
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Qiqing Zhang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Liang Zhao
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Key Laboratory of Cardiac Structure Research, Zhengzhou Seventh People’s Hospital, Zhengzhou, People’s Republic of China
- The Central Lab, The Third People’s Hospital of Datong, Datong, People’s Republic of China
| |
Collapse
|
37
|
Magaz A, Li X, Gough JE, Blaker JJ. Graphene oxide and electroactive reduced graphene oxide-based composite fibrous scaffolds for engineering excitable nerve tissue. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111632. [PMID: 33321671 DOI: 10.1016/j.msec.2020.111632] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/02/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
This study systematically investigates the role of graphene oxide (GO) and reduced GO (rGO)/silk-based composite micro/nano-fibrous scaffolds in regulating neuronal cell behavior in vitro, given the limited comparative studies on the effects of graphene family materials on nerve regeneration. Fibrous scaffolds can mimic the architecture of the native extracellular matrix and are potential candidates for tissue engineering peripheral nerves. Silk/GO micro/nano-fibrous scaffolds were electrospun with GO loadings 1 to 10 wt.%, and optionally post-reduced in situ to explore a family of electrically conductive non-woven silk/rGO scaffolds. Conductivities up to 4 × 10-5 S cm-1 were recorded in the dry state, which increased up to 3 × 10-4 S cm-1 after hydration. Neuronoma NG108-15 cells adhered and were viable on all substrates. Enhanced metabolic activity and proliferation were observed on the GO-containing scaffolds, and these cell responses were further promoted for electroactive silk/rGO. Neurite extensions up to 100 μm were achieved by day 5, with maximum outgrowth up to ~250 μm on some of the conductive substrates. These electroactive composite fibrous scaffolds exhibit potential to enhance the neuronal cell response and could be versatile supportive substrates for neural tissue engineering applications.
Collapse
Affiliation(s)
- Adrián Magaz
- Department of Materials and Henry Royce Institute, The University of Manchester, Manchester M13 9PL, United Kingdom; Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 138634, Singapore
| | - Xu Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 138634, Singapore; Department of Chemistry, National University of Singapore, 117543 Singapore, Singapore.
| | - Julie E Gough
- Department of Materials and Henry Royce Institute, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Jonny J Blaker
- Department of Materials and Henry Royce Institute, The University of Manchester, Manchester M13 9PL, United Kingdom; Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo 0317, Norway.
| |
Collapse
|
38
|
Riboldi SA, Tozzi M, Bagardi M, Ravasio G, Cigalino G, Crippa L, Piccolo S, Nahal A, Spandri M, Catto V, Tironi M, Greco FG, Remuzzi A, Acocella F. A Novel Hybrid Silk Fibroin/Polyurethane Arteriovenous Graft for Hemodialysis: Proof-of-Concept Animal Study in an Ovine Model. Adv Healthc Mater 2020; 9:e2000794. [PMID: 32914588 DOI: 10.1002/adhm.202000794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/17/2020] [Indexed: 12/25/2022]
Abstract
To solve the problem of vascular access failure, a novel semi-degradable hybrid vascular graft, manufactured by electrospinning using silk fibroin and polyurethane (Silkothane), has been previously developed and characterized in vitro. This proof-of-concept animal study aims at evaluating the performances of Silkothane grafts in a sheep model of arteriovenous shunt, in terms of patency and short-term remodeling. Nine Silkothane grafts are implanted between the common carotid artery and the external jugular vein of nine sheep, examined by palpation three times per week, by echo-color Doppler every two weeks, and euthanized at 30, 60, and 90 days (N = 3 per group). At sacrifice, grafts are harvested and submitted for histopathology and/or scanning electron microcopy (SEM). No cases of graft-related complications are recorded. Eight of nine sheep (89%) show 100% primary unassisted patency at the respective time of sacrifice (flow rate 1.76 ± 0.61 L min-1 , one case of surgery-related thrombosis excluded). Histopathology and SEM analysis evidence signs of inflammation and pseudointima inside the graft lumen, especially at the venous anastomosis; however, endoluminal stenosis never impairs the functionality of the shunt and coverage by endothelial cells is observed. In this model, Silkothane grafts grant safety and 100% patency up to 90 days.
Collapse
Affiliation(s)
| | - Matteo Tozzi
- Department of Medicine and Surgery Università degli Studi dell'Insubria Varese 21100 Italy
| | - Mara Bagardi
- Department of Veterinary Medicine Università degli Studi di Milano Milano 20122 Italy
| | - Giuliano Ravasio
- Department of Veterinary Medicine Università degli Studi di Milano Milano 20122 Italy
| | - Giorgio Cigalino
- Centre for Clinical Veterinary Medicine and Experimental Zootechnics Università degli Studi di Milano Milano 20122 Italy
| | - Luca Crippa
- Department of Medicine and Surgery Università degli Studi di Milano Bicocca Milano 20126 Italy
| | - Solange Piccolo
- Department of Medicine and Surgery Università degli Studi dell'Insubria Varese 21100 Italy
| | - Amal Nahal
- Department of Medicine and Surgery Università degli Studi dell'Insubria Varese 21100 Italy
| | | | | | - Matteo Tironi
- Department of Bioengineering IRCCS Istituto di Ricerche Farmacologiche Mario Negri Bergamo 24126 Italy
| | | | - Andrea Remuzzi
- Department of Management, Information and Production Engineering Università degli Studi di Bergamo Bergamo 24129 Italy
| | - Fabio Acocella
- Department of Health, Animal Science and Food Safety Università degli Studi di Milano Milano 20122 Italy
| |
Collapse
|
39
|
Lüchow M, Fortuin L, Malkoch M. Modular, synthetic, thiol‐ene mediated hydrogel networks as potential scaffolds for
3D
cell cultures and tissue regeneration. JOURNAL OF POLYMER SCIENCE 2020. [DOI: 10.1002/pol.20200530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mads Lüchow
- Division of Coating Technology, Department of Fibre and Polymer Technology KTH Stockholm Sweden
| | - Lisa Fortuin
- Division of Coating Technology, Department of Fibre and Polymer Technology KTH Stockholm Sweden
| | - Michael Malkoch
- Division of Coating Technology, Department of Fibre and Polymer Technology KTH Stockholm Sweden
| |
Collapse
|
40
|
Huang Y, Fitzpatrick V, Zheng N, Cheng R, Huang H, Ghezzi C, Kaplan DL, Yang C. Self-Folding 3D Silk Biomaterial Rolls to Facilitate Axon and Bone Regeneration. Adv Healthc Mater 2020; 9:e2000530. [PMID: 32864866 PMCID: PMC7654509 DOI: 10.1002/adhm.202000530] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Biomaterial scaffold designs are needed for self-organizing features related to tissue formation while also simplifying the fabrication processes involved. Toward this goal, silk protein-based self-folding scaffolds to support 3D cell culture, while providing directional guidance and promotion of cell growth and differentiation, are reported. A simple and robust one-step self-folding approach is developed using bilayers consisting of a hydrogel and silk film in aqueous solution. The 3D silk rolls, with patterns transferred from the initially prepared 2D films, guide the directional outgrowth of neurites and also promote the osteogenic differentiation of human mesenchymal stem cells (hMSCs). The osteogenic outcomes are further supported by enhanced biomechanical performance. By utilizing this self-folding method, cocultures of neurons and hMSCs are achieved by patterning cells on silk films and then converting these materials into a 3D format with rolling, mimicking aspects of the structure of osteons and providing physiologically relevant structures to promote bone regeneration. These results demonstrate the utility of self-folded silk rolls as efficient scaffold systems for tissue regeneration, while exploiting relatively simple 2D designs programmed to form more complex 3D structures.
Collapse
Affiliation(s)
- Yimin Huang
- Department of Chemistry, Boston University, Boston, MA, 02215
| | | | - Nan Zheng
- Department of Electrical & Computer Engineering, Boston University, Boston, MA, 02215
| | - Ran Cheng
- Department of Chemistry, Boston University, Boston, MA, 02215
| | - Heyu Huang
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215
| | - Chiara Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02215
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02215
| | - Chen Yang
- Department of Chemistry, Boston University, Boston, MA, 02215
- Department of Electrical & Computer Engineering, Boston University, Boston, MA, 02215
| |
Collapse
|
41
|
Opálková Šišková A, Kozma E, Opálek A, Kroneková Z, Kleinová A, Nagy Š, Kronek J, Rydz J, Eckstein Andicsová A. Diclofenac Embedded in Silk Fibroin Fibers as a Drug Delivery System. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3580. [PMID: 32823655 PMCID: PMC7475829 DOI: 10.3390/ma13163580] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 01/04/2023]
Abstract
Silk fibroin is a biocompatible, non-toxic, mechanically robust protein, and it is commonly used and studied as a material for biomedical applications. Silk fibroin also gained particular interest as a drug carrier vehicle, and numerous silk formats have been investigated for this purpose. Herein, we have prepared electrospun nanofibers from pure silk fibroin and blended silk fibroin/casein, followed by the incorporation of an anti-inflammatory drug, diclofenac. Casein serves as an excipient in pharmaceutical products and has a positive effect on the gradual release of drugs. The characteristics of the investigated composites were estimated by scanning electron microscope, transmission electron microscope, thermogravimetric analysis, and a lifetime of diclofenac by electron paramagnetic resonance analysis. The cumulative release in vitro of diclofenac sodium salt, together with the antiproliferative effect of diclofenac sodium salt-loaded silk nanofibers against the growth of two cancer cell lines, are presented and discussed.
Collapse
Affiliation(s)
- Alena Opálková Šišková
- Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.Š.); (Z.K.); (A.K.); (J.K.)
| | - Erika Kozma
- Institute for Chemical Sciences and Technologies ‘Giulio Natta’ (SCITEC-CNR), Via A. Corti 12, 20133 Milan, Italy;
| | - Andrej Opálek
- Institute of Materials and Machine Mechanics, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.); (Š.N.)
| | - Zuzana Kroneková
- Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.Š.); (Z.K.); (A.K.); (J.K.)
| | - Angela Kleinová
- Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.Š.); (Z.K.); (A.K.); (J.K.)
| | - Štefan Nagy
- Institute of Materials and Machine Mechanics, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.); (Š.N.)
| | - Juraj Kronek
- Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.Š.); (Z.K.); (A.K.); (J.K.)
| | - Joanna Rydz
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34, M. Curie-Skłodowska St., 41-800 Zabrze, Poland;
| | - Anita Eckstein Andicsová
- Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 84541 Bratislava, Slovakia; (A.O.Š.); (Z.K.); (A.K.); (J.K.)
| |
Collapse
|
42
|
Girotti A, Gonzalez-Valdivieso J, Santos M, Martin L, Arias FJ. Functional characterization of an enzymatically degradable multi-bioactive elastin-like recombinamer. Int J Biol Macromol 2020; 164:1640-1648. [PMID: 32758602 DOI: 10.1016/j.ijbiomac.2020.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 10/23/2022]
Abstract
One of the main goals in both tissue engineering and regenerative medicine is to design innovative synthetic scaffolds that can simulate and control the communication pathways between cells and the extracellular matrix (ECM). In this context, we describe herein the characterization of protein polymer, a recombinant elastin-like recombinamer (ELR) designed for developing tissue-engineered devices for use in vascular regeneration. This ELR is composed of an elastin-like backbone that contains a fibronectin domain, which provides specific, endothelial cell adhesion, and a protease target domain directed towards specific proteases involved in ECM remodeling. We also compare the specific response of endothelial and fibroblast cells to ELR scaffolds and show that cell adhesion and spreading on this ELR is significantly higher for endothelial cells than for fibroblasts. The reactivity of this polymer and its hydrogels to specific enzymatic degradation is demonstrated in vitro. As with natural elastin, enzymatic hydrolysis of the ELR produces elastin-derived peptides, or "matrikines", which, in turn, are potentially able to regulate important cell activities.
Collapse
Affiliation(s)
- Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN University of Valladolid, 47011 Valladolid, Spain.
| | - Juan Gonzalez-Valdivieso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN University of Valladolid, 47011 Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN University of Valladolid, 47011 Valladolid, Spain
| | - Laura Martin
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN University of Valladolid, 47011 Valladolid, Spain
| | - F Javier Arias
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN University of Valladolid, 47011 Valladolid, Spain
| |
Collapse
|
43
|
Gupta P, Lorentz KL, Haskett DG, Cunnane EM, Ramaswamy AK, Weinbaum JS, Vorp DA, Mandal BB. Bioresorbable silk grafts for small diameter vascular tissue engineering applications: In vitro and in vivo functional analysis. Acta Biomater 2020; 105:146-158. [PMID: 31958596 PMCID: PMC7050402 DOI: 10.1016/j.actbio.2020.01.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 01/14/2023]
Abstract
The success of tissue-engineered vascular graft (TEVG) predominantly relies on the selection of a suitable biomaterial and graft design. Natural biopolymer silk has shown great promise for various tissue-engineering applications. This study is the first to investigate Indian endemic non-mulberry silk (Antheraea assama-AA) - which inherits naturally superior mechanical and biological traits (e.g., RGD motifs) compared to Bombyx mori-BM silk, for TEVG applications. We designed bi-layered biomimetic small diameter AA-BM silk TEVGs adopting a new fabrication methodology. The inner layer showed ideally sized (~40 µm) pores with interconnectivity to allow cellular infiltration, and an outer dense electrospun layer that confers mechanical resilience. Biodegradation of silk TEVGs into amino acids as resorbable byproducts corroborates their in vivo remodeling ability. Following our previous reports, we surgically implanted human adipose tissue-derived stromal vascular fraction (SVF) seeded silk TEVGs in Lewis rats as abdominal aortic interposition grafts for 8 weeks. Adequate suture retention strength (0.45 ± 0.1 N) without any blood seepage post-implantation substantiate the grafts' viability. AA silk-based TEVGs showed superior animal survival and graft patency compared to BM silk TEVGs. Histological analysis revealed neo-tissue formation, host cell infiltration and graft remodeling in terms of extracellular matrix turnover. Altogether, this study demonstrates promising aspects of AA silk TEVGs for vascular tissue engineering applications. STATEMENT OF SIGNIFICANCE: Clinical 'off the shelf' implementation of tissue-engineered vascular grafts (TEVGs) remains a challenge. Achieving optimal blood vessel regeneration requires the use of bioresorbable materials having suitable degradation rates while producing minimal or no toxic byproducts. Host cell recruitment and preventing acute thrombosis are other pre-requisites for successful graft remodeling. In this study, for the first time we explored the use of naturally derived Indian endemic non-mulberry Antheraea assama silk in combination with Bombyx mori silk for TEVG applications by adopting a new biomimetic approach. Our bi-layered silk TEVGs were optimally porous, mechanically resilient and biodegradable. In vivo implantation in rat aorta showed long-term patency and graft remodeling by host cell infiltration and extracellular matrix deposition corroborating their clinical feasibility.
Collapse
Affiliation(s)
- Prerak Gupta
- Department of Biosciences and Bioengineering, Indian Istitute of Technology Guwahati, Guwahati 781039, India; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Katherine L Lorentz
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Darren G Haskett
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Eoghan M Cunnane
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), Dublin D02 YN77, Ireland
| | - Aneesh K Ramaswamy
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Justin S Weinbaum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - David A Vorp
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Istitute of Technology Guwahati, Guwahati 781039, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, India.
| |
Collapse
|
44
|
Valente F, Allardyce BJ, Hepburn MS, Wijesinghe P, Redmond SL, Chen J, Kennedy BF, Rajkhowa R, Atlas MD, Wang X, Dilley RJ. Enhancing Resistance of Silk Fibroin Material to Enzymatic Degradation by Cross-Linking Both Crystalline and Amorphous Domains. ACS Biomater Sci Eng 2020; 6:2459-2468. [PMID: 33455319 DOI: 10.1021/acsbiomaterials.9b00873] [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] [Indexed: 12/26/2022]
Abstract
Silk fibroin (SF) membranes are finding widespread use as biomaterial scaffolds in a range of tissue engineering applications. The control over SF scaffold degradation kinetics is usually driven by the proportion of SF crystalline domains in the formulation, but membranes with a high β-sheet content are brittle and still contain amorphous domains, which are highly susceptible to enzymatic degradation. In this work, photo-cross-linking of SF using a ruthenium-based method, and with the addition of glycerol, was used to generate robust and flexible SF membranes for long-term tissue engineering applications requiring slow degradation of the scaffolds. The resulting mechanical properties, protein secondary structure, and degradation rate were investigated. In addition, the cytocompatibility and versatility of porous micropatterning of SF films were assessed. The photo-cross-linking reduced the enzymatic degradation of SF in vitro without interfering with the β-sheet content of the SF material, while adding glycerol to the composition grants flexibility to the membranes. By combining these methods, the membrane resistance to protease degradation was significantly enhanced compared to either method alone, and the SF mechanical properties were not impaired. We hypothesize that photo-cross-linking protects the SF amorphous regions from enzymatic degradation and complements the natural protection offered by β-sheets in the crystalline region. Overall, this approach presents broad utility in tissue engineering applications that require a long-term degradation profile and mechanical support.
Collapse
Affiliation(s)
- Filippo Valente
- Ear Science Institute Australia; Subiaco, Australia and Ear Sciences Centre, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands 6009, Australia
| | | | - Matt S Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth 6009, Australia.,Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth 6009, Australia
| | - Philip Wijesinghe
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth 6009, Australia.,Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth 6009, Australia.,SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, U.K
| | - Sharon L Redmond
- Ear Science Institute Australia; Subiaco, Australia and Ear Sciences Centre, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands 6009, Australia
| | - Jingyu Chen
- Institute for Frontier Materials, Deakin University, Geelong 3220, Australia
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth 6009, Australia.,Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Perth 6009, Australia
| | - Rangam Rajkhowa
- Institute for Frontier Materials, Deakin University, Geelong 3220, Australia
| | - Marcus D Atlas
- Ear Science Institute Australia; Subiaco, Australia and Ear Sciences Centre, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands 6009, Australia
| | - Xungai Wang
- Institute for Frontier Materials, Deakin University, Geelong 3220, Australia
| | - Rodney J Dilley
- Ear Science Institute Australia; Subiaco, Australia and Ear Sciences Centre, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands 6009, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands 6009, Australia
| |
Collapse
|
45
|
Zhang L, Gopalakrishnan S, Li K, Wang LS, Han Y, Rotello VM. Fabrication of Collagen Films with Enhanced Mechanical and Enzymatic Stability through Thermal Treatment in Fluorous Media. ACS APPLIED MATERIALS & INTERFACES 2020; 12:6590-6597. [PMID: 31935058 DOI: 10.1021/acsami.9b18256] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Collagen I (Col-I) is widely used in the fabrication of biomaterials due to its biocompatibility; however, Col-I based biomaterials are susceptible to mechanical failure during handling, which limits their applicability to biomaterials. Chemical or physical treatment can improve the mechanical properties of collagen; however, these processes can create issues of cytotoxicity or denaturation. We report here an alternative strategy to improve the stability and mechanical properties of Col-I while preserving its native structure, through thermal treatment in fluorous media. Thermal treatment of Col-I in fluorous solvent generates compact, stable films with significantly increased mechanical strength. Furthermore, the use of fluorous media significantly reduces the extent of swelling and the rate of proteolytic degradation, but it preserves the high cell biocompatibility.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Chemistry , University of Massachusetts, Amherst , Massachusetts 01003 , United States
- State-Key Laboratory for Mechanical Behavior of Materials , Xi'an Jiaotong University , Xi'an 710049 , China
| | - Sanjana Gopalakrishnan
- Department of Chemistry , University of Massachusetts, Amherst , Massachusetts 01003 , United States
| | - Kai Li
- State-Key Laboratory for Mechanical Behavior of Materials , Xi'an Jiaotong University , Xi'an 710049 , China
| | - Li-Sheng Wang
- Department of Chemistry , University of Massachusetts, Amherst , Massachusetts 01003 , United States
| | - Yong Han
- State-Key Laboratory for Mechanical Behavior of Materials , Xi'an Jiaotong University , Xi'an 710049 , China
| | - Vincent M Rotello
- Department of Chemistry , University of Massachusetts, Amherst , Massachusetts 01003 , United States
| |
Collapse
|
46
|
Umuhoza D, Yang F, Long D, Hao Z, Dai J, Zhao A. Strategies for Tuning the Biodegradation of Silk Fibroin-Based Materials for Tissue Engineering Applications. ACS Biomater Sci Eng 2020; 6:1290-1310. [DOI: 10.1021/acsbiomaterials.9b01781] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Diane Umuhoza
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
- Commercial Insect Program, Sericulture, Rwanda Agricultural Board, 5016 Kigali, Rwanda
| | - Fang Yang
- Department of Biomaterials, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Dingpei Long
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Zhanzhang Hao
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Jing Dai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Aichun Zhao
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| |
Collapse
|
47
|
Li J, Zheng H, He Y, Chen B, Liu L, Ouyang Y, Zhu C, Zhou Y, Sun J, Hu Z, Wang B. Ultrasensitive Electrochemical Immunosensor Reveals the Existence of Silk Products on the Maritime Silk Road. ACS Sens 2019; 4:3203-3209. [PMID: 31773952 DOI: 10.1021/acssensors.9b01638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The Maritime Silk Road was the major trade route between eastern and western civilizations in the Middle Ages. However, hardly any silk products have been found along the transoceanic trade route. Thus, the extrasensitive detection of silk relic traces has tremendous importance in research regarding the Maritime Silk Road. In this study, an electrochemical immunosensor based on a tailored monoclonal antibody and gold nanoparticles using the layer-by-layer self-assembly method was devised. The fabricated immunosensor demonstrated preeminent performance in the analysis of silk fibroin, with a linear detection range of 0.01-100 ng mL-1 and a detection limit of 3.8 pg mL-1. In particular, the performance of the immunosensor was excellent in the analysis of ancient silk samples, especially in the qualitative and quantitative detection of soil samples extracted from Nanhai No. 1 shipwreck archeological sites. The proposed electrochemical immunosensor proves the existence of silk products on the Maritime Silk Road and demonstrates enormous potential for studying the formation and development of the ancient transoceanic trading route.
Collapse
Affiliation(s)
| | - Hailing Zheng
- Key Scientific Research Base of Textile Conservation, State Administration for Cultural Heritage, China National Silk Museum, Hangzhou 310002, China
| | | | | | | | | | | | - Yang Zhou
- Key Scientific Research Base of Textile Conservation, State Administration for Cultural Heritage, China National Silk Museum, Hangzhou 310002, China
| | - Jian Sun
- Conservation Center of Underwater Cultural Heritage, National Cultural Heritage Administration, Beijing 100192, China
| | | | | |
Collapse
|
48
|
Zou S, Wang X, Fan S, Zhang J, Shao H, Zhang Y. Fabrication and characterization of regenerated Antheraea pernyi silk fibroin scaffolds for Schwann cell culturing. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.04.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
49
|
Eswaramoorthy SD, Dhiman N, Korra G, Oranges CM, Schaefer DJ, Rath SN, Madduri S. Isogenic-induced endothelial cells enhance osteogenic differentiation of mesenchymal stem cells on silk fibroin scaffold. Regen Med 2019; 14:647-661. [DOI: 10.2217/rme-2018-0166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: We investigated the role of induced endothelial cells (iECs) in mesenchymal stem cells (MSCs)/iECs co-culture and assessed their osteogenic ability on silk fibroin nanofiber scaffolds. Methods: The osteogenic differentiation was assessed by the ALP assay, calcium assay and gene expression studies. Results: The osteogenic differentiation of the iECs co-cultures was found to be higher than the MSCs group and proximal to endothelial cells (ECs) co-cultures. Furthermore, the usage of isogenic iECs for co-culture increased the osteogenic and endothelial gene expression. Conclusion: These findings suggest that iECs mimic endothelial cells when co-cultured with MSCs and that one MSCs source can be used to give rise to both MSCs and iECs. The isogenic MSCs/iECs co-culture provides a new option for bone tissue engineering applications.
Collapse
Affiliation(s)
- Sindhuja D Eswaramoorthy
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, 502285 Telangana, India
| | - Nandini Dhiman
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, 502285 Telangana, India
| | - Gayathri Korra
- Sri Sai Krishna Multi Specialty Hospital, Department of Obstetrics and Gynecology, Sangareddy 502001, Medak, Telangana, India
| | - Carlo M Oranges
- Department of Plastic, Reconstructive, Aesthetic & Hand Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic & Hand Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Subha N Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, 502285 Telangana, India
| | - Srinivas Madduri
- Department of Plastic, Reconstructive, Aesthetic & Hand Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, CH-4123 Allschwil, Switzerland
| |
Collapse
|
50
|
Flora T, González de Torre I, Alonso M, Rodríguez-Cabello JC. Use of proteolytic sequences with different cleavage kinetics as a way to generate hydrogels with preprogrammed cell-infiltration patterns imparted over their given 3D spatial structure. Biofabrication 2019; 11:035008. [DOI: 10.1088/1758-5090/ab10a5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|