1
|
Wei X, Lei L, Luo L, Zhou Y, Zheng Z, Chen W. Advances in osteoimmunomodulation of biomaterials after intrabone implantation: focus on surface hydrophilicity. J Mater Chem B 2024; 12:11089-11104. [PMID: 39387541 DOI: 10.1039/d4tb01907e] [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: 10/15/2024]
Abstract
Biomaterials intended for intrabone implantation are extensively utilized in orthopedic and dental applications. Their surface properties, particularly hydrophilicity, significantly influence the biological interactions surrounding the implant, ultimately determining the implant's in vivo fate. Recently, the role of osteoimmunomodulation in these implantable biomaterials has been recognized for its importance in regulating biomaterial-mediated osteogenesis. Consequently, it is imperative to elucidate the correlation between hydrophilicity and the immune response for the development of osteoimmunomodulatory implants. Herein, this review highlights recent advances in osteoimmunomodulation of biomaterials after intrabone implantation from a novel perspective-surface hydrophilicity, and summarizes the series of immune reactions and subsequent bone remodeling that occur in response to hydrophilic implants, focusing on protein adsorption, the behaviors of major immune cells, and osteoimmunomodulation-enhanced angiogenesis and osteogenesis. Hydrophilic biomaterials have the capacity to alter the surrounding immune microenvironment and accelerate the process of material-tissue bonding, thereby facilitating the successful integration of biomaterials with tissue. Collectively, the authors hope that this article provides strategies for modulating hydrophilicity to achieve osteoimmunomodulatory performance and further promotes the development of novel implantable biomaterials for orthopedic and dental applications.
Collapse
Affiliation(s)
- Xinpeng Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Linshan Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Ying Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zheng Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
2
|
Pazhouhnia Z, Noori A, Farzin A, Khoshmaram K, Hoseinpour M, Ai J, Ebrahimi M, Lotfibakhshaiesh N. 3D-bioprinted GelMA/gelatin/amniotic membrane extract (AME) scaffold loaded with keratinocytes, fibroblasts, and endothelial cells for skin tissue engineering. Sci Rep 2024; 14:12670. [PMID: 38830883 PMCID: PMC11148016 DOI: 10.1038/s41598-024-62926-y] [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: 02/18/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
Gelatin-methacryloyl (GelMA) is a highly adaptable biomaterial extensively utilized in skin regeneration applications. However, it is frequently imperative to enhance its physical and biological qualities by including supplementary substances in its composition. The purpose of this study was to fabricate and characterize a bi-layered GelMA-gelatin scaffold using 3D bioprinting. The upper section of the scaffold was encompassed with keratinocytes to simulate the epidermis, while the lower section included fibroblasts and HUVEC cells to mimic the dermis. A further step involved the addition of amniotic membrane extract (AME) to the scaffold in order to promote angiogenesis. The incorporation of gelatin into GelMA was found to enhance its stability and mechanical qualities. While the Alamar blue test demonstrated that a high concentration of GelMA (20%) resulted in a decrease in cell viability, the live/dead cell staining revealed that incorporation of AME increased the quantity of viable HUVECs. Further, gelatin upregulated the expression of KRT10 in keratinocytes and VIM in fibroblasts. Additionally, the histological staining results demonstrated the formation of well-defined skin layers and the creation of extracellular matrix (ECM) in GelMA/gelatin hydrogels during a 14-day culture period. Our study showed that a 3D-bioprinted composite scaffold comprising GelMA, gelatin, and AME can be used to regenerate skin tissues.
Collapse
Affiliation(s)
- Zahra Pazhouhnia
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- AstraBionics Research Network (ARN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Noori
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Farzin
- Material Engineering Department, Faculty of Engineering, Tarbiat Modares University, Tehran, Iran
| | - Keyvan Khoshmaram
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, 1417935840, Iran
| | - Mahdieh Hoseinpour
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
3
|
Yuan J, Zeng Y, Pan Z, Feng Z, Bao Y, Ye Z, Li Y, Tang J, Liu X, He Y. Amino-Functionalized Zirconium-Based Metal-Organic Frameworks as Bifunctional Nanomaterials to Treat Bone Tumors and Promote Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:53217-53227. [PMID: 37943099 DOI: 10.1021/acsami.3c11787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Bone tumor patients often encounter challenges associated with cancer cell residues and bone defects postoperation. To address this, there is an urgent need to develop a material that can enable tumor treatment and promote bone repair. Metal-organic frameworks (MOFs) have attracted the interest of many researchers due to their special porous structure, which has great potential in regenerative medicine and drug delivery. However, few studies explore MOFs with dual antitumor and bone regeneration properties. In this study, we investigated amino-functionalized zirconium-based MOF nanoparticles (UiO-66-NH2 NPs) as bifunctional nanomaterials for bone tumor treatment and osteogenesis promotion. UiO-66-NH2 NPs loading with doxorubicin (DOX) (DOX@UiO-66-NH2 NPs) showed good antitumor efficacy both in vitro and in vivo. Additionally, DOX@UiO-66-NH2 NPs significantly reduced lung injury compared to free DOX in vivo. Interestingly, the internalized UiO-66-NH2 NPs notably promoted the osteogenic differentiation of preosteoblasts. RNA-sequencing data revealed that PI3K-Akt signaling pathways or MAPK signaling pathways might be involved in this enhanced osteogenesis. Overall, UiO-66-NH2 NPs exhibit dual functionality in tumor treatment and bone repair, making them highly promising as a bifunctional material with broad application prospects.
Collapse
Affiliation(s)
- Jiongpeng Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yaoxun Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhenxing Pan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - ZhenZhen Feng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Ying Bao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhaoyi Ye
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yushan Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Junze Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xujie Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
4
|
Wu P, Yanagi K, Yokota K, Hakamada M, Mabuchi M. Unusual effects of a nanoporous gold substrate on cell adhesion and differentiation because of independent multi-branch signaling of focal adhesions. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:54. [PMID: 37884819 PMCID: PMC10602965 DOI: 10.1007/s10856-023-06760-0] [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: 03/22/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
A variety of cell behaviors, such as cell adhesion, motility, and fate, can be controlled by substrate characteristics such as surface topology and chemistry. In particular, the surface topology of substrates strongly affects cell behaviors, and the topological spacing is a critical factor in inducing cell responses. Various works have demonstrated that cell adhesion was enhanced with decreasing topological spacing although differentiation progressed slowly. However, there are exceptions, and thus, correlations between topological spacing and cell responses are still debated. We show that a nanoporous gold substrate affected cell adhesion while it neither affected osteogenic nor adipogenic differentiation. In addition, the cell adhesion was reduced with decreasing pore size. These do not agree with previous findings. A focal adhesion (FA) is an aggregate of modules comprising specific proteins such as FA kinase, talin, and vinculin. Therefore, it is suggested that because various extracellular signals can be independently branched off from the FA modules, the unusual effects of nanoporous gold substrates are related to the multi-branching of FAs.
Collapse
Affiliation(s)
- Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan.
| | - Kazuya Yanagi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Kazuki Yokota
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| |
Collapse
|
5
|
Homaeigohar S, Assad MA, Azari AH, Ghorbani F, Rodgers C, Dalby MJ, Zheng K, Xu R, Elbahri M, Boccaccini AR. Biosynthesis of Zinc Oxide Nanoparticles on l-Carnosine Biofunctionalized Polyacrylonitrile Nanofibers; a Biomimetic Wound Healing Material. ACS APPLIED BIO MATERIALS 2023; 6:4290-4303. [PMID: 37721636 PMCID: PMC10583230 DOI: 10.1021/acsabm.3c00499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
Multifunctional biohybrid nanofibers (NFs) that can simultaneously drive various cellular activities and confer antibacterial properties are considered desirable in producing advanced wound healing materials. In this study, a bionanohybrid formulation was processed as a NF wound dressing to stimulate the adhesion and proliferation of fibroblast and endothelial cells that play a major role in wound healing. Polyacrylonitrile (PAN) electrospun NFs were hydrolyzed using NaOH and biofunctionalized with l-carnosine (CAR), a dipeptide which could later biosynthesize zinc oxide (ZnO) nanoparticles (NPs) on the NFs surface. The morphological study verified that ZnO NPs are uniformly distributed on the surface of CAR/PAN NFs. Through EDX and XRD analysis, it was validated that the NPs are composed of ZnO and/or ZnO/Zn(OH)2. The presence of CAR and ZnO NPs brought about a superhydrophilicity effect and notably raised the elastic modulus and tensile strength of Zn-CAR/PAN NFs. While CAR ligands were shown to improve the viability of fibroblast (L929) and endothelial (HUVEC) cells, ZnO NPs lowered the positive impact of CAR, most likely due to their repulsive negative surface charge. A scratch assay verified that CAR/PAN NFs and Zn-CAR/PAN NFs aided HUVEC migration more than PAN NFs. Also, an antibacterial assay implied that CAR/PAN NFs and Zn-CAR/PAN NFs are significantly more effective in inhibiting Staphylococcus aureus (S. aureus) than neat PAN NFs are (1000 and 500%, respectively). Taken together, compared to the neat PAN NFs, CAR/PAN NFs with and without the biosynthesized ZnO NPs can support the cellular activities of relevance for wound healing and inactivate bacteria.
Collapse
Affiliation(s)
- Shahin Homaeigohar
- School
of Science and Engineering, University of
Dundee, Dundee DD1 4HN, U.K.
| | - Mhd Adel Assad
- Nanochemistry
and Nanoengineering, Department of Chemistry and Materials Science,
School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| | - Amir Hossein Azari
- Nanochemistry
and Nanoengineering, Department of Chemistry and Materials Science,
School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| | - Farnaz Ghorbani
- Institute
of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Chloe Rodgers
- Centre
for the Cellular Microenvironment, University
of Glasgow, Glasgow 11 6EW, U.K.
| | - Matthew J. Dalby
- Centre
for the Cellular Microenvironment, University
of Glasgow, Glasgow 11 6EW, U.K.
| | - Kai Zheng
- Jiangsu
Province Engineering Research Center of Stomatological Translational
Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Rongyao Xu
- Jiangsu
Province Engineering Research Center of Stomatological Translational
Medicine, Nanjing Medical University, Nanjing 210029, China
- Department
of Oral and Maxillofacial Surgery, Stomatological Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Mady Elbahri
- Nanochemistry
and Nanoengineering, Department of Chemistry and Materials Science,
School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| | - Aldo. R. Boccaccini
- Institute
of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany
| |
Collapse
|
6
|
Benko A, Webster TJ. How to fix a broken heart-designing biofunctional cues for effective, environmentally-friendly cardiac tissue engineering. Front Chem 2023; 11:1267018. [PMID: 37901157 PMCID: PMC10602933 DOI: 10.3389/fchem.2023.1267018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023] Open
Abstract
Cardiovascular diseases bear strong socioeconomic and ecological impact on the worldwide healthcare system. A large consumption of goods, use of polymer-based cardiovascular biomaterials, and long hospitalization times add up to an extensive carbon footprint on the environment often turning out to be ineffective at healing such cardiovascular diseases. On the other hand, cardiac cell toxicity is among the most severe but common side effect of drugs used to treat numerous diseases from COVID-19 to diabetes, often resulting in the withdrawal of such pharmaceuticals from the market. Currently, most patients that have suffered from cardiovascular disease will never fully recover. All of these factors further contribute to the extensive negative toll pharmaceutical, biotechnological, and biomedical companies have on the environment. Hence, there is a dire need to develop new environmentally-friendly strategies that on the one hand would promise cardiac tissue regeneration after damage and on the other hand would offer solutions for the fast screening of drugs to ensure that they do not cause cardiovascular toxicity. Importantly, both require one thing-a mature, functioning cardiac tissue that can be fabricated in a fast, reliable, and repeatable manner from environmentally friendly biomaterials in the lab. This is not an easy task to complete as numerous approaches have been undertaken, separately and combined, to achieve it. This review gathers such strategies and provides insights into which succeed or fail and what is needed for the field of environmentally-friendly cardiac tissue engineering to prosper.
Collapse
Affiliation(s)
| | - Thomas J. Webster
- Department of Biomedical Engineering, Hebei University of Technology, Tianjin, China
- School of Engineering, Saveetha University, Chennai, India
- Program in Materials Science, UFPI, Teresina, Brazil
| |
Collapse
|
7
|
Nitti P, Narayanan A, Pellegrino R, Villani S, Madaghiele M, Demitri C. Cell-Tissue Interaction: The Biomimetic Approach to Design Tissue Engineered Biomaterials. Bioengineering (Basel) 2023; 10:1122. [PMID: 37892852 PMCID: PMC10604880 DOI: 10.3390/bioengineering10101122] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
The advancement achieved in Tissue Engineering is based on a careful and in-depth study of cell-tissue interactions. The choice of a specific biomaterial in Tissue Engineering is fundamental, as it represents an interface for adherent cells in the creation of a microenvironment suitable for cell growth and differentiation. The knowledge of the biochemical and biophysical properties of the extracellular matrix is a useful tool for the optimization of polymeric scaffolds. This review aims to analyse the chemical, physical, and biological parameters on which are possible to act in Tissue Engineering for the optimization of polymeric scaffolds and the most recent progress presented in this field, including the novelty in the modification of the scaffolds' bulk and surface from a chemical and physical point of view to improve cell-biomaterial interaction. Moreover, we underline how understanding the impact of scaffolds on cell fate is of paramount importance for the successful advancement of Tissue Engineering. Finally, we conclude by reporting the future perspectives in this field in continuous development.
Collapse
Affiliation(s)
- Paola Nitti
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (A.N.); (R.P.); (S.V.); (M.M.); (C.D.)
| | | | | | | | | | | |
Collapse
|
8
|
Wang MT, Pang SW. Enhancing Nasopharyngeal Carcinoma Cell Separation with Selective Fibronectin Coating and Topographical Modification on Polydimethylsiloxane Scaffold Platforms. Int J Mol Sci 2023; 24:12409. [PMID: 37569784 PMCID: PMC10418797 DOI: 10.3390/ijms241512409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
The extracellular matrix (ECM) serves as a complex scaffold with diverse physical dimensions and surface properties influencing NPC cell migration. Polydimethylsiloxane (PDMS), a widely used biocompatible material, is hydrophobic and undesirable for cell seeding. Thus, the establishment of a biomimetic model with varied topographies and surface properties is essential for effective NPC43 cell separation from NP460 cells. This study explored how ECM surface properties influence NP460 and NPC43 cell behaviors via plasma treatments and chemical modifications to alter the platform surface. In addition to the conventional oxygen/nitrogen (O2/N2) plasma treatment, O2 and argon plasma treatments were utilized to modify the platform surface, which increased the hydrophilicity of the PDMS platforms, resulting in enhanced cell adhesion. (3-aminopropyl)triethoxysilane and fibronectin (FN) were used to coat the PDMS platforms uniformly and selectively. The chemical coatings significantly affected cell motility and spreading, as cells exhibited faster migration, elongated cell shapes, and larger spreading areas on FN-coated surfaces. Furthermore, narrower top layer trenches with 5 µm width and a lower concentration of 10 µg/mL FN were coated selectively on the platforms to limit NP460 cell movements and enhance NPC43 cell separation efficiency. A significantly high separation efficiency of 99.4% was achieved on the two-layer scaffold platform with 20/5 µm wide ridge/trench (R/T) as the top layer and 40/10 µm wide R/T as the bottom layer, coupling with 10 µg/mL FN selectively coated on the sidewalls of the top and bottom layers. This work demonstrated an innovative application of selective FN coating to direct cell behavior, offering a new perspective to probe into the subtleties of NPC cell separation efficiency. Moreover, this cost-effective and compact microsystem sets a new benchmark for separating cancer cells.
Collapse
Affiliation(s)
| | - S. W. Pang
- Department of Electrical Engineering, Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Hong Kong 999077, China;
| |
Collapse
|
9
|
Chen T, Jinno Y, Atsuta I, Tsuchiya A, Stocchero M, Bressan E, Ayukawa Y. Current surface modification strategies to improve the binding efficiency of emerging biomaterial polyetheretherketone (PEEK) with bone and soft tissue: A literature review. J Prosthodont Res 2023; 67:337-347. [PMID: 36372438 DOI: 10.2186/jpr.jpr_d_22_00138] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
PURPOSE The aim of this study was to review the literature on current surface modification strategies used to improve the binding efficiency of an emerging biological material, polyetheretherketone (PEEK), with bone and soft tissues. STUDY SELECTION This review was based on articles retrieved from PubMed, Google Scholar, Web of Science, and ScienceDirect databases. The main keywords used during the search were "polyetheretherketone (PEEK)," "implant," "surface modification," "biomaterials," "bone," "osseointegration," and "soft tissue." RESULTS The suitability of PEEK surface modification strategies has been critically analyzed and summarized here. Many cell and in vivo experiments in small animals have shown that the use of advanced modification technologies with appropriate surface modification strategies can effectively improve the surface inertness of PEEK, thereby improving its binding efficiency with bone and soft tissues. CONCLUSIONS Surface modifications of PEEK have revealed new possibilities for implant treatment; however, most results are based on in vitro or short-term in vivo evaluations in small animals. To achieve a broad application of PEEK in the field of oral implantology, more in vivo experiments and long-term clinical evaluations are needed to investigate the effects of various surface modifications on the tissue integration ability of PEEK to develop an ideal implant material.
Collapse
Affiliation(s)
- Tianjie Chen
- Section of Implant and Rehabilitative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yohei Jinno
- Section of Implant and Rehabilitative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ikiru Atsuta
- Division of Advanced Dental Devices and Therapeutics, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Michele Stocchero
- Department of Oral and Maxillofacial Surgery and Oral Medicine, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Eriberto Bressan
- Department of Neurosciences, Section of Dentistry, University of Padova, Padova, Italy
| | - Yasunori Ayukawa
- Section of Implant and Rehabilitative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Raptopoulos M, Fischer NG, Aparicio C. Implant surface physicochemistry affects keratinocyte hemidesmosome formation. J Biomed Mater Res A 2023; 111:1021-1030. [PMID: 36621832 DOI: 10.1002/jbm.a.37486] [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: 01/19/2022] [Revised: 08/04/2022] [Accepted: 12/05/2022] [Indexed: 01/10/2023]
Abstract
Previous studies have shown hydrophilic/hydrophobic implant surfaces stimulate/hinder osseointegration. An analogous concept was applied here using common biological functional groups on a model surface to promote oral keratinocytes (OKs) proliferation and hemidesmosomes (HD) to extend implant lifespans through increased soft tissue attachment. However, it is unclear what physicochemistry stimulates HDs. Thus, common biological functional groups (NH2 , OH, and CH3 ) were functionalized on glass using silanization. Non-functionalized plasma-cleaned glass and H silanization were controls. Surface modifications were confirmed with X-ray photoelectron spectroscopy and water contact angle. The amount of bovine serum albumin (BSA) and fibrinogen, and BSA thickness, were assessed to understand how adsorbed protein properties were influenced by physicochemistry and may influence HDs. OKs proliferation was measured, and HDs were quantified with immunofluorescence for collagen XVII and integrin β4. Plasma-cleaned surfaces were the most hydrophilic group overall, while CH3 was the most hydrophobic and OH was the most hydrophilic among functionalized groups. Modification with the OH chemical group showed the highest OKs proliferation and HD expression. The OKs response on OH surfaces appeared to not correlate to the amount or thickness of adsorbed model proteins. These results reveal relevant surface physicochemical features to favor HDs and improve implant soft tissue attachment.
Collapse
Affiliation(s)
- Michail Raptopoulos
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Periodontology, Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas G Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Conrado Aparicio
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
- Basic and Translational Research Division, Department of Odontology, UIC Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- IBEC - Institute for BIoengineering of Catalonia, BIST-Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
11
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Wang J, Yu W, Shi R, Yang S, Zhang J, Han X, Zhou Z, Gao W, Li Y, Zhao J. Osseointegration behavior of carbon fiber reinforced polyetheretherketone composites modified with amino groups: An in vivo study. J Biomed Mater Res B Appl Biomater 2023; 111:505-512. [PMID: 36191250 DOI: 10.1002/jbm.b.35167] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 08/31/2022] [Accepted: 09/14/2022] [Indexed: 01/21/2023]
Abstract
Polyetheretherketone (PEEK) has become increasingly popular in dentistry and orthopedics due to its excellent chemical stability, reliable biosafety, and low elastic modulus. However, PEEK's biomechanical strength and bioactivity are limited and need to be increased as an implant material. The previous study in vitro has shown that the amino-functionalized carbon fiber reinforced PEEK (A-30%-CPEEK) possessed enhanced mechanical property and bioactivity. This study aims to evaluate the effect of amino groups modification on the osseointegration behavior of carbon fiber reinforced PEEK (30%-CPEEK) in rabbits. Herein, 30%-CPEEK and A-30%-CPEEK implant discs were implanted in rabbit skulls for 5 weeks, with pure titanium implants serving as a control. The bone-forming ability and osseointegration in vivo were systematically investigated by micro-computed tomography analysis, scanning electron microscope observation, and histological evaluation. Our results showed that all detection parameters were significantly different between the A-30%-CPEEK and 30%-CPEEK groups, favoring those in the A-30%-CPEEK, whose appraisal parameters were equal to or better than pure titanium. Therefore, this study supported the importance of amino groups in facilitating the new bone formation and bone-implant integration, suggesting that A-30%-CPEEK with enhanced osseointegration will be a promising material for dental or orthopedic implants.
Collapse
Affiliation(s)
- Junyan Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Wanqi Yu
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ruining Shi
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shihui Yang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingjie Zhang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiao Han
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhe Zhou
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Weijia Gao
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yongli Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jinghui Zhao
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
13
|
Effect of Functionalization of the Polycaprolactone Film Surface on the Mechanical and Biological Properties of the Film Itself. Polymers (Basel) 2022; 14:polym14214654. [DOI: 10.3390/polym14214654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
The lack of suitable functional groups for cell adhesion on the surface of Polycaprolactone (PCL) is one of the main limitations in order to use PCL for biomedical applications. The aim of this research is to modify the PCL film surface using arginine, via an aminolysis reaction. In this regard, after PCL films formation by casting method, they were immersed in arginine solutions of various concentration at room temperature or then heated to 40 °C and in the presence of isopropanol or without it. To assess the structure of the modified surface, its wettability, and mechanical properties, methods of measuring the contact angle and the strip tensile test were used, and to compare the degree of attachment and the rate of cell proliferation, the method of fluorescent staining of cultured cells was used. The change in protein synthesis by cells on the modified surface was assessed using Western blotting. The results obtained show that the treatment of PCL films with an aqueous solution of arginine at room temperature for 1 day increases the hydrophilicity of the surface. Wherein surface modification led to a two-fold decrease of mechanical strength and flow stress, but elongation increase by about 30% for PCL films after modification in 0.5 M aqueous arginine solution at room temperature. Moreover, cell attachment and proliferation, as well as collagen synthesis, were significantly enhanced after arginine modification. The proposed simple and effective method for modifying PCL films with arginine significantly expands the possibilities for developing biocompatible scaffolds for tissue engineering.
Collapse
|
14
|
Silver Nanoparticles Produced by Laser Ablation and Re-Irradiation Are Effective Preventing Peri-Implantitis Multispecies Biofilm Formation. Int J Mol Sci 2022; 23:ijms231912027. [PMID: 36233328 PMCID: PMC9570054 DOI: 10.3390/ijms231912027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Implant-associated infection due to biofilm formation is a growing problem. Given that silver nanoparticles (Ag-NPs) have shown antibacterial effects, our goal is to study their effect against multispecies biofilm involved in the development of peri-implantitis. To this purpose, Ag-NPs were synthesized by laser ablation in de-ionized water using two different lasers, leading to the production of colloidal suspensions. Subsequently, part of each suspension was subjected to irradiation one and three times with the same laser source with which it was obtained. Ag-NPs were immobilized on the surface of titanium discs and the resultant materials were compared with unmodified titanium coupons. Nanoparticles were physico-chemically analysed to determine their shape, crystallinity, chemical composition, and mean diameter. The materials were incubated for 90 min or 48 h, to evaluate bacterial adhesion or biofilm formation respectively with Staphylococcus aureus or oral mixed bacterial flora composed of Streptococcus oralis, Actinomyces naeslundii, Veionella dispar, and Porphyromonas gingivalis. Ag-NPs help prevent the formation of biofilms both by S. aureus and by mixed oral bacterial flora. Nanoparticles re-irradiated three times showed the biggest antimicrobial effects. Modifying dental implants in this way could prevent the development of peri-implantitis.
Collapse
|
15
|
Heim M, Nixon IJ, Emmerson E, Callanan A. From hormone replacement therapy to regenerative scaffolds: A review of current and novel primary hypothyroidism therapeutics. Front Endocrinol (Lausanne) 2022; 13:997288. [PMID: 36277721 PMCID: PMC9581390 DOI: 10.3389/fendo.2022.997288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Primary hypothyroidism severely impacts the quality of life of patients through a decrease in the production of the thyroid hormones T3 and T4, leading to symptoms affecting cardiovascular, neurological, cognitive, and metabolic function. The incidence rate of primary hypothyroidism is expected to increase in the near future, partially due to increasing survival of patients that have undergone radiotherapy for head and neck cancer, which induces this disease in over half of those treated. The current standard of care encompasses thyroid hormone replacement therapy, traditionally in the form of synthetic T4. However, there is mounting evidence that this is unable to restore thyroid hormone signaling in all tissues due to often persistent symptoms. Additional complications are also present in the form of dosage difficulties, extensive drug interactions and poor patience compliance. The alternative therapeutic approach employed in the past is combination therapy, which consists of administration of both T3 and T4, either synthetic or in the form of desiccated thyroid extract. Here, issues are present regarding the lack of regulation concerning formulation and lack of data regarding safety and efficacy of these treatment methods. Tissue engineering and regenerative medicine have been applied in conjunction with each other to restore function of various tissues. Recently, these techniques have been adapted for thyroid tissue, primarily through the fabrication of regenerative scaffolds. Those currently under investigation are composed of either biopolymers or native decellularized extracellular matrix (dECM) in conjunction with either primary thyrocytes or stem cells which have undergone directed thyroid differentiation. Multiple of these scaffolds have successfully restored an athyroid phenotype in vivo. However, further work is needed until clinical translation can be achieved. This is proposed in the form of exploration and combination of materials used to fabricate these scaffolds, the addition of peptides which can aid restoration of tissue homeostasis and additional in vivo experimentation providing data on safety and efficacy of these implants.
Collapse
Affiliation(s)
- Maria Heim
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ian J. Nixon
- Department of ENT, Head and Neck Surgery, NHS Lothian, Edinburgh, United Kingdom
| | - Elaine Emmerson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Zhao Y, Bai L, Zhang Y, Yao R, Sun Y, Hang R, Chen X, Wang H, Yao X, Xiao Y, Hang R. Type I collagen decorated nanoporous network on titanium implant surface promotes osseointegration through mediating immunomodulation, angiogenesis, and osteogenesis. Biomaterials 2022; 288:121684. [DOI: 10.1016/j.biomaterials.2022.121684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/10/2022] [Accepted: 07/14/2022] [Indexed: 12/29/2022]
|
17
|
The Single-Step Fabrication of a Poly (Sodium Vinylsulfonate)-Grafted Polyetheretherketone Surface to Ameliorate Its Osteogenic Activity. COATINGS 2022. [DOI: 10.3390/coatings12060868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Polyetheretherketone (PEEK) is considered a potential material for replacing traditional biomedical metals used in orthopedic implants because of its similar elastic modulus to human bone. However, the poor osteogenic activity of PEEK itself hinders its clinical application. In this study, a PEEK surface was grafted with poly (sodium vinylsulfonate) through a single-step ultraviolet-initiated graft polymerization method to ameliorate its osteogenic activity. X-ray photoelectron spectroscopy and water contact angle measurements confirmed that different amounts of poly (sodium vinylsulfonate) were grafted onto the PEEK surface upon varying the ultraviolet irradiation time. Atomic force microscopy revealed that the surface topography and roughness of PEEK before and after surface grafting did not change significantly. The in vitro results showed that grafting with poly (sodium vinylsulfonate) rendered the PEEK surface with improved MC3T3-E1 osteoblast compatibility and osteogenic activity. Moreover, a PEEK surface with a higher grafting amount of poly (sodium vinylsulfonate) was observed to be more beneficial to the proliferation and osteogenic differentiation of MC3T3-E1 osteoblasts. Collectively, by employing this simple and one-step method, the osteogenic activity of PEEK can be enhanced, paving the way for the clinical application of PEEK in orthopedic implants.
Collapse
|
18
|
Xiang J, Zhou L, Xie Y, Zhu Y, Xiao L, Chen Y, Zhou W, Chen D, Wang M, Cai L, Guo L. Mesh-like electrospun membrane loaded with atorvastatin facilitates cutaneous wound healing by promoting the paracrine function of mesenchymal stem cells. Stem Cell Res Ther 2022; 13:190. [PMID: 35526075 PMCID: PMC9080129 DOI: 10.1186/s13287-022-02865-5] [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: 08/17/2021] [Accepted: 04/01/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Functional electrospun membranes are promising dressings for promoting wound healing. However, their microstructure and drug loading capacity need further improvements. It is the first time to design a novel mesh-like electrospun fiber loaded with atorvastatin (ATV) and investigated its effects on paracrine secretion by bone marrow-derived mesenchymal stem cells (BMSCs) and wound healing in vivo. METHODS We fabricated a mesh-like electrospun membrane using a copper mesh receiver. The physical properties of the membranes were evaluated by SEM, FTIR spectroscopy, tensile strength analysis, and contrast angle test. Drug release was measured by plotting concentration as a function of time. We tested the effects of conditioned media (CM) derived from BMSCs on endothelial cell migration and angiogenesis. We used these BMSCs and performed RT-PCR and ELISA to evaluate the expressions of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) genes and proteins, respectively. The involvement of FAK and AKT mechanotransduction pathways in the regulation of BMSC secretion by material surface topography was also investigated. Furthermore, we established a rat model of wound healing, applied ATV-loaded mesh-like membranes (PCL/MAT) seeded with BMSCs on wounds, and assessed their efficacy for promoting wound healing. RESULTS FTIR spectroscopy revealed successful ATV loading in PCL/MAT. Compared with random electrospun fibers (PCL/R) and mesh-like electrospun fibers without drug load (PCL/M), PCL/MAT induced maximum promotion of human umbilical vein endothelial cell (HUVEC) migration. In the PCL/MAT group, the cell sheet scratches were nearly closed after 24 h. However, the cell sheet scratches remained open in other treatments at the same time point. The PCL/MAT promoted angiogenesis and led to the generation of longer tubes than the other treatments. Finally, the PCL/MAT induced maximum gene expression and protein secretion of VEGF and b-FGF. As for material surface topography effect on BMSCs, FAK and AKT signaling pathways were shown to participate in the modulation of MSC morphology and its paracrine function. In vivo, PCL/MAT seeded with BMSCs significantly accelerated healing and improved neovascularization and collagen reconstruction in the wound area compared to the other treatments. CONCLUSIONS The mesh-like topography of fibrous scaffolds combined with ATV release creates a unique microenvironment that promotes paracrine secretion of BMSCs, thereby accelerating wound healing. Hence, drug-loaded mesh-like electrospun membranes may be highly efficacious for wound healing and as artificial skin. It is a promising approach to solve the traumatic skin defect and accelerate recovery, which is essential to developing functional materials for future regenerative medicine.
Collapse
Affiliation(s)
- Jieyu Xiang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ling Zhou
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yufan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yan Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Zhou
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Danyang Chen
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Min Wang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liang Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
19
|
A bioactive porous scaffold containing collagen/ phosphorous-modified polycaprolactone for osteogenesis of adipose-derived mesenchymal stem cells. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
[Osteoimmunomodulatory effects of inorganic biomaterials in the process of bone repair]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:517-522. [PMID: 35426295 PMCID: PMC9011079 DOI: 10.7507/1002-1892.202112025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To review the osteoimmunomodulatory effects and related mechanisms of inorganic biomaterials in the process of bone repair. METHODS A wide range of relevant domestic and foreign literature was reviewed, the characteristics of various inorganic biomaterials in the process of bone repair were summarized, and the osteoimmunomodulatory mechanism in the process of bone repair was discussed. RESULTS Immune cells play a very important role in the dynamic balance of bone tissue. Inorganic biomaterials can directly regulate the immune cells in the body by changing their surface roughness, surface wettability, and other physical and chemical properties, constructing a suitable immune microenvironment, and then realizing dynamic regulation of bone repair. CONCLUSION Inorganic biomaterials are a class of biomaterials that are widely used in bone repair. Fully understanding the role of inorganic biomaterials in immunomodulation during bone repair will help to design novel bone immunomodulatory scaffolds for bone repair.
Collapse
|
21
|
Fischer NG, Kobe AC, Dai J, He J, Wang H, Pizarek JA, De Jong DA, Ye Z, Huang S, Aparicio C. Tapping basement membrane motifs: Oral junctional epithelium for surface-mediated soft tissue attachment to prevent failure of percutaneous devices. Acta Biomater 2022; 141:70-88. [PMID: 34971784 PMCID: PMC8898307 DOI: 10.1016/j.actbio.2021.12.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023]
Abstract
Teeth, long-lasting percutaneous organs, feature soft tissue attachment through adhesive structures, hemidesmosomes, in the junctional epithelium basement membrane adjacent to teeth. This soft tissue attachment prevents bacterial infection of the tooth despite the rich - and harsh - microbial composition of the oral cavity. Conversely, millions of percutaneous devices (catheters, dental, and orthopedic implants) fail from infection yearly. Standard of care antibiotic usage fuels antimicrobial resistance and is frequently ineffective. Infection prevention strategies, like for dental implants, have failed in generating durable soft tissue adhesion - like that seen with the tooth - to prevent bacterial colonization at the tissue-device interface. Here, inspired by the impervious natural attachment of the junctional epithelium to teeth, we synthesized four cell adhesion peptide (CAPs) nanocoatings, derived from basement membranes, to promote percutaneous device soft tissue attachment. The two leading nanocoatings upregulated integrin-mediated hemidesmosomes, selectively increased keratinocyte proliferation compared to fibroblasts, which cannot form hemidesmosomes, and expression of junctional epithelium adhesive markers. CAP nanocoatings displayed marked durability under simulated clinical conditions and the top performer CAP nanocoating was validated in a percutaneous implant murine model. Basement membrane CAP nanocoatings, inspired by the tooth and junctional epithelium, may provide an alternative anti-infective strategy for percutaneous devices to mitigate the worldwide threat of antimicrobial resistance. STATEMENT OF SIGNIFICANCE: Prevention and management of medical device infection is a significant healthcare challenge. Overzealous antibiotic use has motivated alternative material innovations to prevent infection. Here, we report implant cell adhesion peptide nanocoatings that mimic a long-lasting, natural "medical device," the tooth, through formation of cell adhesive structures called hemidesmosomes. Such nanocoatings sidestep the use of antimicrobial or antibiotic elements to form a soft-tissue seal around implants. The top performing nanocoatings prompted expression of hemidesmosomes and defensive factors to mimic the tooth and was validated in an animal model. Application of cell adhesion peptide nanocoatings may provide an alternative to preventing, rather that necessarily treating, medical device infection across a range of device indications, like dental implants.
Collapse
Affiliation(s)
- Nicholas G Fischer
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Alexandra C Kobe
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Jinhong Dai
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Jiahe He
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Hongning Wang
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - John A Pizarek
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States; United States Navy Dental Corps, Naval Medical Leader and Professional Development Command, 8955 Wood Road Bethesda, MD 20889, United States
| | - David A De Jong
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Zhou Ye
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Conrado Aparicio
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States.
| |
Collapse
|
22
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
23
|
Cetin Genc C, Yilmaz HD, Karaca B, Kiran F, Arslan YE. Nano-hydroxyapatite incorporated quince seed mucilage bioscaffolds for osteogenic differentiation of human adipose-derived mesenchymal stem cells. Int J Biol Macromol 2022; 195:492-505. [PMID: 34921891 DOI: 10.1016/j.ijbiomac.2021.12.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
In this study, the therapeutic hydrocolloid quince seed mucilage (QSM) from Cydonia oblonga Miller fruit is enriched with needle-like nano-hydroxyapatite (nHAp) crystals to fabricate a novel biomimetic osteogenic bioscaffold. The molecular weight (Mw) of water-based extracted QSM was measured with GPC (8.67 × 105 g/mol), and the composite blend was prepared at a ratio of 1:1 (w/w) QSMaq and nHAp. The porous bioscaffolds were manufactured by the freeze-drying method, and evaluated in-depth with advanced analyses. The XRD, ATR-FTIR, SEM-EDX, and elemental mapping analyses revealed a uniform coated semi-crystalline structure with no covalent bindings between QSM and nHAp. Moreover, due to the hydrocolloid backbone, a supreme swelling ratio (w/w, 6523 ± 190%) with suitable pore size (208.12 ± 99.22 μm) for osteogenic development was obtained. Further, the cytocompatible bioscaffolds were evaluated for osteogenic differentiation in vitro using human adipose-derived mesenchymal stem cells (hAMSCs). The immuno/histochemical (I/HC) staining revealed that the cells with the spherical morphology invaded the pores of the prepared bioscaffolds. Also, relatively early up-regulated osteogenic markers were observed by the qRT-PCR analyses. Overall, it is believed that the QSM-nHAp bioscaffolds might be favorable in non-load bearing applications, especially in the cranio-maxillofacial region, due to their regenerative, bendable, and durable features.
Collapse
Affiliation(s)
- Cigdem Cetin Genc
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Hilal Deniz Yilmaz
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Burak Karaca
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, 06100 Ankara, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey.
| |
Collapse
|
24
|
Zummo F, Esposito P, Hou H, Wetzl C, Rius G, Tkatchenko R, Guimera A, Godignon P, Prato M, Prats-Alfonso E, Criado A, Scaini D. Bidirectional Modulation of Neuronal Cells Electrical and Mechanical Properties Through Pristine and Functionalized Graphene Substrates. Front Neurosci 2022; 15:811348. [PMID: 35087375 PMCID: PMC8788235 DOI: 10.3389/fnins.2021.811348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years, the quest for surface modifications to promote neuronal cell interfacing and modulation has risen. This course is justified by the requirements of emerging technological and medical approaches attempting to effectively interact with central nervous system cells, as in the case of brain-machine interfaces or neuroprosthetic. In that regard, the remarkable cytocompatibility and ease of chemical functionalization characterizing surface-immobilized graphene-based nanomaterials (GBNs) make them increasingly appealing for these purposes. Here, we compared the (morpho)mechanical and functional adaptation of rat primary hippocampal neurons when interfaced with surfaces covered with pristine single-layer graphene (pSLG) and phenylacetic acid-functionalized single-layer graphene (fSLG). Our results confirmed the intrinsic ability of glass-supported single-layer graphene to boost neuronal activity highlighting, conversely, the downturn inducible by the surface insertion of phenylacetic acid moieties. fSLG-interfaced neurons showed a significant reduction in spontaneous postsynaptic currents (PSCs), coupled to reduced cell stiffness and altered focal adhesion organization compared to control samples. Overall, we have here demonstrated that graphene substrates, both pristine and functionalized, could be alternatively used to intrinsically promote or depress neuronal activity in primary hippocampal cultures.
Collapse
Affiliation(s)
- Francesca Zummo
- Neuroscience Area, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Pietro Esposito
- Neuroscience Area, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Huilei Hou
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastián, Spain
| | - Cecilia Wetzl
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastián, Spain
| | - Gemma Rius
- Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
| | - Raphaela Tkatchenko
- Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
| | - Anton Guimera
- Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Philippe Godignon
- Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Maurizio Prato
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastián, Spain
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
- Basque Foundation for Science (IKERBASQUE), Bilbao, Spain
| | - Elisabet Prats-Alfonso
- Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
- *Correspondence: Elisabet Prats-Alfonso,
| | - Alejandro Criado
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastián, Spain
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, A Coruña, Spain
- Alejandro Criado,
| | - Denis Scaini
- Neuroscience Area, International School for Advanced Studies (SISSA), Trieste, Italy
- Nanomedicine Research Laboratory, Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Denis Scaini,
| |
Collapse
|
25
|
Kim HY, Kim BH, Kim MS. Amine Plasma-Polymerization of 3D Polycaprolactone/β-Tricalcium Phosphate Scaffold to Improving Osteogenic Differentiation In Vitro. MATERIALS 2022; 15:ma15010366. [PMID: 35009509 PMCID: PMC8745968 DOI: 10.3390/ma15010366] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/20/2021] [Accepted: 01/02/2022] [Indexed: 01/19/2023]
Abstract
This study aims to investigate the surface characterization and pre-osteoblast biological behaviors on the three-dimensional (3D) poly(ε-caprolactone)/β-tricalcium phosphate (β-TCP) scaffold modified by amine plasma-polymerization. The 3D PCL scaffolds were fabricated using fused deposition modeling (FDM) 3D printing. To improve the pre-osteoblast bioactivity, the 3D PCL scaffold was modified by adding β-TCP nanoparticles, and then scaffold surfaces were modified by amine plasma-polymerization using monomer allylamine (AA) and 1,2-diaminocyclohexane (DACH). After the plasma-polymerization of PCL/β-TCP, surface characterizations such as contact angle, AFM, XRD, and FTIR were evaluated. In addition, mechanical strength was measured by UTM. The pre-osteoblast bioactivities were evaluated by focal adhesion and cell proliferation. Osteogenic differentiation was investigated by ALP activity, Alizarin red staining, and Western blot. Plasma-polymerization induced the increase in hydrophilicity of the surface of the 3D PCL/β-TCP scaffold due to the deposition of amine polymeric thin film on the scaffold surface. Focal adhesion and proliferation of pre-osteoblast improved, and osteogenic differentiation was increased. These results indicated that 3D PCL/β-TCP scaffolds treated with DACH plasma-polymerization showed the highest bioactivity compared to the other samples. We suggest that 3D PCL/β-TCP scaffolds treated with DACH and AA plasma-polymerization can be used as a promising candidate for osteoblast differentiation of pre-osteoblast.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea;
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Byung-Hoon Kim
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
- Correspondence: (B.-H.K.); (M.-S.K.); Tel.: +82-62-230-6447 (B.-H.K.); +82-62-227-1640 (M.-S.K.)
| | - Myung-Sun Kim
- Department of Orthopaedic Surgery, College of Medicine, Chonnam National University, Gwangju 61469, Korea
- Correspondence: (B.-H.K.); (M.-S.K.); Tel.: +82-62-230-6447 (B.-H.K.); +82-62-227-1640 (M.-S.K.)
| |
Collapse
|
26
|
Covalent attachment of three derivatives of pegylated RGD peptides on the NH2-terminated silicon surfaces: Impact on fibroblast cell behavior. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183770. [PMID: 34517002 DOI: 10.1016/j.bbamem.2021.183770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 11/20/2022]
Abstract
This paper describes a simple strategy for covalent immobilization of the NHS-PEG-RGD peptide with the three different PEG lengths (8, 13, and 22) onto the amine-terminated monolayers with the subsequent investigation of fibroblast cellular response to the three derivatives of pegylated RGD peptides-modified substrates. First, acetamide-terminated monolayers were prepared on the hydride terminated silicon surface to protect NH2-terminated monolayers. This was followed by the removal of the protective groups, and the reaction of NHS-PEG8-RGD, NHS-PEG13-RGD and NHS-PEG22-RGD peptides with the NH2-terminated monolayers while reducing nonspecific protein adsorption. Analysis of X-ray photoelectron spectroscopy (XPS), Fourier Transform Infrared (ATR-FTIR) spectroscopy, and Ellipsometry measurements demonstrated that PEG13-RGD peptide forms relatively a more homogenous, thicker and stable structure compared with those of PEG8-RGD and PEG22-RGD peptide. The quantitative and qualitative assessment of cell adhesion, spreading, and proliferation indicated that relatively further elongated fibroblast cells attached on the PEG13-RGD peptide relative to those on the PEG8-RGD and PEG22-RGD peptide. The results presented here may offer a developed strategy based on the length of the spacer to regulate cellular behavior on the surface substrates.
Collapse
|
27
|
Yang Y, Zhang H, Komasa S, Morimoto Y, Sekino T, Kawazoe T, Okazaki J. UV/ozone irradiation manipulates immune response for antibacterial activity and bone regeneration on titanium. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112377. [PMID: 34579896 DOI: 10.1016/j.msec.2021.112377] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
The immunomodulatory antibacterial activity and osteoimmunomodulatory properties of implantable biomaterials significantly influence bone regeneration. Various types of ultraviolet (UV) instrument are currently in use to greatly enhance the antibacterial activity and osteoconductive capability of titanium, it remains unclear how UV treatment modulates immune response. Compared to traditional UV treatment, the combination of low-dose ozone with UV irradiation is considered a new option to give benefits to surface modification and reduce the drawbacks of UV and ozone individually. Herein, the aim of this study was to elucidate the immune-modulatory properties of macrophages on UV/ozone-irradiated titanium that serve as defense against S. aureus and the crosstalk between immune cells and osteoblasts. Three different cell and bacteria co-culture systems were developed in order to investigate the race between host cells and bacteria to occupy the surface. In vitro immunological experiments indicated that UV/ozone irradiation significantly enhanced the phagocytic and bactericidal activity of macrophages against S. aureus. Further, in vitro and in vivo studies evidenced the favorable osteoimmune environment for osteogenic differentiation and bone formation. This research suggests vital therapeutic potential of UV/ozone irradiation for preventing the biomaterial-associated infections and achieving favorable bone formation simultaneously.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | - Honghao Zhang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | - Satoshi Komasa
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| | - Yukihiro Morimoto
- The Institute of Scientific and Industrial Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tohru Sekino
- The Institute of Scientific and Industrial Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takayoshi Kawazoe
- Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| | - Joji Okazaki
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| |
Collapse
|
28
|
Xu Z, Zhang L, Bentil SA, Bratlie KM. Gellan gum-gelatin viscoelastic hydrogels as scaffolds to promote fibroblast differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112370. [PMID: 34579889 DOI: 10.1016/j.msec.2021.112370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/31/2021] [Accepted: 08/10/2021] [Indexed: 11/18/2022]
Abstract
Fabricating hydrogel scaffolds that are both bioreactive toward fibroblasts while still mechanically compatible with surrounding tissue is a major challenge in tissue engineering. This is because the outcome of scaffold implantation is largely determined by fibroblasts differentiating toward myofibroblasts, which is characterized by the expression of α-smooth muscle actin (α-SMA). Previous studies promoted fibroblasts differentiation by increasing scaffold substrate stiffness. However, the stiffness of scaffold has to be compatible with surrounding tissue, as mismatched stiffness may cause initial hyperplasia and inappropriate endothelial layer development. Therefore, we adjusted the hydrogel chemical component, and thus viscoelasticity to affect the mechano-signaling of fibroblasts and promote fibroblasts differentiation. Elastic gellan gum and viscoelastic gelatin were hybridized at different ratios to fabricate hydrogel scaffold with varied stress-relaxation. Vitronectin (VN) was used to further regulate the interaction between fibroblasts and the substrate. Fibroblast differentiation, characterized by α-SMA area per cell, increased from~3000-4000 μm2/cell on less viscoelastic gels to ~5000 μm2/cell on the most viscoelastic gel. Fibroblasts seeded on hydrogels had a slower migration rate on more viscoelastic hydrogels (slowest at 38 ± 14 μm/h) compared to the migration speed on less viscoelastic hydrogels (74 ± 20 μm/h). VN slowed the migration speed on all hydrogels. The organization of collagen deposited by fibroblasts cultured on the hydrogels was characterized by second harmonic generation (SHG), which showed that collagen was more organized (parallel) on more viscoelastic hydrogels. In summary, we provided a novel strategy to fabricate hydrogel scaffolds that can promote fibroblasts differentiation while keeping the stiffness compatible with blood vessels. The most viscoelastic hydrogel studied here meets these requirements best.
Collapse
Affiliation(s)
- Zihao Xu
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, United States of America
| | - Ling Zhang
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, United States of America
| | - Sarah A Bentil
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, United States of America
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, United States of America; Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States of America.
| |
Collapse
|
29
|
Kodama J, Harumningtyas AA, Ito T, Michlíček M, Sugimoto S, Kita H, Chijimatsu R, Ukon Y, Kushioka J, Okada R, Kamatani T, Hashimoto K, Tateiwa D, Tsukazaki H, Nakagawa S, Takenaka S, Makino T, Sakai Y, Nečas D, Zajíčková L, Hamaguchi S, Kaito T. Amine modification of calcium phosphate by low-pressure plasma for bone regeneration. Sci Rep 2021; 11:17870. [PMID: 34504247 PMCID: PMC8429709 DOI: 10.1038/s41598-021-97460-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Regeneration of large bone defects caused by trauma or tumor resection remains one of the biggest challenges in orthopedic surgery. Because of the limited availability of autograft material, the use of artificial bone is prevalent; however, the primary role of currently available artificial bone is restricted to acting as a bone graft extender owing to the lack of osteogenic ability. To explore whether surface modification might enhance artificial bone functionality, in this study we applied low-pressure plasma technology as next-generation surface treatment and processing strategy to chemically (amine) modify the surface of beta-tricalcium phosphate (β-TCP) artificial bone using a CH4/N2/He gas mixture. Plasma-treated β-TCP exhibited significantly enhanced hydrophilicity, facilitating the deep infiltration of cells into interconnected porous β-TCP. Additionally, cell adhesion and osteogenic differentiation on the plasma-treated artificial bone surfaces were also enhanced. Furthermore, in a rat calvarial defect model, the plasma treatment afforded high bone regeneration capacity. Together, these results suggest that amine modification of artificial bone by plasma technology can provide a high osteogenic ability and represents a promising strategy for resolving current clinical limitations regarding the use of artificial bone.
Collapse
Affiliation(s)
- Joe Kodama
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Anjar Anggraini Harumningtyas
- Center for Atomic and Molecular Technologies (CAMT), Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Center for Accelerator Science and Technology, National Nuclear Energy Agency of Indonesia (BATAN), Jalan Babarsari Kotak Pos 6101 ykbb, Yogyakarta, 55281, Indonesia
| | - Tomoko Ito
- Center for Atomic and Molecular Technologies (CAMT), Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Miroslav Michlíček
- Department of Physical Electronics, Faculty of Science, Masaryk University, Kotlarska 2, 61137, Brno, Czech Republic
| | - Satoshi Sugimoto
- Center for Atomic and Molecular Technologies (CAMT), Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Kita
- Center for Atomic and Molecular Technologies (CAMT), Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Chijimatsu
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuichiro Ukon
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Rintaro Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takashi Kamatani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kunihiko Hashimoto
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Tateiwa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Tsukazaki
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinichi Nakagawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shota Takenaka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takahiro Makino
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Sakai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - David Nečas
- CEITEC - Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, 61200, Czech Republic
| | - Lenka Zajíčková
- CEITEC - Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, 61200, Czech Republic.,Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlarska 2, Brno, 61137, Czech Republic
| | - Satoshi Hamaguchi
- Center for Atomic and Molecular Technologies (CAMT), Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
30
|
Slay EE, Meldrum FC, Pensabene V, Amer MH. Embracing Mechanobiology in Next Generation Organ-On-A-Chip Models of Bone Metastasis. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:722501. [PMID: 35047952 PMCID: PMC8757701 DOI: 10.3389/fmedt.2021.722501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis in breast cancer is associated with high mortality. Biomechanical cues presented by the extracellular matrix play a vital role in driving cancer metastasis. The lack of in vitro models that recapitulate the mechanical aspects of the in vivo microenvironment hinders the development of novel targeted therapies. Organ-on-a-chip (OOAC) platforms have recently emerged as a new generation of in vitro models that can mimic cell-cell interactions, enable control over fluid flow and allow the introduction of mechanical cues. Biomaterials used within OOAC platforms can determine the physical microenvironment that cells reside in and affect their behavior, adhesion, and localization. Refining the design of OOAC platforms to recreate microenvironmental regulation of metastasis and probe cell-matrix interactions will advance our understanding of breast cancer metastasis and support the development of next-generation metastasis-on-a-chip platforms. In this mini-review, we discuss the role of mechanobiology on the behavior of breast cancer and bone-residing cells, summarize the current capabilities of OOAC platforms for modeling breast cancer metastasis to bone, and highlight design opportunities offered by the incorporation of mechanobiological cues in these platforms.
Collapse
Affiliation(s)
- Ellen E. Slay
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Virginia Pensabene
- School of School of Electronic and Electrical Engineering, University of Leeds, Leeds, United Kingdom
- School of Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
31
|
The Influence of the Surface Topographical Cues of Biomaterials on Nerve Cells in Peripheral Nerve Regeneration: A Review. Stem Cells Int 2021; 2021:8124444. [PMID: 34349803 PMCID: PMC8328695 DOI: 10.1155/2021/8124444] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 01/01/2023] Open
Abstract
The surface topographies of artificial implants including surface roughness, surface groove size and orientation, and surface pore size and distribution have a great influence on the adhesion, migration, proliferation, and differentiation of nerve cells in the nerve regeneration process. Optimizing the surface topographies of biomaterials can be a key strategy for achieving excellent cell performance in various applications such as nerve tissue engineering. In this review, we offer a comprehensive summary of the surface topographies of nerve implants and their effects on nerve cell behavior. This review also emphasizes the latest work progress of the layered structure of the natural extracellular matrix that can be imitated by the material surface topology. Finally, the future development of surface topographies on nerve regeneration was prospectively remarked.
Collapse
|
32
|
Wang J, Li J, Liu J, Lin M, Mao S, Wang Y, Luo Y. Adsorption Force of Fibronectin: A Balance Regulator to Transmission of Cell Traction Force and Fluid Shear Stress. Biomacromolecules 2021; 22:3264-3273. [PMID: 34225453 DOI: 10.1021/acs.biomac.1c00375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Osteoblasts actively generate cell traction force (CTF) to sense chemical and mechanical microenvironments. Fluid shear stress (FSS) is a principle mechanical stimulus for bone modeling/remodeling. FSS and CTF share common interconnected elements for force transmission, among which the role of the protein-material interfacial force (Fad) remains unclear. Here, we found that, on the low Fad surface (5.47 ± 1.31 pN/FN), CTF overwhelmed Fad to partially desorb FN, and FSS exacerbated the desorption, resulting in disassembly of the actin cytoskeleton and focal adhesions (FAs) to reduce CTF and establishment of a new mechanical balance at the FN-material interface. Contrarily, on the high Fad surface (27.68 ± 5.24 pN/FN), pure CTF or the combination of CTF and FSS induced no FN desorption, and FSS promoted assembly of actin cytoskeletons and disassembly of FAs, regaining new mechanical balance at the cell-FN interface. These results indicate that Fad is a mechanical regulator for transmission of CTF and FSS, which has never been reported before.
Collapse
Affiliation(s)
- Jinfeng Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Junyao Li
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Juan Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Manping Lin
- Key Laboratory of Emergency and Trauma, Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Shilong Mao
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Yuanliang Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Yanfeng Luo
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
33
|
Strohbach A, Maess F, Wulf K, Petersen S, Grabow N, Schmitz KP, Felix SB, Busch R. The Role of Biodegradable Poly-(L-lactide)-Based Polymers in Blood Cell Activation and Platelet-Monocyte Interaction. Int J Mol Sci 2021; 22:ijms22126340. [PMID: 34199303 PMCID: PMC8231768 DOI: 10.3390/ijms22126340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 06/07/2021] [Indexed: 01/23/2023] Open
Abstract
The main purpose of new stent technologies is to overcome unfavorable material-related incompatibilities by producing bio- and hemo-compatible polymers with anti-inflammatory and anti-thrombogenic properties. In this context, wettability is an important surface property, which has a major impact on the biological response of blood cells. However, the influence of local hemodynamic changes also influences blood cell activation. Therefore, we investigated biodegradable polymers with different wettability to identify possible aspects for a better prediction of blood compatibility. We applied shear rates of 100 s−1 and 1500 s−1 and assessed platelet and monocyte activation as well as the formation of CD62P+ monocyte-bound platelets via flow cytometry. Aggregation of circulating platelets induced by collagen was assessed by light transmission aggregometry. Via live cell imaging, leukocytes were tracked on biomaterial surfaces to assess their average velocity. Monocyte adhesion on biomaterials was determined by fluorescence microscopy. In response to low shear rates of 100 s−1, activation of circulating platelets and monocytes as well as the formation of CD62P+ monocyte-bound platelets corresponded to the wettability of the underlying material with the most favorable conditions on more hydrophilic surfaces. Under high shear rates, however, blood compatibility cannot only be predicted by the concept of wettability. We assume that the mechanisms of blood cell-polymer interactions do not allow for a rule-of-thumb prediction of the blood compatibility of a material, which makes extensive in vitro testing mandatory.
Collapse
Affiliation(s)
- Anne Strohbach
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany; (F.M.); (S.B.F.); (R.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Fleischmannstr. 42-44, 17489 Greifswald, Germany
- Correspondence:
| | - Friedemann Maess
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany; (F.M.); (S.B.F.); (R.B.)
| | - Katharina Wulf
- Institute for Biomedical Engineering, Rostock University Medical Center, Friedrich-Barnewitz-Str. 4, 18119 Rostock, Germany; (K.W.); (S.P.); (N.G.); (K.-P.S.)
| | - Svea Petersen
- Institute for Biomedical Engineering, Rostock University Medical Center, Friedrich-Barnewitz-Str. 4, 18119 Rostock, Germany; (K.W.); (S.P.); (N.G.); (K.-P.S.)
- Faculty of Engineering and Computer Science, University of Applied Sciences, Albrechtstr. 30, 49076 Osnabrück, Germany
| | - Niels Grabow
- Institute for Biomedical Engineering, Rostock University Medical Center, Friedrich-Barnewitz-Str. 4, 18119 Rostock, Germany; (K.W.); (S.P.); (N.G.); (K.-P.S.)
| | - Klaus-Peter Schmitz
- Institute for Biomedical Engineering, Rostock University Medical Center, Friedrich-Barnewitz-Str. 4, 18119 Rostock, Germany; (K.W.); (S.P.); (N.G.); (K.-P.S.)
| | - Stephan B. Felix
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany; (F.M.); (S.B.F.); (R.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Fleischmannstr. 42-44, 17489 Greifswald, Germany
| | - Raila Busch
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany; (F.M.); (S.B.F.); (R.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Fleischmannstr. 42-44, 17489 Greifswald, Germany
| |
Collapse
|
34
|
Ren C, Hao X, Wang L, Hu Y, Meng L, Zheng S, Ren F, Bu W, Wang H, Li D, Zhang K, Sun H. Metformin Carbon Dots for Promoting Periodontal Bone Regeneration via Activation of ERK/AMPK Pathway. Adv Healthc Mater 2021; 10:e2100196. [PMID: 33987977 DOI: 10.1002/adhm.202100196] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/15/2021] [Indexed: 12/14/2022]
Abstract
The osteogenic potential of mesenchymal stem cells (MSCs) is severely impaired under persistent inflammation of periodontitis. A highly efficient way to promote or rescue osteogenic potential of MSCs under inflammation remains an unmet goal. Herein, metformin carbon dots (MCDs) with excellent biocompatibility are prepared from metformin hydrochloride and citric acid via a hydrothermal method. The MCDs can more effectively enhance the alkaline phosphatase (ALP) activity, calcium deposition nodules formation, expression of osteogenic genes and proteins in rat bone marrow mesenchymal stem cells (rBMSCs) than metformin under both inflammatory and normal conditions. Moreover, a novel pathway of extracellular signal-regulated kinases (ERK)/AMP-activated protein kinase (AMPK) signaling is involved in the MCDs-induced osteogenesis. In periodontitis rats, MCDs can effectively regenerate the lost alveolar bone, but not the metformin. Taken together, MCDs can be the promising candidate nanomaterial for periodontitis treatment. This work may provide a new pharmacological target of ERK/AMPK pathway for treating bone loss and also give additional insights into developing nanodrugs from the numerous medications.
Collapse
Affiliation(s)
- Chunxia Ren
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Xinqing Hao
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Lu Wang
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Yue Hu
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Lin Meng
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Shize Zheng
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Feilong Ren
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Wenhuan Bu
- School of Stomatology China Medical University Shenyang 110001 P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Daowei Li
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| | - Kai Zhang
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University Changchun 130012 P. R. China
| | - Hongchen Sun
- Hospital of Stomatology Jilin University Changchun 130021 P. R. China
| |
Collapse
|
35
|
Sarwar M, Sykes PH, Chitcholtan K, Evans JJ. Deciphering Biophysical Modulation in Ovarian Cancer Cells. Cell Biochem Biophys 2021; 79:375-386. [PMID: 33433760 DOI: 10.1007/s12013-020-00964-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2020] [Indexed: 02/08/2023]
Abstract
It has been long known that the oncogenic extracellular environment plays an indispensable role in developing and nurturing cancer cell progression and in resistance to standard treatments. However, by how much the biophysical components of tumour extracellular environment contribute to these processes is uncertain. In particular, the topographic environment is scarcely explored. The biophysical modulation of cell behaviour is primarily facilitated through mechanotransduction-associated mechanisms, including focal adhesion and Rho/ROCK signalling. Dysregulation of these pathways is commonly observed in ovarian cancer and, therefore, biophysical modulation of these mechanisms may be of great importance to ovarian cancer development and progression. In this work, aspects of the biophysical environment were explored using a bioimprinting technique. The study showed that topography-mediated substrate sensing delayed cell attachment, however, cell-cell interactions overrode the effect of topography in some cell lines, such as OVCAR-5. Also, 3D topographical cues were shown to modulate the inhibition of focal adhesion and Rho signalling, which resulted in higher MAPK activity in cells on the bioimprints. It was revealed that c-Src is vital to the biophysical modulation of cell proliferation and inhibition of c-Src could downregulate biophysically modulated MAPK activity. This study provides evidence that the biophysical extracellular environment affects key intracellular mechanisms associated with tumourigenicity in ovarian cancer cells.
Collapse
Affiliation(s)
- Makhdoom Sarwar
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - John J Evans
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
- MacDiarmid Institute of Advanced Materials and Nanotechnology, Christchurch, New Zealand
| |
Collapse
|
36
|
Li Z, Bratlie KM. The Influence of Polysaccharides-Based Material on Macrophage Phenotypes. Macromol Biosci 2021; 21:e2100031. [PMID: 33969643 DOI: 10.1002/mabi.202100031] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 02/03/2023]
Abstract
Macrophage polarization is a key factor in determining the success of implanted tissue engineering scaffolds. Polysaccharides (derived from plants, animals, and microorganisms) are known to modulate macrophage phenotypes by recognizing cell membrane receptors. Numerous studies have developed polysaccharide-based materials into functional biomaterial substrates for tissue regeneration and pharmaceutical application due to their immunostimulatory activities and anti-inflammatory response. They are used as hydrogel substrates, surface coatings, and drug delivery carriers. In addition to their innate immunological functions, the newly endowed physical and chemical properties, including substrate modulus, pore size/porosity, surface binding chemistry, and the mole ratio of polysaccharides in hybrid materials may regulate macrophage phenotypes more precisely. Growing evidence indicates that the sulfation pattern of glycosaminoglycans and proteoglycans expressed on polarized macrophages leads to the changes in protein binding, which may alter macrophage phenotype and influence the immune response. A comprehensive understanding of how different types of polysaccharide-based materials alter macrophage phenotypic changes can be beneficial to predict transplantation/implantation outcomes. This review focuses on recent advances in promoting wound healing and balancing macrophage phenotypes using polysaccharide-based substrates/coatings and new directions to address the limitations in the current understanding of macrophage responses to polysaccharides.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA.,Department of Chemical & Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
37
|
Wei F, Liu S, Chen M, Tian G, Zha K, Yang Z, Jiang S, Li M, Sui X, Chen Z, Guo Q. Host Response to Biomaterials for Cartilage Tissue Engineering: Key to Remodeling. Front Bioeng Biotechnol 2021; 9:664592. [PMID: 34017827 PMCID: PMC8129172 DOI: 10.3389/fbioe.2021.664592] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Biomaterials play a core role in cartilage repair and regeneration. The success or failure of an implanted biomaterial is largely dependent on host response following implantation. Host response has been considered to be influenced by numerous factors, such as immune components of materials, cytokines and inflammatory agents induced by implants. Both synthetic and native materials involve immune components, which are also termed as immunogenicity. Generally, the innate and adaptive immune system will be activated and various cytokines and inflammatory agents will be consequently released after biomaterials implantation, and further triggers host response to biomaterials. This will guide the constructive remolding process of damaged tissue. Therefore, biomaterial immunogenicity should be given more attention. Further understanding the specific biological mechanisms of host response to biomaterials and the effects of the host-biomaterial interaction may be beneficial to promote cartilage repair and regeneration. In this review, we summarized the characteristics of the host response to implants and the immunomodulatory properties of varied biomaterial. We hope this review will provide scientists with inspiration in cartilage regeneration by controlling immune components of biomaterials and modulating the immune system.
Collapse
Affiliation(s)
- Fu Wei
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Shuyun Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Mingxue Chen
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Guangzhao Tian
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Kangkang Zha
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Zhen Yang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | | | - Muzhe Li
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xiang Sui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiwei Chen
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Quanyi Guo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Renkler NZ, Ergene E, Gokyer S, Tuzlakoglu Ozturk M, Yilgor Huri P, Tuzlakoglu K. Facile modification of polycaprolactone nanofibers with egg white protein. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:34. [PMID: 33763760 PMCID: PMC7990845 DOI: 10.1007/s10856-021-06505-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
Synthetic polymers remain to be a major choice for scaffold fabrication due to their structural stability and mechanical strength. However, the lack of functional moieties limits their application for cell-based therapies which necessitate modification and functionalization. Blending synthetic polymers with natural components is a simple and effective way to achieve the desired biological properties for a scaffold. Herein, nanofibrous mats made of polycaprolactone (PCL) and egg white protein (EWP) blend were developed and further evaluated for use as a scaffold for tissue engineering applications. Homogeneous distribution of EWP was achieved throughout the nanofibrous mats, as shown by immunohistochemistry. ATR-FTIR analysis and contact angle measurements have further confirmed the presence of EWP on the surface of the samples. The swelling test showed that PCL/EWP nanofibers have higher water uptake than PCL nanofibrous mats. Also, EWP addition on the nanofibrous mats resulted in an increase in the tensile strength and Young's modulus of the mats, indicating that the presence of protein can greatly enhance the mechanical properties of the mats. A significantly higher, more uniform, and dispersed cell spreading was observed on days 7 and 14 than that on neat PCL mats, demonstrating the importance of providing the required cues for cell homing by the availability of EWP. Hence, EWP is shown to be a simple and low-cost source for the functionalization of PCL nanofibrous mats. EWP is, therefore, a facile candidate to enhance cellular interactions of synthetic polymers for a wide range of tissue engineering applications.
Collapse
Affiliation(s)
| | - Emre Ergene
- Department of Biomedical Engineering, Ankara University, Ankara, Turkey
| | - Seyda Gokyer
- Department of Biomedical Engineering, Ankara University, Ankara, Turkey
| | | | - Pinar Yilgor Huri
- Department of Biomedical Engineering, Ankara University, Ankara, Turkey
| | - Kadriye Tuzlakoglu
- Department of Polymer Engineering, Yalova University, 77200, Yalova, Turkey
| |
Collapse
|
39
|
Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases. Acta Biomater 2021; 123:1-30. [PMID: 33484912 DOI: 10.1016/j.actbio.2021.01.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/05/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The degree of tissue injuries such as the level of scarring or organ dysfunction, and the immune response against them primarily determine the outcome and speed of healing process. The successful regeneration of functional tissues requires proper modulation of inflammation-producing immune cells and bioactive factors existing in the damaged microenvironment. In the tissue repair and regeneration processes, different types of biomaterials are implanted either alone or by combined with other bioactive factors, which will interact with the immune systems including immune cells, cytokines and chemokines etc. to achieve different results highly depending on this interplay. In this review article, the influences of different types of biomaterials such as nanoparticles, hydrogels and scaffolds on the immune cells and the modification of immune-responsive factors such as reactive oxygen species (ROS), cytokines, chemokines, enzymes, and metalloproteinases in tissue microenvironment are summarized. In addition, the recent advances of immune-responsive biomaterials in therapy of inflammation-associated diseases such as myocardial infarction, spinal cord injury, osteoarthritis, inflammatory bowel disease and diabetic ulcer are discussed.
Collapse
|
40
|
Negrescu AM, Cimpean A. The State of the Art and Prospects for Osteoimmunomodulatory Biomaterials. MATERIALS (BASEL, SWITZERLAND) 2021; 14:1357. [PMID: 33799681 PMCID: PMC7999637 DOI: 10.3390/ma14061357] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
The critical role of the immune system in host defense against foreign bodies and pathogens has been long recognized. With the introduction of a new field of research called osteoimmunology, the crosstalk between the immune and bone-forming cells has been studied more thoroughly, leading to the conclusion that the two systems are intimately connected through various cytokines, signaling molecules, transcription factors and receptors. The host immune reaction triggered by biomaterial implantation determines the in vivo fate of the implant, either in new bone formation or in fibrous tissue encapsulation. The traditional biomaterial design consisted in fabricating inert biomaterials capable of stimulating osteogenesis; however, inconsistencies between the in vitro and in vivo results were reported. This led to a shift in the development of biomaterials towards implants with osteoimmunomodulatory properties. By endowing the orthopedic biomaterials with favorable osteoimmunomodulatory properties, a desired immune response can be triggered in order to obtain a proper bone regeneration process. In this context, various approaches, such as the modification of chemical/structural characteristics or the incorporation of bioactive molecules, have been employed in order to modulate the crosstalk with the immune cells. The current review provides an overview of recent developments in such applied strategies.
Collapse
Affiliation(s)
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania;
| |
Collapse
|
41
|
Homaeigohar S, Monavari M, Koenen B, Boccaccini AR. Biomimetic biohybrid nanofibers containing bovine serum albumin as a bioactive moiety for wound dressing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:111965. [PMID: 33812593 DOI: 10.1016/j.msec.2021.111965] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
For the first time, a biohybrid nanofibrous wound dressing is developed via green electrospinning of a blend solution of bovine serum albumin (BSA) (1 and 3 wt%) and polycaprolactone (PCL). In such a system, the components are miscible and interact through hydrogen bonding between the carbonyl group of PCL and the amine group of BSA, as verified by ATR-FTIR. As a result, the biohybrid nanofibers show a superior elastic modulus and elongation (300% and 58%, respectively) compared with the neat PCL nanofibers. The included protein induces a hydrophilicity effect to the PCL nanofibers, notably at the higher BSA content (3 wt%). In contrast to the neat nanofibers, the biohybrid ones are bioactive and encourage formation of biominerals (made of amorphous calcium carbonate) on the surface, after immersion in simulated body fluid (SBF). Based on the WST-8 cell viability tests, NIH3T3 fibroblast cells were seen to properly interact with the biohybrid mats and to proliferate in their proximity. SEM images show that the cells largely adhere onto such nanofibers even more than they do on the neat ones and adopt a flattened and stretched shape. In addition, the live/dead assay and phalloidin/DAPI staining assay confirm large cell viability and normal cell morphology on the biohybrid nanofiber mats after 4 days incubation. Taken together, BSA/PCL nanofibers are able to offer optimum mechanical properties (elasticity) as well as mineralization which can potentially stimulate the wound healing process, and can be considered a suitable candidate for wound dressing applications.
Collapse
Affiliation(s)
- Shahin Homaeigohar
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; School of Science and Engineering, University of Dundee, Dundee DD1 4HN, United Kingdom.
| | - Mahshid Monavari
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Benedict Koenen
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| |
Collapse
|
42
|
Farr N, Thanarak J, Schäfer J, Quade A, Claeyssens F, Green N, Rodenburg C. Understanding Surface Modifications Induced via Argon Plasma Treatment through Secondary Electron Hyperspectral Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003762. [PMID: 33643809 PMCID: PMC7887591 DOI: 10.1002/advs.202003762] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/10/2020] [Indexed: 06/01/2023]
Abstract
Understanding the effects that sterilization methods have on the surface of a biomaterial is a prerequisite for clinical deployment. Sterilization causes alterations in a material's surface chemistry and surface structures that can result in significant changes to its cellular response. Here we compare surfaces resulting from the application of the industry standard autoclave sterilisation to that of surfaces resulting from the use of low-pressure Argon glow discharge within a novel gas permeable packaging method in order to explore a potential new biomaterial sterilisation method. Material surfaces are assessed by applying secondary electron hyperspectral imaging (SEHI). SEHI is a novel low-voltage scanning electron microscopy based characterization technique that, in addition to capturing topographical images, also provides nanoscale resolution chemical maps by utilizing the energy distribution of emitted secondary electrons. Here, SEHI maps are exploited to assess the lateral distributions of diverse functional groups that are effected by the sterilization treatments. This information combined with a range of conventional surface analysis techniques and a cellular metabolic activity assay reveals persuasive reasons as to why low-pressure argon glow discharge should be considered for further optimization as a potential terminal sterilization method for PGS-M, a functionalized form of poly(glycerol sebacate) (PGS).
Collapse
Affiliation(s)
- Nicholas Farr
- Department of Materials Science and EngineeringSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
- Insigneo Institute for In Silico MedicineThe Pam Liversidge BuildingSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| | - Jeerawan Thanarak
- Department of Materials Science and EngineeringSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
- Insigneo Institute for In Silico MedicineThe Pam Liversidge BuildingSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| | - Jan Schäfer
- Leibniz Institute for Plasma Science and Technology (INP e.V.)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| | - Antje Quade
- Leibniz Institute for Plasma Science and Technology (INP e.V.)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| | - Frederik Claeyssens
- Department of Materials Science and EngineeringSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
- Insigneo Institute for In Silico MedicineThe Pam Liversidge BuildingSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| | - Nicola Green
- Department of Materials Science and EngineeringSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
- Insigneo Institute for In Silico MedicineThe Pam Liversidge BuildingSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| | - Cornelia Rodenburg
- Department of Materials Science and EngineeringSir Robert Hadfield BuildingUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| |
Collapse
|
43
|
Cámara-Torres M, Sinha R, Scopece P, Neubert T, Lachmann K, Patelli A, Mota C, Moroni L. Tuning Cell Behavior on 3D Scaffolds Fabricated by Atmospheric Plasma-Assisted Additive Manufacturing. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3631-3644. [PMID: 33448783 PMCID: PMC7880529 DOI: 10.1021/acsami.0c19687] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Three-dimensional (3D) scaffolds with optimum physicochemical properties are able to elicit specific cellular behaviors and guide tissue formation. However, cell-material interactions are limited in scaffolds fabricated by melt extrusion additive manufacturing (ME-AM) of synthetic polymers, and plasma treatment can be used to render the surface of the scaffolds more cell adhesive. In this study, a hybrid AM technology, which combines a ME-AM technique with an atmospheric pressure plasma jet, was employed to fabricate and plasma treat scaffolds in a single process. The organosilane monomer (3-aminopropyl)trimethoxysilane (APTMS) and a mixture of maleic anhydride and vinyltrimethoxysilane (MA-VTMOS) were used for the first time to plasma treat 3D scaffolds. APTMS treatment deposited plasma-polymerized films containing positively charged amine functional groups, while MA-VTMOS introduced negatively charged carboxyl groups on the 3D scaffolds' surface. Argon plasma activation was used as a control. All plasma treatments increased the surface wettability and protein adsorption to the surface of the scaffolds and improved cell distribution and proliferation. Notably, APTMS-treated scaffolds also allowed cell attachment by electrostatic interactions in the absence of serum. Interestingly, cell attachment and proliferation were not significantly affected by plasma treatment-induced aging. Also, while no significant differences were observed between plasma treatments in terms of gene expression, human mesenchymal stromal cells (hMSCs) could undergo osteogenic differentiation on aged scaffolds. This is probably because osteogenic differentiation is rather dependent on initial cell confluency and surface chemistry might play a secondary role.
Collapse
Affiliation(s)
- Maria Cámara-Torres
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Ravi Sinha
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Paolo Scopece
- Nadir
S.r.l., Via Torino, 155/b, 30172 Venice, Italy
| | - Thomas Neubert
- Fraunhofer
Institute for Surface Engineering and Thin Films IST, Bienroder Weg 54E, 38108 Braunschweig, Germany
| | - Kristina Lachmann
- Fraunhofer
Institute for Surface Engineering and Thin Films IST, Bienroder Weg 54E, 38108 Braunschweig, Germany
| | - Alessandro Patelli
- Department
of Physics and Astronomy, Padova University, Via Marzolo, 8, 35131 Padova, Italy
| | - Carlos Mota
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
44
|
Yilmaz HD, Cengiz U, Arslan YE, Kiran F, Ceylan A. From a plant secretion to the promising bone grafts: Cryogels of silicon-integrated quince seed mucilage by microwave-assisted sol-gel reaction. J Biosci Bioeng 2021; 131:420-433. [PMID: 33454223 DOI: 10.1016/j.jbiosc.2020.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/06/2020] [Accepted: 11/25/2020] [Indexed: 01/13/2023]
Abstract
Design and fabrication of biologically active cryogels using novel biopolymer(s) are still of great importance at regenerating bone defects such as traumatic bone injuries, maxillofacial surgery, osteomyelitis, and osteoporosis. Nowadays, plant mucilage, an herbal biomaterial, has been drawn attention by scientists due to their marvelous potential to fabricate 3-dimensional (3D) physical constructs for the field of regenerative medicine. Herein, a 3D cryogel from silicon-integrated quince seed mucilage (QSM) is constructed using microwave-assisted sol-gel reaction, characterized in-depth by attenuated total reflectance Fourier transform-infrared spectroscopy (ATR-FTIR), solid-state silicon cross-polarization magic-angle nuclear magnetic resonance (29Si-CP-MAS NMR), X-ray diffraction (XRD), thermogravimetric analysis (TGA), differential scanning calorimeter (DSC), micro-mechanical testing, porosity, and swelling tests, contact angle measurements, Brunauer-Emmet-Teller and Barret-Joyner-Halenda (BET-BJH) analysis, enzymatic biodegradation test, and field emission-scanning electron microscopy-energy dispersive X-ray spectroscopy (FE-SEM-EDX) mapping. The osteobiologic capacity of the cryogels is determined using human adipose-derived mesenchymal stem cells (hAMSCs) under in vitro conditions. Osteogenic differentiation of hAMSCs on both QSM and silica-modified QSM (Si-QSM) cryogels is analyzed by histochemistry, immunohistochemistry, and quantitative-real time (q-RT) PCR techniques. The results obtained from in vitro experiments demonstrate that the upregulation of osteogenesis-related genes in Si-QSM cryogels presents a stronger and earlier development over QSM cryogels throughout the culture period, which in turn reveals the great potential of this novel Si-incorporated QSM cryogels for bone tissue engineering applications.
Collapse
Affiliation(s)
- Hilal Deniz Yilmaz
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey
| | - Ugur Cengiz
- Surface Science Research Laboratory, Department of Chemical Engineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey.
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara 06100, Turkey
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara 06110, Turkey
| |
Collapse
|
45
|
Wu F, Su Q, Zhou L, Xu P, Dong A, Qian W. A Novel Protein Corona Characterization based on the Reflectometric Interference Spectroscopy with Silica Colloidal Crystal Films. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a20090422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
46
|
Protein Interactions at Material Surfaces. Biomed Mater 2021. [DOI: 10.1007/978-3-030-49206-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
47
|
Benko A, Medina-Cruz D, Duch J, Popiela T, Wilk S, Bińczak M, Nocuń M, Menaszek E, Geoffrion LD, Guisbiers G, Kotarba A, Webster TJ. Conductive all-carbon nanotube layers: Results on attractive physicochemical, anti-bacterial, anticancer and biocompatibility properties. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111703. [DOI: 10.1016/j.msec.2020.111703] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/14/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023]
|
48
|
Tang B, Shen X, Yang Y, Xu Z, Yi J, Yao Y, Cao M, Zhang Y, Xia H. Enhanced cellular osteogenic differentiation on CoFe 2O 4/P(VDF-TrFE) nanocomposite coatings under static magnetic field. Colloids Surf B Biointerfaces 2020; 198:111473. [PMID: 33250417 DOI: 10.1016/j.colsurfb.2020.111473] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 01/15/2023]
Abstract
Cellular responses can be regulated and manipulated through combining stimuli-responsive biomaterial with external stimulus. In this present, the magneto-responsive CoFe2O4/P(VDF-TrFE) nanocomposite coatings were designed to understand cell behaviors of preosteoblasts, as well as get insight into the underlying mechanism of osteogenic differentiation under static magnetic field (SMF). CoFe2O4/P(VDF-TrFE) nanocomposite coatings with differential magnetic property (low, medium and high magnetization) were prepared by incorporation of different mass fraction of CoFe2O4 nanoparticles (6%, 13 %, 20 %) into P(VDF-TrFE) matrix. Cell experiments indicated that all nanocomposite coatings with the assistance of SMF could promote the cell attachment, proliferation and osteogenic differentiation of MC3T3-E1 cells. Among different nanocomposite coatings, low magnetization coating (6%) showed a higher ALP activity and gene expression of Runx2, Col-I, OCN. Molecular biology assays demonstrated that the combination of nanocomposite coatings and SMF could significantly up-regulate the expression level of α2β1 integrin and p-ERK. Whereas, the addition of inhibitor U0126 down-regulated sharply the expression level of p-ERK, which indicated that cellular osteogenic differentiation of MC3T3-E1 cells was governed through α2β1 integrin-mediated MEK/ERK signaling pathways during CoFe2O4/P(VDF-TrFE) nanocomposite coatings were combined with SMF. This work provided a promising strategy to enhance cellular osteogenic differentiation through a remote-control manner, which exhibited great potential in the application of bone tissue repair and regeneration.
Collapse
Affiliation(s)
- Bolin Tang
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China.
| | - Xiaojun Shen
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Yaru Yang
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Zhi Xu
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China.
| | - Jie Yi
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Yongbo Yao
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Miao Cao
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China; Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Yalin Zhang
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Hongqin Xia
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, China
| |
Collapse
|
49
|
Mahdavi R, Belgheisi G, Haghbin-Nazarpak M, Omidi M, Khojasteh A, Solati-Hashjin M. Bone tissue engineering gelatin-hydroxyapatite/graphene oxide scaffolds with the ability to release vitamin D: fabrication, characterization, and in vitro study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:97. [PMID: 33135110 DOI: 10.1007/s10856-020-06430-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/24/2020] [Indexed: 06/11/2023]
Abstract
Developing smart scaffolds with drug release capability is one of the main approaches to bone tissue engineering. The current study involves the fabrication of novel gelatin (G)-hydroxyapatite (HA)-/vitamin D (VD)-loaded graphene oxide (GO) scaffolds with different concentrations through solvent-casting method. Characterizations confirmed the successful synthesis of HA and GO, and VD was loaded in GO with 36.87 ± 4.87% encapsulation efficiency. Physicochemical characterizations showed that the scaffold containing 1% VD-loaded GO had the best mechanical properties and its porosity percentage and density was in the range of natural spongy bone. All scaffolds were degraded after 1-month, subjecting to phosphate buffer saline. The release profile of VD did not match any mathematical kinetics model, porosities and the degradation rate of the scaffolds were dominant controlling factors of release behavior. Studies on the bioactivity of scaffolds immersed in simulated body fluid indicated that VD and HA could encourage the formation of secondary apatite crystals in vitro. Buccal fat pad-derived stem cells (BFPSCs) were seeded on the scaffolds, MTT assay, alkaline phosphatase activity as an indicator of osteoconductivity, and cell adhesion were conducted in order to evaluate in vitro biological responses. All scaffolds highly supported cell adhesion, MTT assay indicated better cell viability in 0.5% VD-loaded GO containing scaffold, and the scaffold enriched with 2% VD-loaded GO performed the most ALP activity. The results demonstrated the potential of these scaffolds to induce bone regeneration. Developing smart scaffolds with drug release capability is one of the main approaches to bone tissue engineering. The current study involves the fabrication of novel gelatin (G)-hydroxyapatite (HA)-/vitamin D (VD)-loaded graphene oxide (GO) scaffolds with different concentrations through solvent-casting method. Characterizations confirmed the successful synthesis of HA and GO, and VD was loaded in GO with 36.87 ± 4.87% encapsulation efficiency. Physicochemical characterizations showed that the scaffold containing 1% VD-loaded GO had the best mechanical properties and its porosity percentage and density was in the range of natural spongy bone. All scaffolds were degraded after 1-month, subjecting to phosphate buffer saline. The release profile of VD did not match any mathematical kinetics model, porosities and the degradation rate of the scaffolds were dominant controlling factors of release behavior. Studies on the bioactivity of scaffolds immersed in simulated body fluid indicated that VD and HA could encourage the formation of secondary apatite crystals in vitro. Buccal fat pad-derived stem cells (BFPSCs) were seeded on the scaffolds, MTT assay, alkaline phosphatase activity as an indicator of osteoconductivity, and cell adhesion were conducted in order to evaluate in vitro biological responses. All scaffolds highly supported cell adhesion, MTT assay indicated better cell viability in 0.5% VD-loaded GO containing scaffold, and the scaffold enriched with 2% VD-loaded GO performed the most ALP activity. The results demonstrated the potential of these scaffolds to induce bone regeneration.
Collapse
Affiliation(s)
- Reza Mahdavi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Ghazal Belgheisi
- Department of Biomedical Engineering, Biofabrication Laboratory, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Masoumeh Haghbin-Nazarpak
- New Technologies Research Center (NTRC), Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Meisam Omidi
- Protein Research Centre, Shahid Beheshti University, GC, Velenjak Tehran, Iran
| | - Arash Khojasteh
- Department of Oral and Maxillofacial Surgery, School of Advanced Technologies in Medicine, Taleghani University Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mehran Solati-Hashjin
- Department of Biomedical Engineering, Biofabrication Laboratory, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| |
Collapse
|
50
|
Enhanced Osteogenic Differentiation of Human Mesenchymal Stem Cells on Amine-Functionalized Titanium Using Humidified Ammonia Supplied Nonthermal Atmospheric Pressure Plasma. Int J Mol Sci 2020; 21:ijms21176085. [PMID: 32846976 PMCID: PMC7503675 DOI: 10.3390/ijms21176085] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
The surface molecular chemistry, such as amine functionality, of biomaterials plays a crucial role in the osteogenic activity of relevant cells and tissues during hard tissue regeneration. Here, we examined the possibilities of creating amine functionalities on the surface of titanium by using the nonthermal atmospheric pressure plasma jet (NTAPPJ) method with humidified ammonia, and the effects on human mesenchymal stem cell (hMSC) were investigated. Titanium samples were subjected to NTAPPJ treatments using nitrogen (N-P), air (A-P), or humidified ammonia (NA-P) as the plasma gas, while control (C-P) samples were not subjected to plasma treatment. After plasma exposure, all treatment groups showed increased hydrophilicity and had more attached cells than the C-P. Among the plasma-treated samples, the A-P and NA-P showed surface oxygen functionalities and exhibited greater cell proliferation than the C-P and N-P. The NA-P additionally showed surface amine-related functionalities and exhibited a higher level of alkaline phosphatase activity and osteocalcin expression than the other samples. The results can be explained by increases in fibronectin absorption and focal adhesion kinase gene expression on the NA-P samples. These findings suggest that NTAPPJ technology with humidified ammonia as a gas source has clinical potential for hard tissue generation.
Collapse
|