1
|
Çalık M, Unal S, Alemdag B, Gündüz O, Tekkeşin MS, Ozen B, Islek I, Kuru L, Agrali OB. Evaluation of the bone regenerative effect of glycogen synthase kinase 3 antagonist Tideglusib carried by different scaffolds on rat calvarial defects. Int J Biol Macromol 2025; 292:139350. [PMID: 39743077 DOI: 10.1016/j.ijbiomac.2024.139350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/16/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
The aim was to explore the efficiency of Tideglusib in bone tissue healing by carrying it with different scaffolds on rat calvarial lesions. Twentyfour male Dawley rats were utilized. Two bone defects of 5 mm in diameter were formed (n = 8). Groups constituted negative control, collagen sponge + Tideglusib (CT), bacterial cellulose carrier (BC), bacterial cellulose carrier + Tideglusib (BC + T), PCL/Gel nanocarrier (Nano) and PCL/Gel + Tideglusib (Nano+T). After four week, histomorphometric and immunohistochemistry investigations were performed. Pairwise comparisons by means of the new bone formation (NBF) effect of Tideglusib demonstrated a significant difference between the control and the Nano+T groups solely (p < 0.05). BC group demonstrated reduced NBF in comparison to the CT group (p < 0.05), Nano group (p < 0.01) and Nano+T group (p < 0.01). Similarly, the BC + T group exhibited a diminished rate of NBF in comparison to both the Nano (p < 0.01) and Nano+T groups (p < 0.01). Type I collagen expression decreased in the BC group (p < 0.05) and BC + T group (p < 0.05) relative to the control. Axin2 expression was increased in the Nano+T group (p < 0.05) compared to the control. Within the limits, Tideglusib delivered with a nanocarrier containing PCL/Gel may have favorable impact on bone regeneration. However, the impact may vary with different carrier.
Collapse
Affiliation(s)
- Mümin Çalık
- Department of Periodontology, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkiye
| | - Semra Unal
- Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, Istanbul, Turkiye.
| | - Berna Alemdag
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkiye
| | - Oğuzhan Gündüz
- Center for Nanotechnology and Biomaterials Application and Research, Marmara University, Istanbul 34722, Turkiye; Department of Metallurgy and Material Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkiye.
| | - Merva Soluk Tekkeşin
- Department of Oral Pathology, Faculty of Dentistry, Istanbul University, Istanbul, Turkiye; Department of Tumour Pathology, Institute of Oncology, Istanbul University, Istanbul, Turkiye.
| | - Bensu Ozen
- Department of Periodontology, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkiye
| | - Ipek Islek
- Department of Periodontology, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkiye
| | - Leyla Kuru
- Department of Periodontology, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkiye.
| | - Omer Birkan Agrali
- Department of Periodontology, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkiye.
| |
Collapse
|
2
|
Beder M, Yemenoglu H, Bostan SA, Kose O, Karakas SM, Mercantepe T, Yılmaz A, Tumkaya L. Investigation of the preventive effect of methylsulfonylmethane on alveolar bone loss and oxidative stress in a rat model of periodontitis. BMC Oral Health 2025; 25:78. [PMID: 39819670 PMCID: PMC11740458 DOI: 10.1186/s12903-025-05447-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: 11/11/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND To investigate the preventive efficacy of methylsulfonylmethane (MSM) on alveolar bone destruction in rats with periodontitis. METHODS Twenty-four male Sprague-Dawley rats were randomly divided into three groups: control, experimental periodontitis (Ep), and Ep-MSM. Periodontitis was induced by placing 4.0 silk sutures in the subparamarginal position on the necks of the mandibular first molars and applying the suture for 5 weeks. The Ep-MSM group was given 500 mg/body weight/day MSM intraperitoneally for 35 days. At the end of the study, bilateral mandibular samples were taken. Periodontal bone loss was measured through histologic sections. Histomorphometric and immunohistochemical (receptor activator of nuclear factor kappa B ligand (RANKL), osteoprotegerin (OPG)) evaluations were performed on right mandibular tissue samples, and biochemical (interleukin (IL)-1 beta (β)/IL-10, malondialdehyde (MDA), glutathione (GSH), oxidative stress index (OSI)) evaluations were performed on left mandibular tissue samples. RESULTS No significant difference was found between the groups in IL-1β and IL-1β/IL-10 values (p > 0.05). A significant decrease in IL-10 levels was observed in the Ep-MSM and Ep groups compared with the control group (p < 0.05). MDA levels significantly increased in the Ep and Ep-MSM groups compared with the control group, and GSH levels significantly decreased in the Ep group compared with the other groups (p < 0.05). OSI values were significantly higher only in the Ep group (p < 0.05). RANKL levels showed a significant increase in the Ep group compared with the other groups. OPG levels were significantly increased only in the Ep-MSM group (p < 0.05). CONCLUSIONS The results of this study may suggest that MSM has preventive effects on alveolar bone loss and oxidative stress.
Collapse
Affiliation(s)
- Melek Beder
- Department of Periodontology, Faculty of Dentistry, Recep Tayyip Erdoğan University, Rize, 53020, Turkey.
| | - Hatice Yemenoglu
- Department of Periodontology, Faculty of Dentistry, Recep Tayyip Erdoğan University, Rize, 53020, Turkey
| | - Semih Alperen Bostan
- Department of Periodontology, Faculty of Dentistry, Recep Tayyip Erdoğan University, Rize, 53020, Turkey
| | - Oğuz Kose
- Department of Periodontology, Faculty of Dentistry, Recep Tayyip Erdoğan University, Rize, 53020, Turkey
| | - Sibel Mataracı Karakas
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Adnan Yılmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
3
|
Choudhury S, Madhu Krishna M, Sen D, Ghosh S, Basak P, Das A. 3D Porous Polymer Scaffold-Conjugated KGF-Mimetic Peptide Promotes Functional Skin Regeneration in Chronic Diabetic Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37418-37434. [PMID: 38980153 DOI: 10.1021/acsami.4c02633] [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: 07/10/2024]
Abstract
The re-epithelialization process gets severely dysregulated in chronic nonhealing diabetic foot ulcers/wounds. Keratinocyte growth factor (KGF or FGF-7) is the major modulator of the re-epithelialization process, which regulates the physiological phenotypes of cutaneous keratinocytes. The existing therapeutic strategies of growth factor administration have several limitations. To overcome these, we have designed a KGF-mimetic peptide (KGFp, 13mer) based on the receptor interaction sites in murine KGF. KGFp enhanced migration and transdifferentiation of mouse bone marrow-derived MSCs toward keratinocyte-like cells (KLCs). A significant increase in the expression of skin-specific markers Bnc1 (28.5-fold), Ck5 (14.6-fold), Ck14 (26.1-fold), Ck10 (187.7-fold), and epithelial markers EpCam (23.3-fold) and Cdh1 (64.2-fold) was associated with the activation of ERK1/2 and STAT3 molecular signaling in the KLCs. Further, to enhance the stability of KGFp in the wound microenvironment, it was conjugated to biocompatible 3D porous polymer scaffolds without compromising its active binding sites followed by chemical characterization using Fourier transform infrared spectroscopy, field-emission scanning electron microscopy, dynamic mechanical analysis, and thermogravimetry. In vitro evaluation of the KGFp-conjugated 3D polymer scaffolds revealed its potential for transdifferentiation of MSCs into KLCs. Transplantation of allogeneic MSCGFP using KGFp-conjugated 3D polymer scaffolds in chronic nonhealing type 2 diabetic wounds (db/db transgenic, 50-52 weeks old male mice) significantly enhanced re-epithelialization-mediated wound closure rate (79.3%) as compared to the control groups (Untransplanted -22.4%, MSCGFP-3D polymer scaffold -38.5%). Thus, KGFp-conjugated 3D porous polymer scaffolds drive the fate of the MSCs toward keratinocytes that may serve as potential stem cell delivery platform technology for tissue engineering and transplantation.
Collapse
Affiliation(s)
- Subholakshmi Choudhury
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Mangali Madhu Krishna
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
- Department of Polymers and Functional Materials, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Debanjan Sen
- BCDA College of Pharmacy and Technology, Hridaypur, Kolkata 700127, West Bengal, India
| | - Subhash Ghosh
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
- Department of Organic Synthesis and Process Chemistry, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Pratyay Basak
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
- Department of Polymers and Functional Materials, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
4
|
Soheilmoghaddam F, Hezaveh H, Rumble M, Cooper-White JJ. Driving Osteocytogenesis from Mesenchymal Stem Cells in Osteon-like Biomimetic Nanofibrous Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39044386 DOI: 10.1021/acsami.3c14785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The treatment of critical-sized bone defects caused by tumor removal, skeletal injuries, or infections continues to pose a major clinical challenge. A popular potential alternative solution to autologous bone grafts is a tissue-engineered approach that utilizes the combination of mesenchymal stromal/stem cells (MSCs) with synthetic biomaterial scaffolds. This approach aims to support new bone formation by mimicking many of the biochemical and biophysical cues present within native bone. Regrettably, osteocyte cells, crucial for bone maturation and homeostasis, are rarely produced within MSC-seeded scaffolds, thereby restricting the development of fully mature cortical bone from these synthetic implants. In this work, we have constructed a multimodal scaffold by combining electrospun poly(lactic-co-glycolic acid) (PLGA) fibrous scaffolds with poly(ethylene glycol) (PEG)-based hydrogels that mimic the functional unit of cortical bone, osteon (osteon-mimetic) scaffolds. These scaffolds were decorated with a novel bone morphogenic protein-6 (BMP6) peptide (BMP6p) after our findings revealed that the BMP6p drives higher levels of Smad signaling than the full-length protein counterpart, soluble or when bound to the PEG hydrogel backbone. We show that our osteon-mimetic scaffolds, in presenting concentric layers of BMP6p-PEG hydrogel overlaid on MSC-seeded PLGA nanofibers, promoted the rapid formation of osteocyte-like cells with a phenotypic dendritic morphology, producing early osteocyte markers, including E11/gp38 (E11). Maturation of these osteocyte-like cells was further confirmed by the observation of significant dentin matrix protein 1 (DMP1) throughout our bilayered scaffolds after 3 weeks, even when cultured in a medium without dexamethasone (DEX) or any other osteogenic supplements. These results demonstrate that these osteon-mimetic scaffolds, in presenting biochemical and topographical cues reminiscent of the forming osteon, can drive the formation of osteocyte-like cells in vitro from hBMSCs without the need for any osteogenic factor media supplementation.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory, The Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Hadi Hezaveh
- Tissue Engineering and Microfluidics Laboratory, The Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
| | - Madeleine Rumble
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Justin J Cooper-White
- Tissue Engineering and Microfluidics Laboratory, The Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
5
|
Wu X, Ni S, Dai T, Li J, Shao F, Liu C, Wang J, Fan S, Tan Y, Zhang L, Jiang Q, Zhao H. Biomineralized tetramethylpyrazine-loaded PCL/gelatin nanofibrous membrane promotes vascularization and bone regeneration of rat cranium defects. J Nanobiotechnology 2023; 21:423. [PMID: 37964381 PMCID: PMC10644548 DOI: 10.1186/s12951-023-02155-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Conventional electrospinning produces nanofibers with smooth surfaces that limit biomineralization ability. To overcome this disadvantage, we fabricated a tetramethylpyrazine (TMP)-loaded matrix-mimicking biomineralization in PCL/Gelatin composite electrospun membranes with bubble-shaped nanofibrous structures. PCL/Gelatin membranes (PG), PCL/Gelatin membranes containing biomineralized hydroxyapatite (HA) (PGH), and PCL/Gelatin membranes containing biomineralized HA and loaded TMP (PGHT) were tested. In vitro results indicated that the bubble-shaped nanofibrous surface increased the surface roughness of the nanofibers and promoted mineralization. Furthermore, sustained-release TMP had an excellent drug release efficiency. Initially released vigorously, it reached stabilization at day 7, and the slow-release rate stabilized at 61.0 ± 1.8% at 28 days. All membranes revealed an intact cytoskeleton, cell viability, and superior adhesion and proliferation when stained with Ghost Pen Cyclic Peptide, CCK-8, cell adhesion, and EdU. In PGHT membranes, the osteogenic and vascularized gene expression of BMSCs and human vascular endothelial cells was significantly upregulated compared with that in other groups, indicating the PGHT membranes exhibited an effective vascularization role. Subsequently, the membranes were implanted in a rat cranium defect model for 4 and 8 weeks. Micro-CT and histological analysis results showed that the PGHT membranes had better bone regenerative patterns. Additionally, the levels of CD31 and VEGF significantly increased in the PGHT membrane compared with those in other membranes. Thus, PGHT membranes could accelerate the repair of cranium defects in vivo via HA and TMP synergistic effects.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Su Ni
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Ting Dai
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Jingyan Li
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Fang Shao
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Chun Liu
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Jiafeng Wang
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Shijie Fan
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Yadong Tan
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
| | - Linxiang Zhang
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China
- Orthopedic Center of Nanjing Jiangbei Hospital, Nanjiang, 210048, China
| | - Qiting Jiang
- Orthopedic Center of Nanjing Jiangbei Hospital, Nanjiang, 210048, China.
| | - Hongbin Zhao
- Laboratory of 3D Printing and Regeneration Medicine, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213164, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213164, China.
| |
Collapse
|
6
|
Pei B, Hu M, Wu X, Lu D, Zhang S, Zhang L, Wu S. Investigations into the effects of scaffold microstructure on slow-release system with bioactive factors for bone repair. Front Bioeng Biotechnol 2023; 11:1230682. [PMID: 37781533 PMCID: PMC10537235 DOI: 10.3389/fbioe.2023.1230682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
In recent years, bone tissue engineering (BTE) has played an essential role in the repair of bone tissue defects. Although bioactive factors as one component of BTE have great potential to effectively promote cell differentiation and bone regeneration, they are usually not used alone due to their short effective half-lives, high concentrations, etc. The release rate of bioactive factors could be controlled by loading them into scaffolds, and the scaffold microstructure has been shown to significantly influence release rates of bioactive factors. Therefore, this review attempted to investigate how the scaffold microstructure affected the release rate of bioactive factors, in which the variables included pore size, pore shape and porosity. The loading nature and the releasing mechanism of bioactive factors were also summarized. The main conclusions were achieved as follows: i) The pore shapes in the scaffold may have had no apparent effect on the release of bioactive factors but significantly affected mechanical properties of the scaffolds; ii) The pore size of about 400 μm in the scaffold may be more conducive to controlling the release of bioactive factors to promote bone formation; iii) The porosity of scaffolds may be positively correlated with the release rate, and the porosity of 70%-80% may be better to control the release rate. This review indicates that a slow-release system with proper scaffold microstructure control could be a tremendous inspiration for developing new treatment strategies for bone disease. It is anticipated to eventually be developed into clinical applications to tackle treatment-related issues effectively.
Collapse
Affiliation(s)
- Baoqing Pei
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Mengyuan Hu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xueqing Wu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Da Lu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shijia Zhang
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Le Zhang
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shuqin Wu
- School of Big Data and Information, Shanxi College of Technology, Taiyuan, Shanxi, China
| |
Collapse
|
7
|
Valizadeh N, Salehi R, Aghazadeh M, Alipour M, Sadeghzadeh H, Mahkam M. Enhanced osteogenic differentiation and mineralization of human dental pulp stem cells using Prunus amygdalus amara (bitter almond) incorporated nanofibrous scaffold. J Mech Behav Biomed Mater 2023; 142:105790. [PMID: 37104899 DOI: 10.1016/j.jmbbm.2023.105790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 04/29/2023]
Abstract
Polyphenol extracts derived from plants are expected to have enhanced osteoblast proliferation and differentiation ability, which has gained much attention in tissue engineering applications. Herein, for the first time, we investigate the effects of Prunus amygdalus amara (bitter almond) (BA) extract loaded on poly (ε-caprolactone) (PCL)/gelatin (Gt) nanofibrous scaffolds on the osteoblast differentiation of human dental pulp stem cells (DPSCs). In this regard, BA (0, 5, 10, and 15% wt)-loaded PCL/Gt nanofibrous scaffolds were prepared by electrospinning with fiber diameters in the range of around 237-276 nm. Morphology, composition, porosity, hydrophilicity, and mechanical properties of the scaffolds were examined by FESEM, ATR-FTIR spectroscopy, BET, contact angle, and tensile tests, respectively. It was found that the addition of BA improved the tensile strength (up to 6.1 times), Young's modulus (up to 3 times), and strain at break (up to 3.2 times) compared to the neat PCL/Gt nanofibers. Evaluations of cell attachment, spreading, and proliferation were done by FESEM observation and MTT assay. Cytocompatibility studies support the biocompatible nature of BA loaded PCL/Gt scaffolds and free BA by demonstrating cell viability of more than 100% in all groups. The results of alkaline phosphatase activity and Alizarin Red assay revealed that osteogenic activity levels of BA loaded PCL/Gt scaffolds and free BA were significantly increased compared to the control group (p < 0.05, p < 0.01, p < 0.001). QRT-PCR results demonstrated that BA loaded PCL/Gt scaffolds and free BA led to a significant increase in osteoblast differentiation of DPSCs through the upregulation of osteogenic related genes compared to the control group (p < 0.05). Based on results, incorporation of BA extract in PCL/Gt scaffolds exhibited synergistic effects on the adhesion, proliferation, and osteogenesis differentiation of hDPSCs and was therefore assumed to be a favorable scaffold for bone tissue engineering applications.
Collapse
Affiliation(s)
- Nasrin Valizadeh
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Marziyeh Aghazadeh
- Stem Cell Research Center and Department of Oral Medicine, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Mahkam
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran.
| |
Collapse
|
8
|
Karahaliloglu Z, Ercan B, Hazer B. Impregnation of polyethylene terephthalate (PET) grafts with BMP-2 loaded functional nanoparticles for reconstruction of anterior cruciate ligament. J Microencapsul 2023; 40:197-215. [PMID: 36881484 DOI: 10.1080/02652048.2023.2188940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Current artificial ligaments based on polyethylene terephthalate (PET) are associated with some disadvantages due to their hydrophobicity and low biocompatibility. In this study, we aimed to modify the surface of PET using polyethylene glycol (PEG)-terminated polystyrene (PS)-linoleic nanoparticles (PLinaS-g-PEG-NPs). We accomplished that BMP-2 in two different concentrations encapsulated in nanoparticles with an efficiency of 99.71 ± 1.5 and 99.95 ± 2.8%. While the dynamic contact angle of plain PET surface reduced from 116° to 115° after a measurement periods of 10 s, that of PLinaS-g-PEG-NPs modified PET from 80° to 17.5° within 0.35 s. According to in vitro BMP2 release study, BMP-2 was released 13.12 ± 1.76% and 45.47 ± 1.78% from 0.05 and 0.1BMP2-PLinaS-g-PEG-NPs modified PET at the end of 20 days, respectively. Findings from this study revealed that BMP2-PLinaS-g-PEG-NPs has a great potential to improve the artificial PET ligaments, and could be effectively applied for ACL reconstruction.
Collapse
Affiliation(s)
| | - Batur Ercan
- Department of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, Ankara, Turkey
- Biomedical Engineering Program, Middle East Technical University, Çankaya, Ankara, Turkey
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Çankaya, Ankara, Turkey
| | - Baki Hazer
- Department of Aircraft Airframe Engine Maintenance, Kapadokya University, Ürgüp, Nevsehir, Turkey
- Department of Chemistry, Bulent Ecevit University, Zonguldak, Turkey
- Department of Nanotechnology Engineering, Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
9
|
Anjum S, Rahman F, Pandey P, Arya DK, Alam M, Rajinikanth PS, Ao Q. Electrospun Biomimetic Nanofibrous Scaffolds: A Promising Prospect for Bone Tissue Engineering and Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23169206. [PMID: 36012473 PMCID: PMC9408902 DOI: 10.3390/ijms23169206] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Skeletal-related disorders such as arthritis, bone cancer, osteosarcoma, and osteoarthritis are among the most common reasons for mortality in humans at present. Nanostructured scaffolds have been discovered to be more efficient for bone regeneration than macro/micro-sized scaffolds because they sufficiently permit cell adhesion, proliferation, and chemical transformation. Nanofibrous scaffolds mimicking artificial extracellular matrices provide a natural environment for tissue regeneration owing to their large surface area, high porosity, and appreciable drug loading capacity. Here, we review recent progress and possible future prospective electrospun nanofibrous scaffolds for bone tissue engineering. Electrospun nanofibrous scaffolds have demonstrated promising potential in bone tissue regeneration using a variety of nanomaterials. This review focused on the crucial role of electrospun nanofibrous scaffolds in biological applications, including drug/growth factor delivery to bone tissue regeneration. Natural and synthetic polymeric nanofibrous scaffolds are extensively inspected to regenerate bone tissue. We focused mainly on the significant impact of nanofibrous composite scaffolds on cell adhesion and function, and different composites of organic/inorganic nanoparticles with nanofiber scaffolds. This analysis provides an overview of nanofibrous scaffold-based bone regeneration strategies; however, the same concepts can be applied to other organ and tissue regeneration tactics.
Collapse
Affiliation(s)
- Shabnam Anjum
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China
| | - Farheen Rahman
- Department of Applied Chemistry, Zakir Husain College of Engineering & Technology, Aligarh Muslim University, Aligarh 202002, India
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Mahmood Alam
- Department of Clinical Medicine, China Medical University, Shenyang 110122, China
| | - Paruvathanahalli Siddalingam Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
- Correspondence: (P.S.R.); (Q.A.)
| | - Qiang Ao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
- Correspondence: (P.S.R.); (Q.A.)
| |
Collapse
|
10
|
Vijayan AN, Solaimuthu A, Murali P, Gopi J, Y MT, R AP, Korrapati PS. Decorin mediated biomimetic PCL-gelatin nano-framework to impede scarring. Int J Biol Macromol 2022; 219:907-918. [PMID: 35952816 DOI: 10.1016/j.ijbiomac.2022.08.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/05/2022]
Abstract
Scars occur as a result of fibrosis after tissue damage or surgery and reports suggest that excessive Transforming growth factor-β (TGF-β) activity during the process of wound healing leads to progressive fibrosis. Decorin is an extracellular matrix (ECM) protein which regulates collagen fibrillogenesis. However, targeted delivery and effective protein therapy remains a challenge owing to degradation byproteases. Hence, we aimed to deliver Decorin in a sustainable mode for the reduction of TGF-β levels and subsequent scar formation. Herein, we have fabricated PCL-Gelatin bio-mimetic scaffolds to optimize the bio-activity and provide localized delivery of recombinant Decorin. The degradation and drug release patterns reveals that this biomaterial is biodegradable and offers sustained release of the recombinant Decorin. Decorin loaded nanofiber displayed lower adhesion and proliferation rates in in-vitro conditions. Moreover, Decorin loaded scaffolds demonstrated morphological changes in cells, specifically targeting the myofibroblast. The expression of TGF-β was also scrutinized to understand the effect of Decorin loaded nanofibers. Besides, in the in-vitro fibrotic model, Decorin loaded nanofibers efficiently reduced the expression of ECM related proteins. Therefore, we report the sustained delivery of the recombinant Decorin from nanofiber dressing to potentially obstruct scar formation during the process of wound healing.
Collapse
Affiliation(s)
- Ane Nishitha Vijayan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anbuthiruselvan Solaimuthu
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Padmaja Murali
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Janani Gopi
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Madhan Teja Y
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Akshaya Priya R
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Purna Sai Korrapati
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
11
|
Backes EH, Harb SV, Beatrice CAG, Shimomura KMB, Passador FR, Costa LC, Pessan LA. Polycaprolactone usage in additive manufacturing strategies for tissue engineering applications: A review. J Biomed Mater Res B Appl Biomater 2021; 110:1479-1503. [PMID: 34918463 DOI: 10.1002/jbm.b.34997] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 08/02/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022]
Abstract
Polycaprolactone (PCL) has been extensively applied on tissue engineering because of its low-melting temperature, good processability, biodegradability, biocompatibility, mechanical resistance, and relatively low cost. The advance of additive manufacturing (AM) technologies in the past decade have boosted the fabrication of customized PCL products, with shorter processing time and absence of material waste. In this context, this review focuses on the use of AM techniques to produce PCL scaffolds for various tissue engineering applications, including bone, muscle, cartilage, skin, and cardiovascular tissue regeneration. The search for optimized geometry, porosity, interconnectivity, controlled degradation rate, and tailored mechanical properties are explored as a tool for enhancing PCL biocompatibility and bioactivity. In addition, rheological and thermal behavior is discussed in terms of filament and scaffold production. Finally, a roadmap for future research is outlined, including the combination of PCL struts with cell-laden hydrogels and 4D printing.
Collapse
Affiliation(s)
- Eduardo Henrique Backes
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| | - Samarah Vargas Harb
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| | - Cesar Augusto Gonçalves Beatrice
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| | - Kawany Munique Boriolo Shimomura
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| | | | - Lidiane Cristina Costa
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| | - Luiz Antonio Pessan
- Materials Engineering Department, Graduate Program in Materials Science and Engineering, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
12
|
Sun CK, Weng PW, Chang JZC, Lin YW, Tsuang FY, Lin FH, Tsai TH, Sun JS. Metformin-Incorporated Gelatin/Hydroxyapatite Nanofiber Scaffold for Bone Regeneration. Tissue Eng Part A 2021; 28:1-12. [PMID: 33971745 DOI: 10.1089/ten.tea.2021.0038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tissue engineering and regenerative medicine has gradually evolved as a promising therapeutic strategy to the modern health care of aging and diseased population. In this study, we developed a novel nanofibrous scaffold and verified its application in the critical bone defect regeneration. The metformin-incorporated nano-gelatin/hydroxyapatite fibers (NGF) was produced by electrospinning, cross-linked, and then characterized by X-ray powder diffractometer and Fourier-transform infrared spectroscopy. Cytotoxicity, cell adhesion, cell differentiation, and quantitative osteogenic gene and protein expression were analyzed by bone marrow stem cells (BMSCs) from rat. Rat forearm critical bone defect model was performed for the in vivo study. The NGF were characterized by their porous structures with proper interconnectivity without significant cytotoxic effects; the adhesion of BMSCs on the NGF could be enhanced. The osteogenic gene and protein expression were upregulated. Postimplantation, the new regenerated bone in bone defect was well demonstrated in the NGF samples. We demonstrated that the metformin-incorporated NGF greatly improved healing potential on the critical-size bone defect. Although metformin-incorporated NGF had advantageous effectiveness during bone regeneration, further validation is required before it can be applied to clinical applications. Impact statement Bone is the structure that supports the rest of the human body. Critical-size bone defect hinders the regeneration of damaged bone tissues and compromises the mechanical strength of the skeletal system. Characterized by their porous structures with proper interconnectivity, the electrospinning nano-gelatin/hydroxyapatite fibrous scaffold developed in this study can greatly improve the healing potential on the critical-size bone defect. Further validation can validate its potential clinical applications.
Collapse
Affiliation(s)
- Chung-Kai Sun
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (Republic of China)
| | - Pei-Wei Weng
- Department of Orthopaedics, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei, Taiwan (Republic of China)
| | - Jenny Zwei-Chieng Chang
- School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan (Republic of China)
| | - Yi-Wen Lin
- Institute of Biomedical Engineering, College of Medicine, National Taiwan University, Taipei, Taiwan.,College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Fon-Yih Tsuang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan (Republic of China)
| | - Feng-Huei Lin
- Institute of Biomedical Engineering, College of Medicine, National Taiwan University, Taipei, Taiwan.,College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (Republic of China)
| | - Jui-Sheng Sun
- Department of Orthopedic Surgery, College of Medicine, China Medical University, Taichung, Taiwan (Republic of China).,Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan (Republic of China)
| |
Collapse
|
13
|
Pharmaceutical electrospinning and 3D printing scaffold design for bone regeneration. Adv Drug Deliv Rev 2021; 174:504-534. [PMID: 33991588 DOI: 10.1016/j.addr.2021.05.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Bone regenerative engineering provides a great platform for bone tissue regeneration covering cells, growth factors and other dynamic forces for fabricating scaffolds. Diversified biomaterials and their fabrication methods have emerged for fabricating patient specific bioactive scaffolds with controlled microstructures for bridging complex bone defects. The goal of this review is to summarize the points of scaffold design as well as applications for bone regeneration based on both electrospinning and 3D bioprinting. It first briefly introduces biological characteristics of bone regeneration and summarizes the applications of different types of material and the considerations for bone regeneration including polymers, ceramics, metals and composites. We then discuss electrospinning nanofibrous scaffold applied for the bone regenerative engineering with various properties, components and structures. Meanwhile, diverse design in the 3D bioprinting scaffolds for osteogenesis especially in the role of drug and bioactive factors delivery are assembled. Finally, we discuss challenges and future prospects in the development of electrospinning and 3D bioprinting for osteogenesis and prominent strategies and directions in future.
Collapse
|
14
|
Siddiqui N, Kishori B, Rao S, Anjum M, Hemanth V, Das S, Jabbari E. Electropsun Polycaprolactone Fibres in Bone Tissue Engineering: A Review. Mol Biotechnol 2021; 63:363-388. [PMID: 33689142 DOI: 10.1007/s12033-021-00311-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 02/20/2021] [Indexed: 01/17/2023]
Abstract
Regeneration of bone tissue requires novel load bearing, biocompatible materials that support adhesion, spreading, proliferation, differentiation, mineralization, ECM production and maturation of bone-forming cells. Polycaprolactone (PCL) has many advantages as a biomaterial for scaffold production including tuneable biodegradation, relatively high mechanical toughness at physiological temperature. Electrospinning produces nanofibrous porous matrices that mimic many properties of natural tissue extracellular matrix with regard to surface area, porosity and fibre alignment. The biocompatibility and hydrophilicity of PCL nanofibres can be improved by combining PCL with other biomaterials to form composite scaffolds for bone regeneration. This work reviews the most recent research on synthesis, characterization and cellular response to nanofibrous PCL scaffolds and the composites of PCL with other natural and synthetic materials for bone tissue engineering.
Collapse
Affiliation(s)
- Nadeem Siddiqui
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh, India.
| | - Braja Kishori
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh, India
| | - Saranya Rao
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh, India
| | - Mohammad Anjum
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh, India
| | - Venkata Hemanth
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhra Pradesh, India
| | - Swati Das
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Esmaiel Jabbari
- Biomaterials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208, USA
| |
Collapse
|
15
|
Raj Preeth D, Saravanan S, Shairam M, Selvakumar N, Selestin Raja I, Dhanasekaran A, Vimalraj S, Rajalakshmi S. Bioactive Zinc(II) complex incorporated PCL/gelatin electrospun nanofiber enhanced bone tissue regeneration. Eur J Pharm Sci 2021; 160:105768. [PMID: 33607242 DOI: 10.1016/j.ejps.2021.105768] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/25/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022]
Abstract
Bone tissue regeneration is augmented by biocompatible nanofiber scaffolds, that supports reliable and enhanced bone formation. Zinc is an essential mineral that is vital for routine skeletal growth and it emerges to be able to improve bone regeneration. Phytochemicals, particularly flavonoids have achieved prominent interest for their therapeutic ability, they have demonstrated promising effects on bone by encouraging osteoblastogenesis, which finally leads to bone formation. In this study, we have synthesized bioactive zinc(II) quercetin complex material and used for nanofibers scaffold fabrication to enhance bone tissue regeneration property. Two derivatives of zinc(II) quercetin complexes [(Zn(quercetin) (H2O)2) (Zn+Q), and Zn(quercetin)(phenanthroline) (Zn+Q(PHt)) have been synthesized and characterized using UV-Visible spectrophotometer and Fourier Transform-IR spectroscopy. The UV-Visible absorption and IR spectra prove the B-ring chelation of the flavonoid quercetin to zinc(II) rather C-ring chelation. The potential ability of the above synthesized metal complexes on osteogenesis and angiogenesis have been studied. Besides the bioactivity of the metal complexes, the control quercetin has also been examined. The chick embryo chorioallantoic membrane (CAM) assay demonstrated that the angiogenic parameters were increased by the (Zn+Q(PHt)) complex. Amongst, (Zn+Q(PHt)) complex showed significant activity and thereby this complex has been further examined for the bone tissue activity by incorporating the complex into a nanofiber through electrospinning method. At the molecular level, Runx2, mRNA and protein, ALP and type 1 collagen mRNAs, and osteoblast-specific microRNA, pre-mir-15b were examined using real time RT-PCR and Western blot assay. Histology studies showed that the (PCL/gelatin/Zn+Q(PHt)) was biocompatibility in-ovo. Overall, the present study showed that quercetin-zinc complex (Zn+Q(PHt)) incorporated into PCL/gelatin nanofiber can act as a pharmacological agent for treating bone associated defects and promote bone regeneration.
Collapse
Affiliation(s)
- Desingh Raj Preeth
- Chemical Biology and Nanobiotechnology Laboratory, AU-KBC Research Centre, Anna University, MIT, Campus, Chrompet, Chennai 600 044, India
| | - Sekaran Saravanan
- Centre for Nanotechnology & Advance Biomaterials (CeNTAB), Department of Bioengineering, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613 401, Tamil Nadu, India
| | - Manickaraj Shairam
- Chemical Biology and Nanobiotechnology Laboratory, AU-KBC Research Centre, Anna University, MIT, Campus, Chrompet, Chennai 600 044, India
| | | | | | | | - Selvaraj Vimalraj
- Centre for Biotechnology, Anna University, Guindy, Chennai 600 025, India; Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600 077, Tamil Nadu, India.
| | - Subramaniyam Rajalakshmi
- Chemical Biology and Nanobiotechnology Laboratory, AU-KBC Research Centre, Anna University, MIT, Campus, Chrompet, Chennai 600 044, India.
| |
Collapse
|
16
|
Castillo-Henríquez L, Castro-Alpízar J, Lopretti-Correa M, Vega-Baudrit J. Exploration of Bioengineered Scaffolds Composed of Thermo-Responsive Polymers for Drug Delivery in Wound Healing. Int J Mol Sci 2021; 22:1408. [PMID: 33573351 PMCID: PMC7866792 DOI: 10.3390/ijms22031408] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Innate and adaptive immune responses lead to wound healing by regulating a complex series of events promoting cellular cross-talk. An inflammatory response is presented with its characteristic clinical symptoms: heat, pain, redness, and swelling. Some smart thermo-responsive polymers like chitosan, polyvinylpyrrolidone, alginate, and poly(ε-caprolactone) can be used to create biocompatible and biodegradable scaffolds. These processed thermo-responsive biomaterials possess 3D architectures similar to human structures, providing physical support for cell growth and tissue regeneration. Furthermore, these structures are used as novel drug delivery systems. Locally heated tumors above the polymer lower the critical solution temperature and can induce its conversion into a hydrophobic form by an entropy-driven process, enhancing drug release. When the thermal stimulus is gone, drug release is reduced due to the swelling of the material. As a result, these systems can contribute to the wound healing process in accelerating tissue healing, avoiding large scar tissue, regulating the inflammatory response, and protecting from bacterial infections. This paper integrates the relevant reported contributions of bioengineered scaffolds composed of smart thermo-responsive polymers for drug delivery applications in wound healing. Therefore, we present a comprehensive review that aims to demonstrate these systems' capacity to provide spatially and temporally controlled release strategies for one or more drugs used in wound healing. In this sense, the novel manufacturing techniques of 3D printing and electrospinning are explored for the tuning of their physicochemical properties to adjust therapies according to patient convenience and reduce drug toxicity and side effects.
Collapse
Affiliation(s)
- Luis Castillo-Henríquez
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200 San José, Costa Rica;
- Physical Chemistry Laboratory, Faculty of Pharmacy, University of Costa Rica, 11501-2060 San José, Costa Rica
| | - Jose Castro-Alpízar
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Costa Rica, 11501-2060 San José, Costa Rica;
| | - Mary Lopretti-Correa
- Nuclear Research Center, Faculty of Science, Universidad de la República (UdelaR), 11300 Montevideo, Uruguay;
| | - José Vega-Baudrit
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200 San José, Costa Rica;
- Laboratory of Polymers (POLIUNA), Chemistry School, National University of Costa Rica, 86-3000 Heredia, Costa Rica
| |
Collapse
|
17
|
Toprak Ö, Topuz B, Monsef YA, Oto Ç, Orhan K, Karakeçili A. BMP-6 carrying metal organic framework-embedded in bioresorbable electrospun fibers for enhanced bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111738. [PMID: 33545881 DOI: 10.1016/j.msec.2020.111738] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022]
Abstract
Biomolecule carrier structures have attracted substantial interest owing to their potential utilizations in the field of bone tissue engineering. In this study, MOF-embedded electrospun fiber scaffold for the controlled release of BMP-6 was developed for the first time, to enrich bone regeneration efficacy. The scaffolds were achieved by first, one-pot rapid crystallization of BMP-6 encapsulated ZIF-8 nanocrystals-as a novel carrier for growth factor molecules- and then electrospinning of the blending solution composed of poly (ε-caprolactone) and BMP-6 encapsulated ZIF-8 nanocrystals. BMP-6 molecule encapsulation efficiency for ZIF-8 nanocrystals was calculated as 98%. The in-vitro studies showed that, the bioactivity of BMP-6 was preserved and the release lasted up to 30 days. The release kinetics fitted the Korsmeyer-Peppas model exhibiting a pseudo-Fickian behavior. The in-vitro osteogenesis studies revealed the superior effect of sustained release of BMP-6 towards osteogenic differentiation of MC3T3-E1 pre-osteoblasts. In-vivo studies also revealed that the sustained slow release of BMP-6 was responsible for the generation of well-mineralized, new bone formation in a rat cranial defect. Our results proved that; MOF-carriers embedded in electrospun scaffolds can be used as an effective platform for bone regeneration in bone tissue engineering applications. The proposed approach can easily be adapted for various growth factor molecules for different tissue engineering applications.
Collapse
Affiliation(s)
- Özge Toprak
- Ankara University, Faculty of Engineering, Chemical Engineering Department, 06100 Ankara, Turkey
| | - Berna Topuz
- Ankara University, Faculty of Engineering, Chemical Engineering Department, 06100 Ankara, Turkey
| | - Yanad Abou Monsef
- Ankara University, Faculty of Veterinary Medicine, Department of Pathology, 06110 Ankara, Turkey
| | - Çağdaş Oto
- Ankara University, Faculty of Veterinary Medicine, Department of Anatomy, 06110 Ankara, Turkey; Ankara University Medical Design Application and Research Center (MEDITAM), Ankara, Turkey
| | - Kaan Orhan
- Ankara University, Faculty of Dentistry, Department of DentoMaxillofacial Radiology, 06100, Ankara, Turkey; Ankara University Medical Design Application and Research Center (MEDITAM), Ankara, Turkey
| | - Ayşe Karakeçili
- Ankara University, Faculty of Engineering, Chemical Engineering Department, 06100 Ankara, Turkey.
| |
Collapse
|
18
|
Sukul M, Sahariah P, Lauzon HL, Borges J, Másson M, Mano JF, Haugen HJ, Reseland JE. In vitro biological response of human osteoblasts in 3D chitosan sponges with controlled degree of deacetylation and molecular weight. Carbohydr Polym 2020; 254:117434. [PMID: 33357907 DOI: 10.1016/j.carbpol.2020.117434] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/03/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022]
Abstract
We have studied the effect of chitosan sponges, produced from chitosan batches with distinct degree of deacetylation (DDA) and molecular weight (Mw), on the adhesion, growth and differentiation of primary human osteoblasts with an aim to offer a suitable tool for guided bone regeneration. All the chitosan sponges revealed similar microstructure, irrespective of the DDA (58, 73, 82, 88, and 91 %) and Mw (749, 547, 263, 215, and 170 kDa, respectively). Cell spreading was higher on sponges having a higher DDA. Higher DDA induced a more pronounced increase in alkaline phosphatase activity, osteopontin (OPN), vascular endothelial growth factor-A (VEGF), interleukin-6 (IL-6), and reduction in monocyte chemoattractant protein-1 (MCP-1), sclerostin (SOST) and dickkopf related protein-1 as compared to lower DDA. Lower DDA induced the increased secretion of osteoprotegerin and SOST as compared to higher DDA. The combination of higher DDA and Mw induced an increased secretion of VEGF and IL-6, however reduced the secretion of OPN as compared to chitosan with similar DDA but with lower Mw. In summary, the variations in cellular responses to the different chitosan sponges indicate a potential for individual tailoring of desired responses in guided bone regeneration.
Collapse
Affiliation(s)
- Mousumi Sukul
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway.
| | - Priyanka Sahariah
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavík, Iceland
| | | | - João Borges
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Már Másson
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavík, Iceland
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Håvard J Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway
| | - Janne E Reseland
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
19
|
Conjugation with Methylsulfonylmethane Improves Hyaluronic Acid Anti-Inflammatory Activity in a Hydrogen Peroxide-Exposed Tenocyte Culture In Vitro Model. Int J Mol Sci 2020; 21:ijms21217956. [PMID: 33114764 PMCID: PMC7662253 DOI: 10.3390/ijms21217956] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 12/11/2022] Open
Abstract
Rotator cuff tears (RCTs) and rotator cuff disease (RCD) are important causes of disability in middle-aged individuals affected by nontraumatic shoulder dysfunctions. Our previous studies have demonstrated that four different hyaluronic acid preparations (HAPs), including Artrosulfur® hyaluronic acid (HA) (Alfakjn S.r.l., Garlasco, Italy), may exert a protective effect in human RCT-derived tendon cells undergoing oxidative stress damage. Recently, methylsulfonylmethane (MSM) (Barentz, Paderno Dugnano, Italy) has proven to have anti-inflammatory properties and to cause pain relief in patients affected by tendinopathies. This study aims at evaluating three preparations (Artrosulfur® HA, MSM, and Artrosulfur® MSM + HA) in the recovery from hydrogen peroxide-induced oxidative stress damage in human tenocyte. Cell proliferation, Lactate Dehydrogenase (LDH) release, and inducible nitric oxide synthases (iNOS) and prostaglandin E2 (PGE2) modulation were investigated. In parallel, expression of metalloproteinases 2 (MMP2) and 14 (MMP14) and collagen types I and III were also examined. Results demonstrate that Artrosulfur® MSM + HA improves cell escape from oxidative stress by decreasing cytotoxicity and by reducing iNOS and PGE2 secretion. Furthermore, it differentially modulates MMP2 and MMP14 levels and enhances collagen III expression after 24 h, proteins globally related to rapid acceleration of the extracellular matrix (ECM) remodelling and thus tendon healing. By improving the anti-cytotoxic effect of HA, the supplementation of MSM may represent a feasible strategy to ameliorate cuff tendinopathies.
Collapse
|
20
|
Stutz C, Strub M, Clauss F, Huck O, Schulz G, Gegout H, Benkirane-Jessel N, Bornert F, Kuchler-Bopp S. A New Polycaprolactone-Based Biomembrane Functionalized with BMP-2 and Stem Cells Improves Maxillary Bone Regeneration. NANOMATERIALS 2020; 10:nano10091774. [PMID: 32911737 PMCID: PMC7558050 DOI: 10.3390/nano10091774] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/06/2020] [Indexed: 12/12/2022]
Abstract
Oral diseases have an impact on the general condition and quality of life of patients. After a dento-alveolar trauma, a tooth extraction, or, in the case of some genetic skeletal diseases, a maxillary bone defect, can be observed, leading to the impossibility of placing a dental implant for the restoration of masticatory function. Recently, bone neoformation was demonstrated after in vivo implantation of polycaprolactone (PCL) biomembranes functionalized with bone morphogenic protein 2 (BMP-2) and ibuprofen in a mouse maxillary bone lesion. In the present study, human bone marrow derived mesenchymal stem cells (hBM-MSCs) were added on BMP-2 functionalized PCL biomembranes and implanted in a maxillary bone lesion. Viability of hBM-MSCs on the biomembranes has been observed using the "LIVE/DEAD" viability test and scanning electron microscopy (SEM). Maxillary bone regeneration was observed for periods ranging from 90 to 150 days after implantation. Various imaging methods (histology, micro-CT) have demonstrated bone remodeling and filling of the lesion by neoformed bone tissue. The presence of mesenchymal stem cells and BMP-2 allows the acceleration of the bone remodeling process. These results are encouraging for the effectiveness and the clinical use of this new technology combining growth factors and mesenchymal stem cells derived from bone marrow in a bioresorbable membrane.
Collapse
Affiliation(s)
- Céline Stutz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
| | - Marion Strub
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Periodontology, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Georg Schulz
- Core Facility Micro- and Nanotomography, Biomaterials Science Center (BMC), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland;
| | - Hervé Gegout
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
| | - Fabien Bornert
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Oral Medicine and Oral Surgery, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Correspondence: ; Tel.: +33-619610523
| |
Collapse
|
21
|
Jang HY, Shin JY, Oh SH, Byun JH, Lee JH. PCL/HA Hybrid Microspheres for Effective Osteogenic Differentiation and Bone Regeneration. ACS Biomater Sci Eng 2020; 6:5172-5180. [PMID: 33455267 DOI: 10.1021/acsbiomaterials.0c00550] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The purpose of this study is to develop a bioactive bone graft based on polycaprolactone (PCL, synthetic polymer; used in clinical practices as a grafting material for craniofacial bone defects) and hyaluronic acid (HA, bioactive natural polymer; known as a promoting substance for bone regeneration) that would be fabricated by clinically available procedures (mild condition without toxic chemicals) and provide bioactivity for sufficient period, and thus effectively induce bone reconstruction. For this, PCL/HA hybrid microspheres were produced by a spray-precipitation technique using clinically adapted solvents. The HA was stably and evenly entrapped in the PCL/HA hybrid microspheres. It was demonstrated that the PCL/HA hybrid microspheres provide an appropriate environment for proliferation and osteogenic differentiation of human periosteum-derived cells (hPDCs) (in vitro) and allow significantly enhanced bone regeneration (in vivo) compared with PCL microspheres without HA. The PCL/HA hybrid microspheres can be a simple but clinically applicable bioactive bone graft for large-sized bone defects.
Collapse
Affiliation(s)
- Hee Yun Jang
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| | - Jun Yung Shin
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea.,Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jin Ho Lee
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| |
Collapse
|
22
|
Sukul M, Cama G, Dubruel P, Reseland JE, Haugen HJ. Methacrylation increase growth and differentiation of primary human osteoblasts for gelatin hydrogels. EMERGENT MATERIALS 2020; 3:559-566. [DOI: 10.1007/s42247-020-00101-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/15/2020] [Indexed: 01/06/2025]
Abstract
AbstractThe role of gelatin methacrylate hydrogels with varying degrees of methacrylation (69% and 84%) was accessed with FTIR, NMR, microCT, and subsequent exposure to human osteoblasts. The cells responded positively to the degree of methacrylation and showed attachment, growth, and proliferated on both hydrogels. The cell reacted differently to the degree of methacrylation with higher proliferation on higher substitution; however, cell differentiation behavior was improved for less substitution. The secretion of late osteogenic markers (osteoprotegerin (OPG), osteopontin (OPN), and osteocalcin (OCN)) and angiogenic factor vascular endothelial growth factor (VEGF) was increased for gelatin methacrylate hydrogels with 69% degree of methacrylation and thus would be the better candidate for future bone regenerative applications amongst the three tested hydrogels.
Collapse
|
23
|
Zafar MS, Amin F, Fareed MA, Ghabbani H, Riaz S, Khurshid Z, Kumar N. Biomimetic Aspects of Restorative Dentistry Biomaterials. Biomimetics (Basel) 2020; 5:E34. [PMID: 32679703 PMCID: PMC7557867 DOI: 10.3390/biomimetics5030034] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Biomimetic has emerged as a multi-disciplinary science in several biomedical subjects in recent decades, including biomaterials and dentistry. In restorative dentistry, biomimetic approaches have been applied for a range of applications, such as restoring tooth defects using bioinspired peptides to achieve remineralization, bioactive and biomimetic biomaterials, and tissue engineering for regeneration. Advancements in the modern adhesive restorative materials, understanding of biomaterial-tissue interaction at the nano and microscale further enhanced the restorative materials' properties (such as color, morphology, and strength) to mimic natural teeth. In addition, the tissue-engineering approaches resulted in regeneration of lost or damaged dental tissues mimicking their natural counterpart. The aim of the present article is to review various biomimetic approaches used to replace lost or damaged dental tissues using restorative biomaterials and tissue-engineering techniques. In addition, tooth structure, and various biomimetic properties of dental restorative materials and tissue-engineering scaffold materials, are discussed.
Collapse
Affiliation(s)
- Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
| | - Faiza Amin
- Science of Dental Materials Department, Dow Dental College, Dow University of Health Sciences, Karachi 74200, Pakistan;
| | - Muhmmad Amber Fareed
- Adult Restorative Dentistry, Dental Biomaterials and Prosthodontics Oman Dental College, Muscat 116, Sultanate of Oman;
| | - Hani Ghabbani
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
| | - Samiya Riaz
- School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudia Arabia;
| | - Naresh Kumar
- Department of Science of Dental Materials, Dow University of Health Sciences, Karachi 74200, Pakistan;
| |
Collapse
|
24
|
Behere I, Pardawala Z, Vaidya A, Kale V, Ingavle G. Osteogenic differentiation of an osteoblast precursor cell line using composite PCL-gelatin-nHAp electrospun nanofiber mesh. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1767619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Isha Behere
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
| | - Zain Pardawala
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
| | - Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
25
|
Faruq O, Sayed S, Kim B, Im S, Lee B. A biphasic calcium phosphate ceramic scaffold loaded with oxidized cellulose nanofiber–gelatin hydrogel with immobilized simvastatin drug for osteogenic differentiation. J Biomed Mater Res B Appl Biomater 2020; 108:1229-1238. [DOI: 10.1002/jbm.b.34471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Omar Faruq
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Shithima Sayed
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Boram Kim
- Institute of Tissue Regeneration, College of MedicineSoonchunhyang University Cheonan South Korea
| | - Soo‐Bin Im
- Department of Neurosurgery, College of MedicineSoonchunhyang University, Bucheon Hospital Bucheon South Korea
| | - Byong‐Taek Lee
- Department of Regenerative Medicine, College of MedicineSoonchunhyang University Cheonan South Korea
- Institute of Tissue Regeneration, College of MedicineSoonchunhyang University Cheonan South Korea
| |
Collapse
|
26
|
Electrospun gelatin matrices with bioactive pDNA polyplexes. Int J Biol Macromol 2020; 149:296-308. [DOI: 10.1016/j.ijbiomac.2020.01.252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/15/2022]
|
27
|
Aljohani H, Senbanjo LT, Chellaiah MA. Methylsulfonylmethane increases osteogenesis and regulates the mineralization of the matrix by transglutaminase 2 in SHED cells. PLoS One 2019; 14:e0225598. [PMID: 31805069 PMCID: PMC6894810 DOI: 10.1371/journal.pone.0225598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/07/2019] [Indexed: 01/09/2023] Open
Abstract
Methylsulfonylmethane (MSM) is a naturally occurring, sulfate-containing, organic compound. It has been shown to stimulate the differentiation of mesenchymal stem cells into osteoblast-like cells and bone formation. In this study, we investigated whether MSM influences the differentiation of stem cells from human exfoliated deciduous teeth (SHED) into osteoblast-like cells and their osteogenic potential. Here, we report that MSM induced osteogenic differentiation through the expression of osteogenic markers such as osterix, osteopontin, and RUNX2, at both mRNA and protein levels in SHED cells. An increase in the activity of alkaline phosphatase and mineralization confirmed the osteogenic potential of MSM. These MSM-induced effects were observed in cells grown in basal medium but not osteogenic medium. MSM induced transglutaminase-2 (TG2), which may be responsible for the cross-linking of extracellular matrix proteins (collagen or osteopontin), and the mineralization process. Inhibition of TG2 ensued a significant decrease in the differentiation of SHED cells and cross-linking of matrix proteins. A comparison of mineralization with the use of mineralized and demineralized bone particles in the presence of MSM revealed that mineralization is higher with mineralized bone particles than with demineralized bone particles. In conclusion, these results indicated that MSM could promote differentiation and osteogenic potential of SHED cells. This osteogenic property is more in the presence of mineralized bone particles. TG2 is a likely cue in the regulation of differentiation and mineral deposition of SHED cells in response to MSM.
Collapse
Affiliation(s)
- Hanan Aljohani
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States of America
- Department of Oral Medicine and Diagnostics Sciences, King Saud University School of Dentistry, Riyadh, KSA
| | - Linda T. Senbanjo
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States of America
| | - Meenakshi A. Chellaiah
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States of America
| |
Collapse
|
28
|
Li X, Yin HM, Luo E, Zhu S, Wang P, Zhang Z, Liao GQ, Xu JZ, Li ZM, Li JH. Accelerating Bone Healing by Decorating BMP-2 on Porous Composite Scaffolds. ACS APPLIED BIO MATERIALS 2019; 2:5717-5726. [PMID: 35021565 DOI: 10.1021/acsabm.9b00761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xiang Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Hua-Mo Yin
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Peng Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Gui-Qing Liao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Ji-Hua Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
29
|
John JV, Choksi M, Chen S, Boda SK, Su Y, McCarthy A, Teusink MJ, Reinhardt RA, Xie J. Tethering peptides onto biomimetic and injectable nanofiber microspheres to direct cellular response. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 22:102081. [PMID: 31400571 PMCID: PMC6904511 DOI: 10.1016/j.nano.2019.102081] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023]
Abstract
Biomimetic and injectable nanofiber microspheres (NMs) could be ideal candidate for minimally invasive tissue repair. Herein, we report a facile approach to fabricate peptide-tethered NMs by combining electrospinning, electrospraying, and surface conjugation techniques. The composition and size of NMs can be tuned by varying the processing parameters during the fabrication. Further, bone morphogenic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) mimicking peptides have been successfully tethered onto poly(ε-caprolactone) (PCL):gelatin:(gelatin-methacryloyl) (GelMA)(1:0.5:0.5) NMs through photocrosslinking of the methacrylic group in GelMA and octenyl alanine (OCTAL) in the modified peptides. The BMP-2-OCTAL peptide-tethered NMs significantly promote osteogenic differentiation of bone marrow-derived stem cells (BMSCs). Moreover, human umbilical vein endothelial cells (HUVECs) seeded on VEGF mimicking peptide QK-OCTAL-tethered NMs significantly up-regulated vascular-specific proteins, leading to microvascularization. The strategy developed in this work holds great potential in developing a biomimetic and injectable carrier to efficiently direct cellular response (Osteogenesis and Angiogenesis) for tissue repair.
Collapse
Affiliation(s)
- Johnson V John
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Meera Choksi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shixuan Chen
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sunil Kumar Boda
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alec McCarthy
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew J Teusink
- Department of Orthopaedic Surgery and Rehabilitation, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Richard A Reinhardt
- Department of Surgical Specialties, College of Dentistry, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska Lincoln, Lincoln, NE, USA.
| |
Collapse
|
30
|
Thermal cycling effect on osteogenic differentiation of MC3T3-E1 cells loaded on 3D-porous Biphasic Calcium Phosphate (BCP) scaffolds for early osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110027. [PMID: 31546388 DOI: 10.1016/j.msec.2019.110027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/21/2019] [Accepted: 07/26/2019] [Indexed: 12/30/2022]
Abstract
The application of heat stress on a defect site during the healing process is a promising technique for early bone regeneration. The primary goal of this study was to investigate the effect of periodic heat shock on bone formation. MC3T3-E1 cells were seeded onto biphasic calcium phosphate (BCP) scaffolds, followed by periodic heating to evaluate osteogenic differentiation. Heat was applied to cells seeded onto scaffolds at 41 °C for 1 h once, twice, and four times a day for seven days and their viability, morphology, and differentiation were analyzed. BCP scaffolds with interconnected porous structures mimic bone biology for cellular studies. MTT and confocal studies have shown that heat shock significantly increased cell proliferation without any toxic effects. Compared to non-heated samples, heat shock enhanced calcium deposition and mineralization, which could be visualized by SEM observation and Alizarin red S staining. Immunostaining images showed the localization of osteogenic proteins ALP and OPN on heat-shocked cells. qRT-PCR analysis revealed the presence of more osteospecific markers, osteopontin (OPN), osteocalcin, collagen type X, and Runx2, in the heat-shocked samples than in the non-heated sample. Periodic heat shock significantly upregulated both heat shock proteins (HSP70 and HSP27) in differentiated MC3T3-E1 cells. The results of this study demonstrated that periodically heat applied especially two times a day was better approach for osteogenic differentiation. Hence, this work provides a define temperature and time schedule for the development of a clinical heating device in future for early bone regeneration during the postsurgical period.
Collapse
|
31
|
Ranganathan S, Balagangadharan K, Selvamurugan N. Chitosan and gelatin-based electrospun fibers for bone tissue engineering. Int J Biol Macromol 2019; 133:354-364. [DOI: 10.1016/j.ijbiomac.2019.04.115] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/06/2019] [Accepted: 04/16/2019] [Indexed: 12/29/2022]
|
32
|
Balagangadharan K, Trivedi R, Vairamani M, Selvamurugan N. Sinapic acid-loaded chitosan nanoparticles in polycaprolactone electrospun fibers for bone regeneration in vitro and in vivo. Carbohydr Polym 2019; 216:1-16. [PMID: 31047045 DOI: 10.1016/j.carbpol.2019.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022]
Abstract
Sinapic acid (SA) is a plant-derived phenolic compound known for its multiple biological properties, but its role in the promotion of bone formation is not yet well-studied. Moreover, the delivery of SA is hindered by its complex hydrophobic nature, limiting its bioavailability. In this study, we fabricated a drug delivery system using chitosan nanoparticles (nCS) loaded with SA at different concentrations. These were incorporated into polycaprolactone (PCL) fibers via an electrospinning method. nCS loaded with 50 μM SA in PCL fibers promoted osteoblast differentiation. Furthermore, SA treatment activated the osteogenesis signaling pathways in mouse mesenchymal stem cells. A critical-sized rat calvarial bone defect model system identified that the inclusion of SA into PCL/nCS fibers accelerated bone formation. Collectively, these data suggest that SA promoted osteoblast differentiation in vitro and bone formation in vivo, possibly by activating the TGF-β1/BMP/Smads/Runx2 signaling pathways, suggesting SA might have therapeutic benefits in bone regeneration.
Collapse
Affiliation(s)
- Kalimuthu Balagangadharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ritu Trivedi
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow 226031, Uttar Pradesh, India
| | - Mariappanadar Vairamani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
33
|
Jang CH, Lee J, Kim G. Synergistic effect of alginate/BMP-2/Umbilical cord serum-coated on 3D-printed PCL biocomposite for mastoid obliteration model. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.12.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
34
|
Nobile S, Nobile L. Recent Research Progress on Scaffolds for Bone Repair and Regeneration. IFMBE PROCEEDINGS 2019. [DOI: 10.1007/978-981-10-9023-3_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
35
|
Zheng S, Guan Y, Yu H, Huang G, Zheng C. Poly-l-lysine-coated PLGA/poly(amino acid)-modified hydroxyapatite porous scaffolds as efficient tissue engineering scaffolds for cell adhesion, proliferation, and differentiation. NEW J CHEM 2019. [DOI: 10.1039/c9nj01675a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ideal bone tissue engineering scaffolds should be biocompatible, biodegradable, and mechanically robust and have the ability to regulate cell function.
Collapse
Affiliation(s)
- Shuang Zheng
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- P. R. China
| | - Yonghong Guan
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- P. R. China
| | - Haichi Yu
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- P. R. China
| | - Ge Huang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- P. R. China
| | - Changjun Zheng
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- P. R. China
| |
Collapse
|
36
|
Chen L, Shao L, Wang F, Huang Y, Gao F. Enhancement in sustained release of antimicrobial peptide and BMP-2 from degradable three dimensional-printed PLGA scaffold for bone regeneration. RSC Adv 2019; 9:10494-10507. [PMID: 35515290 PMCID: PMC9062520 DOI: 10.1039/c8ra08788a] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/26/2019] [Indexed: 11/24/2022] Open
Abstract
One of the goals of bone tissue engineering is to create scaffolds with well-defined, inter-connected pores, excellent biocompatibility and osteoinductive ability. Three-dimensional (3D)-printed polymer scaffold coated with bioactive peptide are an effective approach to fabricating ideal bone tissue engineering scaffolds for bone defect repair. However, the current strategy of adding bioactive peptides generally cause degradation to the polymer materials or damage the bioactivity of the biomolecules. Thus, in this study, we used a biomimetic process via poly(dopamine) coating to prepare functional 3D PLGA porous scaffolds with immobilized BMP-2 and ponericin G1 that efficiently regulate the osteogenic differentiation of preosteoblasts (MC3T3-E1) and simultaneously inhibit of pathogenic microbes, thereby enhancing biological activity. In this study, we analysed a 3D PLGA porous scaffold by scanning electron microscopy, water contact angle measurements, and materials testing. Subsequently, we examined the adsorption, release and in vitro antimicrobial activity of the 3D PLGA. Surface characterization showed that poly(dopamine) surface modification could more efficiently mediate the immobilization of BMP-2 and ponericin G1 onto the scaffold surfaces than physical adsorption, and that ponericin G1-immobilized 3D PLGA scaffolds were able to maintain long-term antibacterial activity. We evaluated the influence on cell adhesion, proliferation and differentiation by culturing MC3T3-E1 cells on different modified 3D PLGA scaffolds in vitro. The biological results indicate that MC3T3-E1 cell attachment and proliferation on BMP-2/ponericin G1-immobilized 3D PLGA scaffolds were much higher than those on other groups. Calcium deposition, and gene expression results showed that the osteogenic differentiation of cells was effectively improved by loading the 3D PLGA scaffold with BMP-2 and ponericin G1. In summary, our findings indicated that the polydopamine-assisted surface modification method can be a useful tool for grafting biomolecules onto biodegradable implants, and the dual release of BMP-2 and ponericin G1 can enhance the osteointegration of bone implants and simultaneously inhibit of pathogenic microbes. Therefore, we conclude that the BMP-2/ponericin G1-loaded PLGA 3D scaffolds are versatile and biocompatible scaffolds for bone tissue engineering. One of the goals of bone tissue engineering is to create scaffolds with well-defined, inter-connected pores, excellent biocompatibility and osteoinductive ability.![]()
Collapse
Affiliation(s)
- Lei Chen
- Department of Joints and Sports Medicine
- The First Hospital of Jilin University
- Changchun
- PR China
| | - Liping Shao
- Department of Joints and Sports Medicine
- The First Hospital of Jilin University
- Changchun
- PR China
| | - Fengping Wang
- Department of Joints and Sports Medicine
- The First Hospital of Jilin University
- Changchun
- PR China
| | - Yifan Huang
- Department of Joints Surgery
- The First Hospital of Jilin University
- Changchun
- PR China
| | - Fenghui Gao
- Department of Orthopedic
- The First Hospital of Jilin University
- Changchun
- PR China
| |
Collapse
|
37
|
Vashisth P, Bellare JR. Development of hybrid scaffold with biomimetic 3D architecture for bone regeneration. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1325-1336. [DOI: 10.1016/j.nano.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 01/27/2023]
|
38
|
Makkar P, Kang HJ, Padalhin AR, Park I, Moon BG, Lee BT. Development and properties of duplex MgF2/PCL coatings on biodegradable magnesium alloy for biomedical applications. PLoS One 2018; 13:e0193927. [PMID: 29608572 PMCID: PMC5880343 DOI: 10.1371/journal.pone.0193927] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 02/21/2018] [Indexed: 11/18/2022] Open
Abstract
The present work addresses the performance of polycaprolactone (PCL) coating on fluoride treated (MgF2) biodegradable ZK60 magnesium alloy (Mg) for biomedical application. MgF2 conversion layer was first produced by immersing Mg alloy substrate in hydrofluoric acid solution. The outer PCL coating was then prepared using dip coating technique. Morphology, elements profile, phase structure, roughness, mechanical properties, invitro corrosion, and biocompatibility of duplex MgF2/PCL coating were then characterized and compared to those of fluoride coated and uncoated Mg samples. The invivo degradation behavior and biocompatibility of duplex MgF2/PCL coating with respect to ZK60 Mg alloy were also studied using rabbit model for 2 weeks. SEM and TEM analysis showed that the duplex coating was uniform and comprised of porous PCL film (~3.3 μm) as upper layer with compact MgF2 (~2.2 μm) as inner layer. No significant change in microhardness was found on duplex coating compared with uncoated ZK60 Mg alloy. The duplex coating showed improved invitro corrosion resistance than single layered MgF2 or uncoated alloy samples. The duplex coating also resulted in better cell viability, cell adhesion, and cell proliferation compared to fluoride coated or uncoated alloy. Preliminary invivo studies indicated that duplex MgF2/PCL coating reduced the degradation rate of ZK60 Mg alloy and exhibited good biocompatibility. These results suggested that duplex MgF2/PCL coating on magnesium alloy might have great potential for orthopedic applications.
Collapse
Affiliation(s)
- Preeti Makkar
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Hoe Jin Kang
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Andrew R. Padalhin
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Ihho Park
- Sk Innovation Global Technology, Daejeon, South Korea
| | - Byoung-Gi Moon
- Advanced Metals Division, Korea Institute of Materials Science, Changwon, Gyeongnam, South Korea
| | - Byong Taek Lee
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
- * E-mail:
| |
Collapse
|
39
|
Casanova MR, Reis RL, Martins A, Neves NM. The Use of Electrospinning Technique on Osteochondral Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1058:247-263. [PMID: 29691825 DOI: 10.1007/978-3-319-76711-6_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Electrospinning, an electrostatic fiber fabrication technique, has attracted significant interest in recent years due to its versatility and ability to produce highly tunable nanofibrous meshes. These nanofibrous meshes have been investigated as promising tissue engineering scaffolds since they mimic the scale and morphology of the native extracellular matrix. The sub-micron diameter of fibers produced by this process presents various advantages like the high surface area to volume ratio, tunable porosity, and the ability to manipulate the nanofiber composition in order to get desired properties and functionality. Electrospun fibers can be oriented or arranged randomly, giving control over both mechanical properties and the biological response to the fibrous scaffold. Moreover, bioactive molecules can be integrated with the electrospun nanofibrous scaffolds in order to improve the cellular response. This chapter presents an overview of the developments on electrospun polymer nanofibers including processing, structure, and their applications in the field of osteochondral tissue engineering.
Collapse
Affiliation(s)
- Marta R Casanova
- 3B's Research Group-Biomaterials, Biodegradable and Biomimetics, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group-Biomaterials, Biodegradable and Biomimetics, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Albino Martins
- 3B's Research Group-Biomaterials, Biodegradable and Biomimetics, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno M Neves
- 3B's Research Group-Biomaterials, Biodegradable and Biomimetics, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco/Guimarães, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
40
|
Prolonged delivery of BMP-2 by a non-polymer hydrogel for bone defect regeneration. Drug Deliv Transl Res 2017; 8:178-190. [DOI: 10.1007/s13346-017-0451-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Kim B, Ventura R, Lee BT. Functionalization of porous BCP scaffold by generating cell-derived extracellular matrix from rat bone marrow stem cells culture for bone tissue engineering. J Tissue Eng Regen Med 2017; 12:e1256-e1267. [PMID: 28752541 DOI: 10.1002/term.2529] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 05/24/2017] [Accepted: 07/24/2017] [Indexed: 12/24/2022]
Abstract
The potential of decellularized cell-derived extracellular matrix (ECM) deposited on biphasic calcium phosphate (BCP) scaffold for bone tissue engineering was investigated. Rat derived bone marrow mesenchymal stem cells were cultured on porous BCP scaffolds for 3 weeks and decellularized with two different methods (freeze-thaw [F/T] or sodium dodecyl sulfate [SDS]). The decellularized ECM deposited scaffolds (dECM-BCP) were characterized through scanning electron microscopy, energy dispersive X-ray spectrometer, and confocal microscopy. The efficiency of decellularization was evaluated by quantifying remaining DNA, sulfated glycosaminoglycans, and collagens. Results revealed that F/T method was more effective procedure for removing cellular components of cultured cells (95.21% DNA reduction) than SDS treatment (92.49%). Although significant loss of collagen was observed after decellularization with both F/T (56.68%) and SDS (70.87%) methods, F/T treated sample showed higher retaining amount of sulfated glycosaminoglycans content (75.64%) than SDS (33.28%). In addition, we investigated the cell biocompatibility and osteogenic effect of dECM-BCP scaffolds using preosteoblasts (MC3T3-E1). Compared to bare BCP scaffolds, dECM-BCP_F/T scaffolds showed improved cell attachment and proliferation based on immunofluorescence staining and water soluble tetrazolium salts assay (p < .001). Moreover, dECM-BCP scaffolds showed increased osteoblastic differentiation of newly seeded preosteoblasts by up-regulating three types of osteoblastic genes (osteopontin, alkaline phosphatase, and bone morphogenic protein-2). This study demonstrated that functionalization of BCP scaffold using cell-derived ECM could be useful for improving the bioactivity of materials and providing suitable microenvironment, especially for osteogenesis. Further study is needed to determine the potential of dECM-BCP scaffold for bone formation and regeneration in vivo.
Collapse
Affiliation(s)
- Boram Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Reiza Ventura
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Byong-Taek Lee
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Korea.,Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
42
|
Preparation of P3HB4HB/(Gelatin + PVA) Composite Scaffolds by Coaxial Electrospinning and Its Biocompatibility Evaluation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9251806. [PMID: 29349086 PMCID: PMC5733976 DOI: 10.1155/2017/9251806] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 06/19/2017] [Accepted: 09/06/2017] [Indexed: 01/23/2023]
Abstract
This study was conducted to prepare coaxial electrospun scaffolds of P3HB4HB/(gelatin + PVA) with various concentration ratios with P3HB4HB as the core solution and gelatin + PVA mixture as the shell solution; the mass ratios of gelatin and PVA in each 10 mL shell mixture were 0.6 g : 0.2 g (Group A), 0.4 g : 0.4 g (Group B), and 0.2 g : 0.6 g (Group C). The results showed that the pore size, porosity, and cell proliferation rate of Group C were better than those of Groups A and B. The ascending order of the tensile strength and modulus of elasticity was Group A < Group B < Group C. The surface roughness was Group C > Group B > Group A. The osteogenic and chondrogenic-specific staining showed that Group C was stronger than Groups A and B. This study demonstrates that when the mass ratio of gelatin : PVA was 0.2 g : 0.6 g, a P3HB4HB/(gelatin + PVA) composite scaffold with a core-shell structure can be prepared, and the scaffold has good biocompatibility that it may be an ideal scaffold for tissue engineering.
Collapse
|
43
|
Choi JS, Park JS, Kim B, Lee BT, Yim JH. In vitro biocompatibility of vapour phase polymerised conductive scaffolds for cell lines. POLYMER 2017. [DOI: 10.1016/j.polymer.2017.07.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Li G, Zhou T, Lin S, Shi S, Lin Y. Nanomaterials for Craniofacial and Dental Tissue Engineering. J Dent Res 2017; 96:725-732. [PMID: 28463533 DOI: 10.1177/0022034517706678] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- G. Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - T. Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - S. Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - S. Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Y. Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
45
|
Heterogeneous electrospun polycaprolactone/polyethylene glycol membranes with improved wettability, biocompatibility, and mineralization. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2017.01.054] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
46
|
Nobile S, Nobile L. Nanotechnology for biomedical applications: Recent advances in neurosciences and bone tissue engineering. POLYM ENG SCI 2017. [DOI: 10.1002/pen.24595] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Stefano Nobile
- Maternal and Child Department; Ospedali Riuniti di Ancona; via F. Corridoni 11 Ancona 60123 Italy
| | - Lucio Nobile
- Department DICAM-Campus of Cesena; University of Bologna; Via Cavalcavia 61 Cesena 47521 Italy
| |
Collapse
|
47
|
Lee J, Perikamana SKM, Ahmad T, Lee MS, Yang HS, Kim DG, Kim K, Kwon B, Shin H. Controlled Retention of BMP-2-Derived Peptide on Nanofibers Based on Mussel-Inspired Adhesion for Bone Formation. Tissue Eng Part A 2017; 23:323-334. [DOI: 10.1089/ten.tea.2016.0363] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sajeesh Kumar Madhurakkat Perikamana
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Taufiq Ahmad
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Min Suk Lee
- Department of Nanobio Medical Science, Dankook University, Chonan, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobio Medical Science, Dankook University, Chonan, Republic of Korea
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, Ohio
| | - Kyobum Kim
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Bosun Kwon
- Wooridul Life Sciences & WINNOVA Research Institute, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Abstract
Growth factors are essential orchestrators of the normal bone fracture healing response. For non-union defects, delivery of exogenous growth factors to the injured site significantly improves healing outcomes. However, current clinical methods for scaffold-based growth factor delivery are fairly rudimentary, and there is a need for greater spatial and temporal regulation to increase their in vivo efficacy. Various approaches used to provide spatiotemporal control of growth factor delivery from bone tissue engineering scaffolds include physical entrapment, chemical binding, surface modifications, biomineralization, micro- and nanoparticle encapsulation, and genetically engineered cells. Here, we provide a brief review of these technologies, describing the fundamental mechanisms used to regulate release kinetics. Examples of their use in pre-clinical studies are discussed, and their capacities to provide tunable, growth factor delivery are compared. These advanced scaffold systems have the potential to provide safer, more effective therapies for bone regeneration than the systems currently employed in the clinic.
Collapse
|
49
|
Dunn LL, de Valence S, Tille JC, Hammel P, Walpoth BH, Stocker R, Imhof BA, Miljkovic-Licina M. Biodegradable and plasma-treated electrospun scaffolds coated with recombinant Olfactomedin-like 3 for accelerating wound healing and tissue regeneration. Wound Repair Regen 2016; 24:1030-1035. [DOI: 10.1111/wrr.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/24/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Louise L. Dunn
- Vascular Biology Division, Victor Chang Cardiac Research Institute, and School of Medical Sciences; University of New South Wales; Sydney Australia
| | - Sarra de Valence
- School of Pharmaceutical Sciences; University of Geneva, University of Lausanne; Geneva Switzerland
| | | | - Philippe Hammel
- Department of Pathology and Immunology; University of Geneva Medical Center; Geneva Switzerland
| | - Beat H. Walpoth
- Service of Cardiovascular Surgery; Geneva University Hospital; Geneva Switzerland
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, and School of Medical Sciences; University of New South Wales; Sydney Australia
| | - Beat A. Imhof
- Department of Pathology and Immunology; University of Geneva Medical Center; Geneva Switzerland
| | | |
Collapse
|
50
|
Fabrication of Core-Shell PEI/pBMP2-PLGA Electrospun Scaffold for Gene Delivery to Periodontal Ligament Stem Cells. Stem Cells Int 2016; 2016:5385137. [PMID: 27313626 PMCID: PMC4899599 DOI: 10.1155/2016/5385137] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/18/2016] [Accepted: 04/11/2016] [Indexed: 12/04/2022] Open
Abstract
Bone tissue engineering is the most promising technology for enhancing bone regeneration. Scaffolds loaded with osteogenic factors improve the therapeutic effect. In this study, the bioactive PEI (polyethylenimine)/pBMP2- (bone morphogenetic protein-2 plasmid-) PLGA (poly(D, L-lactic-co-glycolic acid)) core-shell scaffolds were prepared using coaxial electrospinning for a controlled gene delivery to hPDLSCs (human periodontal ligament stem cells). The pBMP2 was encapsulated in the PEI phase as a core and PLGA was employed to control pBMP2 release as a shell. First, the scaffold characterization and mechanical properties were evaluated. Then the gene release behavior was analyzed. Our results showed that pBMP2 was released at high levels in the first few days, with a continuous release behavior in the next 28 days. At the same time, PEI/pBMP2 showed high transfection efficiency. Moreover, the core-shell electrospun scaffold showed BMP2 expression for a much longer time (more than 28 days) compared with the single axial electrospun scaffold, as evaluated by qRT-PCR and western blot after culturing with hPDLSCs. These results suggested that the core-shell PEI/pBMP2-PLGA scaffold fabricated by coaxial electrospinning had a good gene release behavior and showed a prolonged expression time with a high transfection efficiency.
Collapse
|