1
|
Farjaminejad S, Farjaminejad R, Garcia-Godoy F. Nanoparticles in Bone Regeneration: A Narrative Review of Current Advances and Future Directions in Tissue Engineering. J Funct Biomater 2024; 15:241. [PMID: 39330217 PMCID: PMC11432802 DOI: 10.3390/jfb15090241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/28/2024] Open
Abstract
The rising demand for effective bone regeneration has underscored the limitations of traditional methods like autografts and allografts, including donor site morbidity and insufficient biological signaling. This review examines nanoparticles (NPs) in tissue engineering (TE) to address these challenges, evaluating polymers, metals, ceramics, and composites for their potential to enhance osteogenesis and angiogenesis by mimicking the extracellular matrix (ECM) nanostructure. The methods involved synthesizing and characterizing nanoparticle-based scaffoldsand integrating hydroxyapatite (HAp) with polymers to enhance mechanical properties and osteogenic potential. The results showed that these NPs significantly promote cell growth, differentiation, and bone formation, with carbon-based NPs like graphene and carbon nanotubes showing promise. NPs offer versatile, biocompatible, and customizable scaffolds that enhance drug delivery and support bone repair. Despite promising results, challenges with cytotoxicity, biodistribution, and immune responses remain. Addressing these issues through surface modifications and biocompatible molecules can improve the biocompatibility and efficacy of nanomaterials. Future research should focus on long-term in vivo studies to assess the safety and efficacy of NP-based scaffolds and explore synergistic effects with other bioactive molecules or growth factors. This review underscores the transformative potential of NPs in advancing BTE and calls for further research to optimize these technologies for clinical applications.
Collapse
Affiliation(s)
- Samira Farjaminejad
- School of Health and Psychological Sciences, Department of Health Services Research and Management, City University of London, London WC1E 7HU, UK
| | - Rosana Farjaminejad
- School of Health and Psychological Sciences, Department of Health Services Research and Management, City University of London, London WC1E 7HU, UK
| | - Franklin Garcia-Godoy
- Department of Bioscience Research, Bioscience Research Center, College of Dentistry, University of Tennessee Health Science Center, 875 Union Avenue, Memphis, TN 38163, USA
| |
Collapse
|
2
|
Alazmi SO. A review on guided bone regeneration using titanium mesh. Bioinformation 2024; 20:562-565. [PMID: 39132237 PMCID: PMC11309112 DOI: 10.6026/973206300200562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 08/13/2024] Open
Abstract
The gold standard for bone regeneration in atrophic ridge patients is guided bone regeneration (GBR). This makes it possible to get enough bone volume for an appropriate implant-prosthetic rehabilitation. The barrier membranes must meet the primary GBR design requirements, which include adequate integration with the surrounding tissue, spaciousness and clinical manageability. Titanium mesh's superior mechanical qualities and biocompatibility have broadened the indications of GBR technology, enabling it to be used to restore alveolar ridges with more significant bone defects. GBR with titanium mesh is being used in many clinical settings and for a range of clinical procedures. Furthermore, several advancements in digitalization and material modification have resulted from the study of GBR using titanium mesh. Hence, we report a review on the various characteristics of titanium mesh and its current use in clinical settings for bone augmentation.
Collapse
Affiliation(s)
- Saad Obaid Alazmi
- Department of Periodontology and Implant Dentistry, College of Dentistry, Qassim University, Saudi Arabia
| |
Collapse
|
3
|
Hassan M, Abdelnabi HA, Mohsin S. Harnessing the Potential of PLGA Nanoparticles for Enhanced Bone Regeneration. Pharmaceutics 2024; 16:273. [PMID: 38399327 PMCID: PMC10892810 DOI: 10.3390/pharmaceutics16020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Recently, nanotechnologies have become increasingly prominent in the field of bone tissue engineering (BTE), offering substantial potential to advance the field forward. These advancements manifest in two primary ways: the localized application of nanoengineered materials to enhance bone regeneration and their use as nanovehicles for delivering bioactive compounds. Despite significant progress in the development of bone substitutes over the past few decades, it is worth noting that the quest to identify the optimal biomaterial for bone regeneration remains a subject of intense debate. Ever since its initial discovery, poly(lactic-co-glycolic acid) (PLGA) has found widespread use in BTE due to its favorable biocompatibility and customizable biodegradability. This review provides an overview of contemporary advancements in the development of bone regeneration materials using PLGA polymers. The review covers some of the properties of PLGA, with a special focus on modifications of these properties towards bone regeneration. Furthermore, we delve into the techniques for synthesizing PLGA nanoparticles (NPs), the diverse forms in which these NPs can be fabricated, and the bioactive molecules that exhibit therapeutic potential for promoting bone regeneration. Additionally, we addressed some of the current concerns regarding the safety of PLGA NPs and PLGA-based products available on the market. Finally, we briefly discussed some of the current challenges and proposed some strategies to functionally enhance the fabrication of PLGA NPs towards BTE. We envisage that the utilization of PLGA NP holds significant potential as a potent tool in advancing therapies for intractable bone diseases.
Collapse
Affiliation(s)
| | | | - Sahar Mohsin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
4
|
Mofarrah M, Jafari-Gharabaghlou D, Farhoudi-Sefidan-Jadid M, Zarghami N. Potential application of inorganic nano-materials in modulation of macrophage function: Possible application in bone tissue engineering. Heliyon 2023; 9:e16309. [PMID: 37292328 PMCID: PMC10245018 DOI: 10.1016/j.heliyon.2023.e16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
Nanomaterials indicate unique physicochemical properties for drug delivery in osteogenesis. Benefiting from high surface area grades, high volume ratio, ease of functionalization by biological targeting moieties, and small size empower nanomaterials to pass through biological barriers for efficient targeting. Inorganic nanomaterials for bone regeneration include inorganic synthetic polymers, ceramic nanoparticles, metallic nanoparticles, and magnetic nanoparticles. These nanoparticles can effectively modulate macrophage polarization and function, as one of the leading players in osteogenesis. Bone healing procedures in close cooperation with the immune system. Inflammation is one of the leading triggers of the bone fracture healing barrier. Macrophages commence anti-inflammatory signaling along with revascularization in the damaged site to promote the formation of a soft callus, bone mineralization, and bone remodeling. In this review, we will discuss the role of macrophages in bone hemostasis and regeneration. Furthermore, we will summarize the influence of the various inorganic nanoparticles on macrophage polarization and function in the benefit of osteogenesis.
Collapse
Affiliation(s)
- Mohsen Mofarrah
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Farhoudi-Sefidan-Jadid
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
5
|
Ilhan M, Kilicarslan M, Alcigir ME, Bagis N, Ekim O, Orhan K. Clindamycin phosphate and bone morphogenetic protein-7 loaded combined nanoparticle-graft and nanoparticle-film formulations for alveolar bone regeneration - An in vitro and in vivo evaluation. Int J Pharm 2023; 636:122826. [PMID: 36918117 DOI: 10.1016/j.ijpharm.2023.122826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Commonly utilized techniques for healing alveolar bone destruction such as the use of growth factors, suffering from short half-life, application difficulties, and the ability to achieve bioactivity only in the presence of high doses of growth factor. The sustained release of growth factors through a scaffold-based delivery system offers a promising and innovative tool in dentistry. Furthermore, it is suggested to guide the host response by using antimicrobials together with growth factors to prevent recovery and achieve ideal regeneration. Herein, the aim was to prepare and an in vitro - in vivo evaluation of bone morphogenetic protein 7 (BMP-7) and clindamycin phosphate (CDP) loaded polymeric nanoparticles, and their loading into the alginate-chitosan polyelectrolyte complex film or alloplastic graft to accelerate hard tissue regeneration. PLGA nanoparticles containing CDP and BMP-7, separately or together, were prepared using the double emulsion solvent evaporation technique. Through in vitro assays, it was revealed that spherical particles were homogeneously distributed in the combination formulations, and sustained release could be achieved for >12 weeks with all formulations. Also, results from the micro-CT and histopathological analyses indicated that CDP and BMP-7 loaded nanoparticle-film formulations were more effective in treatment than the nanoparticle loaded grafts.
Collapse
Affiliation(s)
- Miray Ilhan
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06560 Ankara, Türkiye; Duzce University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 81620 Duzce, Türkiye.
| | - Muge Kilicarslan
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06560 Ankara, Türkiye.
| | - Mehmet Eray Alcigir
- Kirikkale University, Faculty of Veterinary Medicine, Department of Pathology, 71450 Kirikkale, Türkiye.
| | - Nilsun Bagis
- Ankara University, Faculty of Dentistry, Department of Periodontology, 06560 Ankara, Türkiye.
| | - Okan Ekim
- Ankara University, Faculty of Veterinary Medicine, Department of Anatomy, 06110 Ankara, Türkiye.
| | - Kaan Orhan
- Ankara University, Faculty of Dentistry, Department of Dentomaxillofacial Radiology, 06560 Ankara, Türkiye.
| |
Collapse
|
6
|
Bian Y, Hu T, Lv Z, Xu Y, Wang Y, Wang H, Zhu W, Feng B, Liang R, Tan C, Weng X. Bone tissue engineering for treating osteonecrosis of the femoral head. EXPLORATION (BEIJING, CHINA) 2023; 3:20210105. [PMID: 37324030 PMCID: PMC10190954 DOI: 10.1002/exp.20210105] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/12/2022] [Indexed: 06/16/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and complicated disease with an unclear etiology. Femoral head-preserving surgeries have been devoted to delaying and hindering the collapse of the femoral head since their introduction in the last century. However, the isolated femoral head-preserving surgeries cannot prevent the natural progression of ONFH, and the combination of autogenous or allogeneic bone grafting often leads to many undesired complications. To tackle this dilemma, bone tissue engineering has been widely developed to compensate for the deficiencies of these surgeries. During the last decades, great progress has been made in ingenious bone tissue engineering for ONFH treatment. Herein, we comprehensively summarize the state-of-the-art progress made in bone tissue engineering for ONFH treatment. The definition, classification, etiology, diagnosis, and current treatments of ONFH are first described. Then, the recent progress in the development of various bone-repairing biomaterials, including bioceramics, natural polymers, synthetic polymers, and metals, for treating ONFH is presented. Thereafter, regenerative therapies for ONFH treatment are also discussed. Finally, we give some personal insights on the current challenges of these therapeutic strategies in the clinic and the future development of bone tissue engineering for ONFH treatment.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Tingting Hu
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yingjie Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Wei Zhu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Bin Feng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Chaoliang Tan
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Lee S, Park H, Oh JS, Byun K, Kim DY, Yun HS, Kang BJ. Hydroxyapatite microbeads containing BMP-2 and quercetin fabricated via electrostatic spraying to encourage bone regeneration. Biomed Eng Online 2023; 22:15. [PMID: 36803418 PMCID: PMC9938985 DOI: 10.1186/s12938-023-01078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/10/2023] [Indexed: 02/20/2023] Open
Abstract
BACKGROUND Hydroxyapatite (HAp) possesses osteoconductive properties, and its granular form can serve as an effective drug delivery vehicle for bone regeneration. Quercetin (Qct), a plant-derived bioflavonoid, is known to promote bone regeneration; however, its comparative and synergistic effects with the commonly used bone morphogenetic protein-2 (BMP-2) have not been investigated. METHODS We examined the characteristics of newly formed HAp microbeads using an electrostatic spraying method and analyzed the in vitro release pattern and osteogenic potential of ceramic granules containing Qct, BMP-2, and both. In addition, HAp microbeads were transplanted into a rat critical-sized calvarial defect and the osteogenic capacity was assessed in vivo. RESULTS The manufactured beads had a microscale size of less than 200 μm, a narrow size distribution, and a rough surface. The alkaline phosphatase (ALP) activity of osteoblast-like cells cultured with the BMP-2-and-Qct-loaded HAp was significantly higher than that of either Qct- or BMP-2-loaded HAp groups. The mRNA levels of osteogenic marker genes such as ALP and runt-related transcription factor 2 were found to be upregulated in the HAp/BMP-2/Qct group compared to the other groups. In micro-computed tomographic analysis, the amount of newly formed bone and bone surface area within the defect was significantly higher in the HAp/BMP-2/Qct group, followed by the HAp/BMP-2 and HAp/Qct groups, which is consistent with the histomorphometrical results. CONCLUSIONS These results imply that electrostatic spraying can be an efficient strategy to produce homogenous ceramic granules and that the BMP-2-and-Qct-loaded HAp microbeads can serve as effective implants for bone defect healing.
Collapse
Affiliation(s)
- Seoyun Lee
- grid.31501.360000 0004 0470 5905Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826 South Korea ,grid.31501.360000 0004 0470 5905BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826 South Korea
| | - Honghyun Park
- grid.410902.e0000 0004 1770 8726Department of Advanced Biomaterials Research, Ceramics Materials Division, Korea Institute of Materials Science (KIMS), Changwon, 51508 South Korea
| | - Jeong-Seop Oh
- grid.31501.360000 0004 0470 5905Department of Veterinary Pathology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826 South Korea
| | - Kyubin Byun
- grid.410902.e0000 0004 1770 8726Department of Advanced Biomaterials Research, Ceramics Materials Division, Korea Institute of Materials Science (KIMS), Changwon, 51508 South Korea ,grid.412786.e0000 0004 1791 8264Department of Advanced Materials Engineering, University of Science & Technology (UST), Daejeon, 34113 South Korea
| | - Dae-Yong Kim
- grid.31501.360000 0004 0470 5905Department of Veterinary Pathology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826 South Korea
| | - Hui-suk Yun
- grid.410902.e0000 0004 1770 8726Department of Advanced Biomaterials Research, Ceramics Materials Division, Korea Institute of Materials Science (KIMS), Changwon, 51508 South Korea ,grid.412786.e0000 0004 1791 8264Department of Advanced Materials Engineering, University of Science & Technology (UST), Daejeon, 34113 South Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea. .,BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
8
|
Khamkongkaeo A, Jiamprasertboon A, Jinakul N, Srabua P, Tantavisut S, Wongrakpanich A. Antibiotic-loaded hydroxyapatite scaffolds fabricated from Nile tilapia bones for orthopaedics. Int J Pharm X 2023; 5:100169. [PMID: 36861068 PMCID: PMC9969256 DOI: 10.1016/j.ijpx.2023.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/01/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
This work aimed to develop new antibiotic-coated/ antibiotic-loaded hydroxyapatite (HAp) scaffolds for orthopaedic trauma, specifically to treat the infection after fixation of skeletal fracture. The HAp scaffolds were fabricated from the Nile tilapia (Oreochromis niloticus) bones and fully characterized. The HAp scaffolds were coated with 12 formulations of poly (lactic-co-glycolic acid) (PLGA) or poly (lactic acid) (PLA), blended with vancomycin. The vancomycin release, surface morphology, antibacterial properties, and the cytocompatibility of the scaffolds were conducted. The HAp powder contains elements identical to those found in human bones. This HAp powder is suitable as a starting material to build scaffolds. After the scaffold fabrication, The ratio of HAp to β-TCP changed, and the phase transformation of β-TCP to α-TCP was observed. All antibiotic-coated/ antibiotic-loaded HAp scaffolds can release vancomycin into the phosphate-buffered saline (PBS) solution. PLGA-coated scaffolds obtained faster drug release profiles than PLA-coated scaffolds. The low polymer concentration in the coating solutions (20%w/v) gave a faster drug release profile than the high polymer concentration (40%w/v). All groups showed a trace of surface erosion after being submerged in PBS for 14 days. Most of the extracts can inhibit Staphylococcus aureus (S. aureus) and methicillin-resistant S. aureus (MRSA). The extracts not only caused no cytotoxicity to Saos-2 bone cells but also can increase cell growth. This study demonstrates that it is possible to use these antibiotic-coated/ antibiotic-loaded scaffolds in the clinic as an antibiotic bead replacement.
Collapse
Key Words
- Antibiotic
- Antibiotic-coated
- Antibiotic-loaded
- CLSI, The Clinical and Laboratory Standards Institute
- DI, Deionized water
- DMSO, Dimethyl sulfoxide
- F10[PLGA40-Hvanc], Formulation 10, HAp saffolds containing high concentration of vancomycin, coated with PLGA 40%w/v
- F11[PLA20-Hvanc], Formulation 11, HAp saffolds containing high concentration of vancomycin, coated with PLA 20%w/v
- F12[PLA40-Hvanc], Formulation 12, HAp saffolds containing high concentration of vancomycin, coated with PLA 40%w/v
- F1[V-PLGA20-Lvanc], Formulation 1, HAp saffolds containing low concentration of vancomycin, coated with PLGA 20%w/v blended with vancomycin
- F2[V-PLGA40-Lvanc], Formulation 2, HAp saffolds containing low concentration of vancomycin, coated with PLGA 40%w/v blended with vancomycin
- F3[V-PLA20-Lvanc], Formulation 3, HAp saffolds containing low concentration of vancomycin, coated with PLA 20%w/v blended with vancomycin
- F4[V-PLA40-Lvanc], Formulation 4, HAp saffolds containing low concentration of vancomycin, coated with PLA 40%w/v blended with vancomycin
- F5[PLGA20-Lvanc], Formulation 5, HAp saffolds containing low concentration of vancomycin, coated with PLGA 20%w/v
- F6[PLGA40-Lvanc], Formulation 6, HAp saffolds containing low concentration of vancomycin, coated with PLGA 40%w/v
- F7[PLA20-Lvanc], Formulation 7, HAp saffolds containing low concentration of vancomycin, coated with PLA 20%w/v
- F8[PLA40-Lvanc], Formulation 8, HAp saffolds containing low concentration of vancomycin, coated with PLA 40%w/v
- F9[PLGA20-Hvanc], Formulation 9, HAp saffolds containing high concentration of vancomycin, coated with PLGA 20%w/v
- FDA, Food and Drug Administration
- FTIR, Fourier transforms infrared spectroscopy
- HAp, Hydroxyapatite
- Hydroxyapatite
- IFSF, The infection after fixation of skeletal fracture
- Nile tilapia
- P.U., Polyurethane
- PBS, Phosphate-buffered saline
- PLA, Poly(lactic acid)
- PLGA, Poly(lactic-co-glycolic acid)
- PVA, Polyvinyl alcohol
- SEM, Scanning electron microscopy
- Scaffold
- Vancomycin
- XRD, X-ray diffraction
- XRF, X-ray fluorescence spectroscopy
- α-TCP, α-tricalcium phosphate
- β-TCP, β-tricalcium phosphate
Collapse
Affiliation(s)
- Atchara Khamkongkaeo
- Department of Metallurgical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
| | - Arreerat Jiamprasertboon
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand,Institute of Research and Development, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Nanthawan Jinakul
- Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Phatraya Srabua
- Scientific and Technological Research Equipment Center (STREC), Chulalongkorn University, Bangkok, Thailand
| | - Saran Tantavisut
- Department of Orthopaedics, Chulalongkorn University, Bangkok, Thailand,Hip Fracture Research Unit, Department of Orthopaedics, Chulalongkorn University, Bangkok, Thailand
| | - Amaraporn Wongrakpanich
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Corresponding author.
| |
Collapse
|
9
|
He Y, de Araújo Júnior RF, Cavalcante RS, Yu Z, Schomann T, Gu Z, Eich C, Cruz LJ. Effective breast cancer therapy based on palmitic acid-loaded PLGA nanoparticles. BIOMATERIALS ADVANCES 2023; 145:213270. [PMID: 36603405 DOI: 10.1016/j.bioadv.2022.213270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Although new strategies for breast cancer treatment have yielded promising results, most drugs can lead to serious side effects when applied systemically. Doxorubicin (DOX), currently the most effective chemotherapeutic drug to treat breast cancer, is poorly selective towards tumor cells and treatment often leads to the development of drug resistance. Recent studies have indicated that several fatty acids (FAs) have beneficial effects on inhibiting tumorigenesis. The saturated FA palmitic acid (PA) showed anti-tumor activities in several types of cancer, as well as effective repolarization of M2 macrophages towards the anti-tumorigenic M1 phenotype. However, water insolubility and cellular impermeability limit the use of PA in vivo. To overcome these limitations, here, we encapsulated PA into a poly(d,l-lactic co-glycolic acid) (PLGA) nanoparticle (NP) platform, alone and in combination with DOX, to explore PA's potential as mono or combinational breast cancer therapy. Our results showed that PLGA-PA-DOX NPs and PLGA-PA NPs significantly reduced the viability and migratory capacity of breast cancer cells in vitro. In vivo studies in mice bearing mammary tumors demonstrated that PLGA-PA-NPs were as effective in reducing primary tumor growth and metastasis as NPs loaded with DOX, PA and DOX, or free DOX. At the molecular level, PLGA-PA NPs reduced the expression of genes associated with multi-drug resistance and inhibition of apoptosis, and induced apoptosis via a caspase-3-independent pathway in breast cancer cells. In addition, immunohistochemical analysis of residual tumors showed a reduction in M2 macrophage content and infiltration of leukocytes after treatment of PLGA-PA NPs and PLGA-PA-DOX NPs, suggesting immunomodulatory properties of PA in the tumor microenvironment. In conclusion, the use of PA alone or in combination with DOX may represent a promising novel strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yuanyuan He
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Raimundo Fernandes de Araújo Júnior
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, 59064-720, Brazil; Cancer and Inflammation Research Laboratory (LAICI), Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal, 59064-720, Brazil; Percuros B.V., 2333, CL, Leiden, the Netherlands
| | - Rômulo S Cavalcante
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, 59064-720, Brazil; Cancer and Inflammation Research Laboratory (LAICI), Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal, 59064-720, Brazil
| | - Zhenfeng Yu
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Timo Schomann
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands; Percuros B.V., 2333, CL, Leiden, the Netherlands
| | - Zili Gu
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands
| | - Christina Eich
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands.
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333, ZA, Leiden, the Netherlands.
| |
Collapse
|
10
|
Liu Y, Amissah OB, Huangfang X, Wang L, Dieu Habimana JD, Lv L, Ding X, Li J, Chen M, Zhu J, Mukama O, Sun Y, Li Z, Huang R. Large-scale expansion of human umbilical cord-derived mesenchymal stem cells using PLGA@PLL scaffold. BIORESOUR BIOPROCESS 2023; 10:18. [PMID: 36915643 PMCID: PMC9994782 DOI: 10.1186/s40643-023-00635-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/31/2023] [Indexed: 03/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are highly important in biomedicine and hold great potential in clinical treatment for various diseases. In recent years, the capabilities of MSCs have been under extensive investigation for practical application. Regarding therapy, the efficacy usually depends on the amount of MSCs. Nevertheless, the yield of MSCs is still limited due to the traditional cultural methods. Herein, we proposed a three-dimensional (3D) scaffold prepared using poly lactic-co-glycolic acid (PLGA) nanofiber with polylysine (PLL) grafting, to promote the growth and proliferation of MSCs derived from the human umbilical cord (hUC-MSCs). We found that the inoculated hUC-MSCs adhered efficiently to the PLGA scaffold with good affinity, fast growth rate, and good multipotency. The harvested cells were ideally distributed on the scaffold and we were able to gain a larger yield than the traditional culturing methods under the same condition. Thus, our cell seeding with a 3D scaffold could serve as a promising strategy for cell proliferation in the large-scale production of MSCs. Moreover, the simplicity and low preparation cost allow this 3D scaffold to extend its potential application beyond cell culture. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1186/s40643-023-00635-6.
Collapse
Affiliation(s)
- Yujie Liu
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,Guangzhou Junyuankang Biotechnology Co., Ltd., Guangzhou, 510530 China
| | - Obed Boadi Amissah
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | | | - Ling Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China
| | - Jean de Dieu Habimana
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Linshuang Lv
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xuanyan Ding
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Junyi Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ming Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China
| | - Jinmin Zhu
- GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| | - Omar Mukama
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Yirong Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Zhiyuan Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China.,School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China.,GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China.,GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013 China
| | - Rongqi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| |
Collapse
|
11
|
Breast Cancer and Arsenic Anticancer Effects: Systematic Review of the Experimental Data from In Vitro Studies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8030931. [PMID: 36619302 PMCID: PMC9815927 DOI: 10.1155/2022/8030931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/31/2022]
Abstract
Arsenic is a known environmental carcinogenic agent. However, under certain circumstances, it may exert anticancer effects. In this systematic review, we aim to provide information on recent developments in studies on arsenic antitumor effects in breast cancer. Research included in the review refers to experimental data from in vitro studies. The data was collected using search terms "breast cancer," "arsenic," and "anticancer" (25.05.2021). Only studies in English and published in the last 10 years were included. The search identified 123 studies from the EBSCOhost, PubMed, and Scopus databases. In the selection process, thirty full-texts were evaluated as eligible for the review. The literature of the last decade provides a lot of information on mechanisms behind anticancer effects of arsenic on breast cancer. Similar to arsenic-induced carcinogenesis, these mechanisms include the activation of the redox system and the increased production of free radicals. Targets of arsenic action are systems of cell membranes, mitochondria, pathways of intracellular transmission, and the genetic apparatus of the cell. Beneficial effects of arsenic use are possible due to significant metabolic differences between cancer and healthy cells. Further efforts are needed in order to establish modes and doses of treatment with arsenic that would provide anticancer activity with minimal toxicity.
Collapse
|
12
|
Chen H, Zhang Y, Yu T, Song G, Xu T, Xin T, Lin Y, Han B. Nano-Based Drug Delivery Systems for Periodontal Tissue Regeneration. Pharmaceutics 2022; 14:2250. [PMID: 36297683 PMCID: PMC9612159 DOI: 10.3390/pharmaceutics14102250] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2022] Open
Abstract
Periodontitis is a dysbiotic biofilm-induced and host-mediated inflammatory disease of tooth supporting tissues that leads to progressive destruction of periodontal ligament and alveolar bone, thereby resulting in gingival recession, deep periodontal pockets, tooth mobility and exfoliation, and aesthetically and functionally compromised dentition. Due to the improved biopharmaceutical and pharmacokinetic properties and targeted and controlled drug release, nano-based drug delivery systems have emerged as a promising strategy for the treatment of periodontal defects, allowing for increased efficacy and safety in controlling local inflammation, establishing a regenerative microenvironment, and regaining bone and attachments. This review provides an overview of nano-based drug delivery systems and illustrates their practical applications, future prospects, and limitations in the field of periodontal tissue regeneration.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunfan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tingting Yu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Guangying Song
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianmin Xu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianyi Xin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yifan Lin
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Bing Han
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
13
|
Cao Y, Ahmed AMQ, Du HH, Sun W, Lu X, Xu Z, Tao J, Cao QR. Combretastatin A4-loaded Poly (Lactic-co-glycolic Acid)/Soybean Lecithin Nanoparticles with Enhanced Drug Dissolution Rate and Antiproliferation Activity. Curr Drug Deliv 2022; 19:918-927. [PMID: 35139789 DOI: 10.2174/1567201819666220209093443] [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: 06/16/2021] [Revised: 08/24/2021] [Accepted: 12/12/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed to prepare combretastatin A4 (CA4)-loaded nanoparticles (CA4 NPs) using poly(lactic-co-glycolic acid) (PLGA) and soybean lecithin (Lipoid S100) as carriers, and further evaluate the physicochemical properties and cytotoxicities of CA4 NPs against cancer cells. METHODS CA4 NPs were prepared using a solvent evaporation technique. The effects of formulations on CA4 NPs were investigated in terms of particle size, zeta potential, encapsulation efficacy, and drug loading. The physicochemical properties of CA4 NPs were characterized using transmission electron microscopy, X-ray powder diffraction, differential scanning calorimetry, and Fourier transform infrared spectra. The drug release from CA4NPs was performed using a dialysis method. In addition, the cytotoxicity of CA4NPs against human alveolar basal epithelial (A549) cells was also evaluated. RESULTS CA4 NPs prepared with a low organic/water phase ratio (1:20) and high drug/PLGA mass ratio (1:2.5) exhibited a uniform hydrodynamic particle size of 142 nm, the zeta potential of -1.66 mV, and encapsulation efficacy and drug loading of 92.1% and 28.3%, respectively. CA4 NPs showed a significantly higher release rate than pure CA4 in pH 7.4 phosphate-buffered solution with 0.5% Tween 80. It was found that the drug molecules could change from the crystal state to an amorphous form when loaded into the PLGA/Lipoid S100 matrix, and some molecular interactions could also occur between the drug and PLGA. Importantly, CA4 NPs showed a remarkably higher antiproliferation activity against A549 cancer cells compared to pure CA4. CONCLUSION These results suggested the promising potential of PLGA/Lipoid S100 nanoparticles as the drug delivery system of CA4 for effective cancer therapy.
Collapse
Affiliation(s)
- Yue Cao
- Department of Pharmacy, Beijing Health Vocational College, Beijing 100053, People's Republic of China
| | | | - Huan-Huan Du
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Wei Sun
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaojuan Lu
- PharmaMax Pharmaceuticals, Ltd., China Medical City, Taizhou 225300, People's Republic of China
| | - Zhao Xu
- PharmaMax Pharmaceuticals, Ltd., China Medical City, Taizhou 225300, People's Republic of China
| | - Jing Tao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Qing-Ri Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
14
|
Polymer Texture Influences Cell Responses in Osteogenic Microparticles. Cell Mol Bioeng 2022; 15:409-423. [DOI: 10.1007/s12195-022-00729-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
|
15
|
Higino T, França R. Drug-delivery nanoparticles for bone-tissue and dental applications. Biomed Phys Eng Express 2022; 8. [PMID: 35439740 DOI: 10.1088/2057-1976/ac682c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/19/2022] [Indexed: 11/11/2022]
Abstract
The use of nanoparticles as biomaterials with applications in the biomedical field is growing every day. These nanomaterials can be used as contrast imaging agents, combination therapy agents, and targeted delivery systems in medicine and dentistry. Usually, nanoparticles are found as synthetic or natural organic materials, such as hydroxyapatite, polymers, and lipids. Besides that, they are could also be inorganic, for instance, metallic or metal-oxide-based particles. These inorganic nanoparticles could additionally present magnetic properties, such as superparamagnetic iron oxide nanoparticles. The use of nanoparticles as drug delivery agents has many advantages, for they help diminish toxicity effects in the body since the drug dose reduces significantly, increases drugs biocompatibility, and helps target drugs to specific organs. As targeted-delivery agents, one of the applications uses nanoparticles as drug delivery particles for bone-tissue to treat cancer, osteoporosis, bone diseases, and dental treatments such as periodontitis. Their application as drug delivery agents requires a good comprehension of the nanoparticle properties and composition, alongside their synthesis and drug attachment characteristics. Properties such as size, shape, core-shell designs, and magnetic characteristics can influence their behavior inside the human body and modify magnetic properties in the case of magnetic nanoparticles. Based on that, many different studies have modified the synthesis methods for these nanoparticles and developed composite systems for therapeutics delivery, adapting, and improving magnetic properties, shell-core designs, and particle size and nanosystems characteristics. This review presents the most recent studies that have been presented with different nanoparticle types and structures for bone and dental drug delivery.
Collapse
Affiliation(s)
- Taisa Higino
- Biomedical Engineering Program, University of Manitoba, Winnipeg, Canada
| | - Rodrigo França
- Biomedical Engineering Program, University of Manitoba, Winnipeg, Canada.,Dental Biomaterials Research Lab, Department of Restorative Dentistry, College of Dentistry, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
16
|
Current Advances of Nanomedicines Delivering Arsenic Trioxide for Enhanced Tumor Therapy. Pharmaceutics 2022; 14:pharmaceutics14040743. [PMID: 35456577 PMCID: PMC9026299 DOI: 10.3390/pharmaceutics14040743] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/18/2022] Open
Abstract
Arsenic trioxide (ATO) is one of the first-line chemotherapeutic drugs for acute promyelocytic leukemia. Its anti-cancer activities against various human neoplastic diseases have been extensively studied. However, the clinical use of ATO for solid tumors is limited, and these limitations are because of severe systemic toxicity, low bioavailability, and quick renal elimination before it reaches the target site. Although without much success, several efforts have been made to boost ATO bioavailability toward solid tumors without raising its dose. It has been found that nanomedicines have various advantages for drug delivery, including increased bioavailability, effectiveness, dose-response, targeting capabilities, and safety as compared to traditional drugs. Therefore, nanotechnology to deliver ATO to solid tumors is the main topic of this review, which outlines the previous and present medical applications of ATO. We also summarised ATO anti-cancer mechanisms, limitations, and outcomes of combinatorial treatment with chemo agents. As a result, we strongly recommend conducting pre-clinical and clinical studies of ATO, especially nano-system-based ones that might lead to a novel combination therapy for cancer treatment with high efficacy, bioavailability, and low toxicity for cancer patients.
Collapse
|
17
|
Controlling the Spatiotemporal Release of Nerve Growth Factor by Chitosan/Polycaprolactone Conduits for Use in Peripheral Nerve Regeneration. Int J Mol Sci 2022; 23:ijms23052852. [PMID: 35269991 PMCID: PMC8911064 DOI: 10.3390/ijms23052852] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Tubular polymeric structures have been recognized in the treatment of peripheral nerves as comparable to autologous grafting. The best therapeutic outcomes are obtained with conduits releasing therapeutic molecules. In this study, a new approach for the incorporation of biologically active agent-loaded microspheres into the structure of chitosan/polycaprolactone conduits was developed. The support of a polycaprolactone helix formed by 3D melt extrusion was coated with dopamine in order to adsorb nerve growth factor-loaded microspheres. The complex analysis of the influence of process factors on the coverage efficiency of polycaprolactone helix by nerve grow factor-loaded microspheres was analyzed. Thus, the PCL helix characterized with the highest adsorption of microspheres was subjected to nerve growth factor release studies, and finally incorporated into chitosan hydrogel deposit through the process of electrophoretic deposition. It was demonstrated by chemical and physical tests that the chitosan/polycaprolactone conduit meets the requirements imposed on peripheral nerve implants, particularly mimicking mechanical properties of surrounding soft tissue. Moreover, the conduit may support regrowing nerves for a prolonged period, as its structure and integrity persist upon incubation in lysozyme-contained PBS solution up to 28 days at body temperature. In vitro cytocompatibility toward mHippoE-18 embryonic hippocampal cells of the chitosan/polycaprolactone conduit was proven. Most importantly, the developed conduits stimulate axonal growth and support monocyte activation, the latter is advantageous especially at early stages of nerve regeneration. It was demonstrated that, through the described approach for controlling spatiotemporal release of nerve growth factors, these biocompatible structures adjusted to the specific peripheral nerve injury case can be manufactured.
Collapse
|
18
|
Abstract
Bone regeneration is a central focus of maxillofacial research, especially when dealing with dental implants or critical sized wound sites. While bone has great regeneration potential, exogenous delivery of growth factors can greatly enhance the speed, duration, and quality of osseointegration, making a difference in a patient’s quality of life. Bone morphogenic protein 2 (BMP-2) is a highly potent growth factor that acts as a recruiting molecule for mesenchymal stromal cells, induces a rapid differentiation of them into osteoblasts, while also maintaining their viability. Currently, the literature data shows that the liposomal direct delivery or transfection of plasmids containing BMP-2 at the bone wound site often results in the overexpression of osteogenic markers and result in enhanced mineralization with formation of new bone matrix. We reviewed the literature on the scientific data regarding BMP-2 delivery with the help of liposomes. This may provide the ground for a future new bone regeneration strategy with real chances of reaching clinical practice.
Collapse
|
19
|
Encapsulation of Large-Size Plasmids in PLGA Nanoparticles for Gene Editing: Comparison of Three Different Synthesis Methods. NANOMATERIALS 2021; 11:nano11102723. [PMID: 34685164 PMCID: PMC8541650 DOI: 10.3390/nano11102723] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
The development of new gene-editing technologies has fostered the need for efficient and safe vectors capable of encapsulating large nucleic acids. In this work we evaluate the synthesis of large-size plasmid-loaded PLGA nanoparticles by double emulsion (considering batch ultrasound and microfluidics-assisted methodologies) and magnetic stirring-based nanoprecipitation synthesis methods. For this purpose, we characterized the nanoparticles and compared the results between the different synthesis processes in terms of encapsulation efficiency, morphology, particle size, polydispersity, zeta potential and structural integrity of loaded pDNA. Our results demonstrate particular sensibility of large pDNA for shear and mechanical stress degradation during double emulsion, the nanoprecipitation method being the only one that preserved plasmid integrity. However, plasmid-loaded PLGA nanoparticles synthesized by nanoprecipitation did not show cell expression in vitro, possibly due to the slow release profile observed in our experimental conditions. Strong electrostatic interactions between the large plasmid and the cationic PLGA used for this synthesis may underlie this release kinetics. Overall, none of the methods evaluated satisfied all the requirements for an efficient non-viral vector when applied to large-size plasmid encapsulation. Further optimization or alternative synthesis methods are thus in current need to adapt PLGA nanoparticles as delivery vectors for gene editing therapeutic technologies.
Collapse
|
20
|
Kim SM, Patel M, Patel R. PLGA Core-Shell Nano/Microparticle Delivery System for Biomedical Application. Polymers (Basel) 2021; 13:3471. [PMID: 34685230 PMCID: PMC8540999 DOI: 10.3390/polym13203471] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/24/2022] Open
Abstract
Core-shell particles are very well known for their unique features. Their distinctive inner core and outer shell structure allowed promising biomedical applications at both nanometer and micrometer scales. The primary role of core-shell particles is to deliver the loaded drugs as they are capable of sequence-controlled release and provide protection of drugs. Among other biomedical polymers, poly (lactic-co-glycolic acid) (PLGA), a food and drug administration (FDA)-approved polymer, has been recognized for the vehicle material. This review introduces PLGA core-shell nano/microparticles and summarizes various drug-delivery systems based on these particles for cancer therapy and tissue regeneration. Tissue regeneration mainly includes bone, cartilage, and periodontal regeneration.
Collapse
Affiliation(s)
- Se Min Kim
- Life Science and Biotechnology Department (LSBT), Underwood Division (UD), Underwood International College, Yonsei University, Sinchon, Seoul 03722, Korea;
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Woman’s University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea;
| | - Rajkumar Patel
- Energy and Environmental Science and Engineering (EESE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdogwahak-ro, Yeonsugu, Incheon 21983, Korea
| |
Collapse
|
21
|
Fereig SA, El-Zaafarany GM, Arafa MG, Abdel-Mottaleb MMA. Tacrolimus-loaded chitosan nanoparticles for enhanced skin deposition and management of plaque psoriasis. Carbohydr Polym 2021; 268:118238. [PMID: 34127220 DOI: 10.1016/j.carbpol.2021.118238] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/18/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
Tacrolimus is a natural macrolide that exhibits an anti-proliferative action by T-lymphocytic cells inhibition. Hence, it was tested as a potential topical treatment to improve and control psoriatic plaques. In this study, for the first time the lipophilic tacrolimus in chitosan nanoparticles was used to achieve the desired response and dermal retention of the drug using a modified ionic gelation technique. The hydrophobic drug, tacrolimus, was successfully encapsulated into the synthesized positively-charged particles (140.8 nm ± 50.0) and EE of (65.5% ± 1.3). Local skin deposition of the drug was significantly enhanced with 82.0% ± 0.6 of the drug retained in the skin compared to 34.0% ± 0.9 from tarolimus® ointment. An outstanding response to the prepared formula was the enhanced hair growth rate in the treated animals, which can be considered an excellent sign of the skin recovery from the induced psoriatic plaques after only three days of treatment.
Collapse
Affiliation(s)
- Salma A Fereig
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt (BUE), El Sherouk City, Egypt
| | - Ghada M El-Zaafarany
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona G Arafa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt (BUE), El Sherouk City, Egypt; Chemotherapeutic Unit, Mansoura University Hospitals, Mansoura, Egypt
| | - Mona M A Abdel-Mottaleb
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
22
|
Song H, Zhang Y, Zhang Z, Xiong S, Ma X, Li Y. Hydroxyapatite/NELL-1 Nanoparticles Electrospun Fibers for Osteoinduction in Bone Tissue Engineering Application. Int J Nanomedicine 2021; 16:4321-4332. [PMID: 34211273 PMCID: PMC8241815 DOI: 10.2147/ijn.s309567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Background As commonly bone defect is a disease of jaw that can seriously affect implant restoration, the bioactive scaffold can be used as potential systems to provide effective repair for bone defect. Purpose A osteoinductive bone tissue engineering scaffold has been prepared in order to explore the effect of bioactive materials on bone tissue engineering. Methods In this study, NELL-1 nanoparticles (Chi/NNP) and nano hydroxyapatite were incorporated in composite scaffolds by electrospinning and characterized using TEM, SEM, contact angle, tensile tests and in vitro drug release. In vitro biological activities such as MC3T3-E1 cell attachment, proliferation and osteogenic activity were studied. Results With the addition of nHA and nanoparticles, the fiber diameter of PCL/BNPs group, PCL/NNPs group and PCL/nHA/NNPs group was significantly increased. Moreover, the hydrophilic hydroxyl group and amino group presented in nHA and nanoparticles had improved the hydrophilicity of the composite fibers. The composite electrospun containing Chi/NNPs can form a double protective barrier which can effectively prolong the release time of NELL-1 growth factor. In addition, the hydroxyapatite/NELL-1 nanoparticles electrospun fibers can promote attachment, proliferation, differentiation of MC3T3-E1 cells and good cytocompatibility, indicating better ability of inducing osteogenic differentiation. Conclusion A multi-functional PCL/nHA/NNPs composite fiber with long-term bioactivity and osteoinductivity was successfully prepared by electrospinning. This potential composite could be used as scaffolds in bone tissue engineering application after in vivo studies.
Collapse
Affiliation(s)
- Hualei Song
- Department of Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Yuntao Zhang
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Zihan Zhang
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Shijiang Xiong
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, 250012, People's Republic of China
| | - Xiangrui Ma
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Yourui Li
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| |
Collapse
|
23
|
Cheng L, Suresh K S, He H, Rajput RS, Feng Q, Ramesh S, Wang Y, Krishnan S, Ostrovidov S, Camci-Unal G, Ramalingam M. 3D Printing of Micro- and Nanoscale Bone Substitutes: A Review on Technical and Translational Perspectives. Int J Nanomedicine 2021; 16:4289-4319. [PMID: 34211272 PMCID: PMC8239380 DOI: 10.2147/ijn.s311001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
Recent developments in three-dimensional (3D) printing technology offer immense potential in fabricating scaffolds and implants for various biomedical applications, especially for bone repair and regeneration. As the availability of autologous bone sources and commercial products is limited and surgical methods do not help in complete regeneration, it is necessary to develop alternative approaches for repairing large segmental bone defects. The 3D printing technology can effectively integrate different types of living cells within a 3D construct made up of conventional micro- or nanoscale biomaterials to create an artificial bone graft capable of regenerating the damaged tissues. This article reviews the developments and applications of 3D printing in bone tissue engineering and highlights the numerous conventional biomaterials and nanomaterials that have been used in the production of 3D-printed scaffolds. A comprehensive overview of the 3D printing methods such as stereolithography (SLA), selective laser sintering (SLS), fused deposition modeling (FDM), and ink-jet 3D printing, and their technical and clinical applications in bone repair and regeneration has been provided. The review is expected to be useful for readers to gain an insight into the state-of-the-art of 3D printing of bone substitutes and their translational perspectives.
Collapse
Affiliation(s)
- Lijia Cheng
- School of Basic Medicine, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Shoma Suresh K
- Biomaterials and Organ Engineering Group, Centre for Biomaterials, Cellular, and Molecular Theranostics, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Hongyan He
- School of Basic Medicine, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Ritu Singh Rajput
- Biomaterials and Organ Engineering Group, Centre for Biomaterials, Cellular, and Molecular Theranostics, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Qiyang Feng
- School of Basic Medicine, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Saravanan Ramesh
- Biomaterials and Organ Engineering Group, Centre for Biomaterials, Cellular, and Molecular Theranostics, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Yuzhuang Wang
- School of Basic Medicine, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Sasirekha Krishnan
- Biomaterials and Organ Engineering Group, Centre for Biomaterials, Cellular, and Molecular Theranostics, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Serge Ostrovidov
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Murugan Ramalingam
- Biomaterials and Organ Engineering Group, Centre for Biomaterials, Cellular, and Molecular Theranostics, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| |
Collapse
|
24
|
Zhang Y, Wu D, Zhao X, Pakvasa M, Tucker AB, Luo H, Qin KH, Hu DA, Wang EJ, Li AJ, Zhang M, Mao Y, Sabharwal M, He F, Niu C, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Chen C, Wagstaff W, Reid RR, Athiviraham A, Ho S, Lee MJ, Hynes K, Strelzow J, He TC, El Dafrawy M. Stem Cell-Friendly Scaffold Biomaterials: Applications for Bone Tissue Engineering and Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:598607. [PMID: 33381499 PMCID: PMC7767872 DOI: 10.3389/fbioe.2020.598607] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic organ with high regenerative potential and provides essential biological functions in the body, such as providing body mobility and protection of internal organs, regulating hematopoietic cell homeostasis, and serving as important mineral reservoir. Bone defects, which can be caused by trauma, cancer and bone disorders, pose formidable public health burdens. Even though autologous bone grafts, allografts, or xenografts have been used clinically, repairing large bone defects remains as a significant clinical challenge. Bone tissue engineering (BTE) emerged as a promising solution to overcome the limitations of autografts and allografts. Ideal bone tissue engineering is to induce bone regeneration through the synergistic integration of biomaterial scaffolds, bone progenitor cells, and bone-forming factors. Successful stem cell-based BTE requires a combination of abundant mesenchymal progenitors with osteogenic potential, suitable biofactors to drive osteogenic differentiation, and cell-friendly scaffold biomaterials. Thus, the crux of BTE lies within the use of cell-friendly biomaterials as scaffolds to overcome extensive bone defects. In this review, we focus on the biocompatibility and cell-friendly features of commonly used scaffold materials, including inorganic compound-based ceramics, natural polymers, synthetic polymers, decellularized extracellular matrix, and in many cases, composite scaffolds using the above existing biomaterials. It is conceivable that combinations of bioactive materials, progenitor cells, growth factors, functionalization techniques, and biomimetic scaffold designs, along with 3D bioprinting technology, will unleash a new era of complex BTE scaffolds tailored to patient-specific applications.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Andrew Blake Tucker
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Laboratory Diagnostic Medicine, The Affiliated Hospital of the University of Chinese Academy of Sciences, Chongqing General Hospital, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery Section of Plastic and Reconstructive Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
25
|
Tan W, Gao C, Feng P, Liu Q, Liu C, Wang Z, Deng Y, Shuai C. Dual-functional scaffolds of poly(L-lactic acid)/nanohydroxyapatite encapsulated with metformin: Simultaneous enhancement of bone repair and bone tumor inhibition. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111592. [PMID: 33545810 DOI: 10.1016/j.msec.2020.111592] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/01/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022]
Abstract
Bone defects caused by tumors are difficult to repair clinically because of their poor morphology and residual tumor cell-induced recurrence. Scaffolds with the dual function of bone repair and bone tumor treatment are urgently needed to resolve this problem. In this study, a poly(L-lactic acid) (PLLA)/nanoscale hydroxyapatite (nHA)/metformin (MET) nanocomposite scaffold was constructed via selective laser sintering. The scaffolds were expected to combine the excellent mechanical strength and biodegradability of PLLA, the good bioactivity of nHA, and the water solubility and antitumor properties of MET. The PLLA/nHA/MET scaffolds showed improved cell adhesion, appropriate porosity, good biocompatibility and osteogenic-induced ability in vitro because metformin improves water solubility and promotes the osteogenic differentiation of cells within the scaffold. The PLLA/nHA/MET scaffold had an extended drug release time because the MET particles were wrapped in the biodegradable polymer PLLA and the wrapped MET particles were slowly released into body fluids as the PLLA was degraded. Moreover, the scaffold induced osteosarcoma (OS) cell apoptosis by upregulating apoptosis-related gene expression and showed excellent tumor inhibition characteristics in vitro. In addition, the scaffold induced osteogenic differentiation of bone marrow mesenchymal cells (BMSCs) by promoting osteogenic gene expression. The results suggest that the PLLA/nHA/MET composite scaffold has the dual function of tumor inhibition and bone repair and therefore it provides a promising new approach for the treatment of tumor-induced bone defects.
Collapse
Affiliation(s)
- Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013, People's Republic of China; Department of Spinal Orthopedics, Huizhou Third People's Hospital, Guangzhou Medical University, No.1, Xuebei Road, Huizhou, Guangdong 516002, People's Republic of China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, Hunan, People's Republic of China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, Hunan, People's Republic of China
| | - Qing Liu
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013, People's Republic of China
| | - Congcong Liu
- Department of Spine Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| | - Zhenting Wang
- Department of Urinary Surgery, Haikou People's Hospital, Haikou, Hainan 570208, P.R. China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013, People's Republic of China.
| | - Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
26
|
Sousa-Batista AJ, Arruda-Costa N, Escrivani DO, Reynaud F, Steel PG, Rossi-Bergmann B. Single-dose treatment for cutaneous leishmaniasis with an easily synthesized chalcone entrapped in polymeric microparticles. Parasitology 2020; 147:1032-1037. [PMID: 32364107 PMCID: PMC10317656 DOI: 10.1017/s0031182020000712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/21/2020] [Accepted: 04/25/2020] [Indexed: 11/07/2022]
Abstract
Cutaneous leishmaniasis (CL) is a major health problem in many countries and its current treatment involves multiple parenteral injections with toxic drugs and requires intensive health services. Previously, the efficacy of a single subcutaneous injection with a slow-release formulation consisting of poly(lactide-co-glycolide) (PLGA) microparticles loaded with an antileishmanial 3-nitro-2-hydroxy-4,6-dimethoxychalcone (CH8) was demonstrated in mice model. In the search for more easily synthesized active chalcone derivatives, and improved microparticle loading, CH8 analogues were synthesized and tested for antileishmanial activity in vitro and in vivo. The 3-nitro-2',4',6'-trimethoxychalcone (NAT22) analogue was chosen for its higher selectivity against intracellular amastigotes (selectivity index = 1489, as compared with 317 for CH8) and more efficient synthesis (89% yield, as compared with 18% for CH8). NAT22 was loaded into PLGA / polyvinylpyrrolidone (PVP) polymeric blend microspheres (NAT22-PLGAk) with average diameter of 1.9 μm. Although NAT22-PLGAk showed similar activity to free NAT22 in killing intracellular parasites in vitro (IC50 ~ 0.2 μm), in vivo studies in Leishmania amazonensis - infected mice demonstrated the significant superior efficacy of NAT22-PLGAk to reduce the parasite load. A single intralesional injection with NAT22-PLGAk was more effective than eight injections with free NAT22. Together, these results show that NAT22-PLGAk is a promising alternative for single-dose localized treatment of CL.
Collapse
Affiliation(s)
- Ariane J. Sousa-Batista
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering – COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia Arruda-Costa
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas O. Escrivani
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Franceline Reynaud
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Bartira Rossi-Bergmann
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Prediction of the enhanced insulin absorption across a triple co-cultured intestinal model using mucus penetrating PLGA nanoparticles. Int J Pharm 2020; 585:119516. [DOI: 10.1016/j.ijpharm.2020.119516] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/25/2020] [Accepted: 06/05/2020] [Indexed: 01/26/2023]
|
28
|
Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic Nanoparticles and Their Targeted Delivery Applications. Molecules 2020; 25:E2193. [PMID: 32397080 PMCID: PMC7248934 DOI: 10.3390/molecules25092193] [Citation(s) in RCA: 339] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Nanotechnology offers many advantages in various fields of science. In this regard, nanoparticles are the essential building blocks of nanotechnology. Recent advances in nanotechnology have proven that nanoparticles acquire a great potential in medical applications. Formation of stable interactions with ligands, variability in size and shape, high carrier capacity, and convenience of binding of both hydrophilic and hydrophobic substances make nanoparticles favorable platforms for the target-specific and controlled delivery of micro- and macromolecules in disease therapy. Nanoparticles combined with the therapeutic agents overcome problems associated with conventional therapy; however, some issues like side effects and toxicity are still debated and should be well concerned before their utilization in biological systems. It is therefore important to understand the specific properties of therapeutic nanoparticles and their delivery strategies. Here, we provide an overview on the unique features of nanoparticles in the biological systems. We emphasize on the type of clinically used nanoparticles and their specificity for therapeutic applications, as well as on their current delivery strategies for specific diseases such as cancer, infectious, autoimmune, cardiovascular, neurodegenerative, ocular, and pulmonary diseases. Understanding of the characteristics of nanoparticles and their interactions with the biological environment will enable us to establish novel strategies for the treatment, prevention, and diagnosis in many diseases, particularly untreatable ones.
Collapse
Affiliation(s)
- Abuzer Alp Yetisgin
- Materials Science and Nano-Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey;
| | - Merve Zuvin
- Mechatronics Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; (M.Z.); (A.K.)
| | - Ali Kosar
- Mechatronics Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; (M.Z.); (A.K.)
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| | - Ozlem Kutlu
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
29
|
Bedair TM, Lee CK, Kim DS, Baek SW, Bedair HM, Joshi HP, Choi UY, Park KH, Park W, Han I, Han DK. Magnesium hydroxide-incorporated PLGA composite attenuates inflammation and promotes BMP2-induced bone formation in spinal fusion. J Tissue Eng 2020; 11:2041731420967591. [PMID: 33178410 PMCID: PMC7592173 DOI: 10.1177/2041731420967591] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
Spinal fusion has become a common surgical technique to join two or more vertebrae to stabilize a damaged spine; however, the rate of pseudarthrosis (failure of fusion) is still high. To minimize pseudarthrosis, bone morphogenetic protein-2 (BMP2) has been approved for use in humans. In this study, we developed a poly(lactide-co-glycolide) (PLGA) composite incorporated with magnesium hydroxide (MH) nanoparticles for the delivery of BMP2. This study aimed to evaluate the effects of released BMP2 from BMP2-immobilized PLGA/MH composite scaffold in an in vitro test and an in vivo mice spinal fusion model. The PLGA/MH composite films were fabricated via solvent casting technique. The surface of the PLGA/MH composite scaffold was modified with polydopamine (PDA) to effectively immobilize BMP2 on the PLGA/MH composite scaffold. Analyzes of the scaffold revealed that using PLGA/MH-PDA improved hydrophilicity, degradation performance, neutralization effects, and increased BMP2 loading efficiency. In addition, releasing BMP2 from the PLGA/MH scaffold significantly promoted the proliferation and osteogenic differentiation of MC3T3-E1 cells. Furthermore, the pH neutralization effect significantly increased in MC3T3-E1 cells cultured on the BMP2-immobilized PLGA/MH scaffold. In our animal study, the PLGA/MH scaffold as a BMP2 carrier attenuates inflammatory responses and promotes BMP2-induced bone formation in posterolateral spinal fusion model. These results collectively demonstrate that the BMP2-immobilized PLGA/MH scaffold offers great potential in effectively inducing bone formation in spinal fusion surgery.
Collapse
Affiliation(s)
- Tarek M. Bedair
- Department of Biomedical Science, CHA
University, Seongnam-si, Gyeonggi-do, Republic of Korea
- Chemistry Department, Faculty of
Science, Minia University, El-Minia, Egypt
| | - Chang Kyu Lee
- Department of Neurosurgery, Keimyung
University Dongsan Medical Center, Daegu, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA
University, Seongnam-si, Gyeonggi-do, Republic of Korea
- School of Integrative Engineering,
Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA
University, Seongnam-si, Gyeonggi-do, Republic of Korea
- Department of Biomedical Engineering,
Sungkyunkwan University, Jangan-gu, Gyeonggi-do, Republic of Korea
| | - Hanan M. Bedair
- Department of Clinical Pathology,
National Liver Institute, Menoufia University, Menoufia, Egypt
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA
University School of Medicine, CHA Bungdang Medical Center, Seongnam-si,
Gyeonggi-do, Republic of Korea
| | - Un Yong Choi
- Department of Neurosurgery, CHA
University School of Medicine, CHA Bungdang Medical Center, Seongnam-si,
Gyeonggi-do, Republic of Korea
| | - Keun-Hong Park
- Department of Biomedical Science, CHA
University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Wooram Park
- Department of Biomedical-Chemical
Engineering, The Catholic University of Korea, Bucheon-Si, Gyeonggi-do, Republic of
Korea
| | - InBo Han
- Department of Neurosurgery, CHA
University School of Medicine, CHA Bungdang Medical Center, Seongnam-si,
Gyeonggi-do, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA
University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
30
|
Banstola A, Pham TT, Jeong JH, Yook S. Polydopamine-tailored paclitaxel-loaded polymeric microspheres with adhered NIR-controllable gold nanoparticles for chemo-phototherapy of pancreatic cancer. Drug Deliv 2019; 26:629-640. [PMID: 31237149 PMCID: PMC6598510 DOI: 10.1080/10717544.2019.1628118] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
Chemotherapeutic drugs often used as a first-line treatment of pancreatic cancer (PC) exhibit challenges due to resistance development, lack of selectivity, and tumor heterogeneity. Currently, combination chemo-photothermal therapy is known to enhance the therapeutic efficacy of chemotherapeutic drugs in PC. In this study, we develop adherent gold nanoparticles (GNPs) and paclitaxel (PTX)-loaded PLGA microspheres for the treatment of PC. Polydopamine (pD) was used as a linker to adhere GNPs to the surface of PLGA-Ms and characterized using TEM. Short-term cytotoxicity of GNPs-pD-PTX-PLGA-Ms with or without NIR treatment was evaluated using CCK-8 assays. ROS and western blot assay were performed to determine the intensity of ROS following the treatment of GNPs-pD-PTX-PLGA-Ms with or without NIR in Panc-1 cell line. Successful adhesion of GNPs on the microspheres was confirmed by TEM. CCK-8 assay revealed that GNPs-pD-PTX-PLGA-Ms with NIR showed three-fold higher cytotoxicity, compared to the group without NIR. Furthermore, ROS and western blot assay suggest that GNPs-pD-PTX-PLGA-Ms with NIR showed more ROS generation, followed by downregulation of the expression levels of antioxidant enzyme (SOD2 and CATALASE). These results suggest that the GNPs-pD-PTX-PLGA-Ms in combination with NIR irradiation can provide a synergistic chemo-photothermal therapy for the treatment of PC.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Tung Thanh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, South Korea
| |
Collapse
|
31
|
Filippi M, Born G, Felder-Flesch D, Scherberich A. Use of nanoparticles in skeletal tissue regeneration and engineering. Histol Histopathol 2019; 35:331-350. [PMID: 31721139 DOI: 10.14670/hh-18-184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bone and osteochondral defects represent one of the major causes of disabilities in the world. Derived from traumas and degenerative pathologies, these lesions cause severe pain, joint deformity, and loss of joint motion. The standard treatments in clinical practice present several limitations. By producing functional substitutes for damaged tissues, tissue engineering has emerged as an alternative in the treatment of defects in the skeletal system. Despite promising preliminary clinical outcomes, several limitations remain. Nanotechnologies could offer new solutions to overcome those limitations, generating materials more closely mimicking the structures present in naturally occurring systems. Nanostructures comparable in size to those appearing in natural bone and cartilage have thus become relevant in skeletal tissue engineering. In particular, nanoparticles allow for a unique combination of approaches (e.g. cell labelling, scaffold modification or drug and gene delivery) inside single integrated systems for optimized tissue regeneration. In the present review, the main types of nanoparticles and the current strategies for their application to skeletal tissue engineering are described. The collection of studies herein considered confirms that advanced nanomaterials will be determinant in the design of regenerative therapeutic protocols for skeletal lesions in the future.
Collapse
Affiliation(s)
- Miriam Filippi
- Department of Biomedical Engineering, University of Basel, Allschwil, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Gordian Born
- Department of Biomedical Engineering, University of Basel, Allschwil, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Delphine Felder-Flesch
- Institut de Physique et Chimie des Matériaux Strasbourg, UMR CNRS-Université de Strasbourg, Strasbourg, France
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Allschwil, Basel, Switzerland.
| |
Collapse
|
32
|
Pranskunas M, Galindo-Moreno P, Padial-Molina M. Extraction Socket Preservation Using Growth Factors and Stem Cells: a Systematic Review. EJOURNAL OF ORAL MAXILLOFACIAL RESEARCH 2019; 10:e7. [PMID: 31620269 PMCID: PMC6788421 DOI: 10.5037/jomr.2019.10307] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Objectives To evaluate the reported literature on the use of stem cells or growth factors for post extraction treatment of the alveolar bone. Material and Methods A NCBI PubMed and PubMed Central databases search was conducted between September 2010 and August 2018, to identify animal or clinical studies reporting the clinical, radiographical and/or histological outcomes of socket preservation techniques after applying mesenchymal stem cells or growth factors. Only studies published in English language in the last 10 years were included in the study. Results Eleven studies were identified fulfilling the inclusion criteria. They evaluate a total of 386 post extraction sockets. The main tested materials identified in the current review were bone morphogenetic protein-2 - 3 studies and mesenchymal stem cells - 3 studies. Other comparators were bone morphogenetic protein-9, platelet-derived growth factor-BB homodimers and bone marrow. Overall evaluation indicate positive results for all test groups showing differences in final socket width between 0.64 and 1.28 mm favouring the test groups. Histologically, no particular differences are detected between test and control groups. Most of the studies present low risk of bias. Conclusions In general, the use of mesenchymal stem cells or bioactive osteogenic molecules favours bone regeneration after tooth extraction, as evaluated clinically, radiographically and histologically. However, specific differences that support particular recommendations are still unclear in light of the current published evidence. Future studies should include the standardization of the mesenchymal stem cells selection and purification as well as dosage and delivery methods of bioactive molecules.
Collapse
Affiliation(s)
- Mindaugas Pranskunas
- Department of Oral and Maxillofacial Surgery, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, KaunasLithuania
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry. School of Dentistry, University of Granada, GranadaSpain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry. School of Dentistry, University of Granada, GranadaSpain
| |
Collapse
|
33
|
Formulation, Colloidal Characterization, and In Vitro Biological Effect of BMP-2 Loaded PLGA Nanoparticles for Bone Regeneration. Pharmaceutics 2019; 11:pharmaceutics11080388. [PMID: 31382552 PMCID: PMC6723283 DOI: 10.3390/pharmaceutics11080388] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/15/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) based on the polymer poly (lactide-co-glycolide) acid (PLGA) have been widely studied in developing delivery systems for drugs and therapeutic biomolecules, due to the biocompatible and biodegradable properties of the PLGA. In this work, a synthesis method for bone morphogenetic protein (BMP-2)-loaded PLGA NPs was developed and optimized, in order to carry out and control the release of BMP-2, based on the double-emulsion (water/oil/water, W/O/W) solvent evaporation technique. The polymeric surfactant Pluronic F68 was used in the synthesis procedure, as it is known to have an effect on the reduction of the size of the NPs, the enhancement of their stability, and the protection of the encapsulated biomolecule. Spherical solid polymeric NPs were synthesized, showing a reproducible multimodal size distribution, with diameters between 100 and 500 nm. This size range appears to allow the protein to act on the cell surface and at the cytoplasm level. The effect of carrying BMP-2 co-adsorbed with bovine serum albumin on the NP surface was analyzed. The colloidal properties of these systems (morphology by SEM, hydrodynamic size, electrophoretic mobility, temporal stability, protein encapsulation, and short-term release profile) were studied. The effect of both BMP2-loaded NPs on the proliferation, migration, and osteogenic differentiation of mesenchymal stromal cells from human alveolar bone (ABSC) was also analyzed in vitro.
Collapse
|
34
|
Fabrication of microcomposites based on silk sericin and monetite for bone tissue engineering. Polym Bull (Berl) 2019. [DOI: 10.1007/s00289-019-02754-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
35
|
Flichy-Fernández AJ, Blaya-Tárraga JA, O'Valle F, Padial-Molina M, Peñarrocha-Diago M, Galindo-Moreno P. Sinus floor elevation using particulate PLGA-coated biphasic calcium phosphate bone graft substitutes: A prospective histological and radiological study. Clin Implant Dent Relat Res 2019; 21:895-902. [PMID: 30895713 DOI: 10.1111/cid.12741] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/03/2018] [Accepted: 10/23/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Poly (lactic-co-glycolic acid) (PLGA) is widely used for the development of delivery systems for drugs and therapeutic biomolecules in tissue engineering applications. Particles of biphasic calcium phosphate can be covered by PLGA to change their manipulating characteristics. PURPOSE Aim of this study was to investigate the radiological and histomorphometric results of the use of PLGA-coated biphasic calcium phosphate granules in sinus floor elevation and to analyze the underlying molecular processes by immunohistochemical staining. MATERIALS AND METHODS A randomized clinical study was designed to include patients in need of sinus floor elevation. Patients were assigned to receive either PLGA-coated biphasic calcium phosphate particles (group I) or the equivalent but noncoated particles (group II). Cone beam computed tomography (CBCT) scans were performed before and 6 months after the procedure to assess the bone height gain. At the time of implant placement, bone core biopsies were obtained at the site of implant placement. Histological sections were subjected to histomorphometric and immunohistochemical evaluation of differentiation markers (Musashi-1 [MSI1]). RESULTS No statistically significant differences were observed between groups for the radiologic parameters. No differences were observed histologically or histomorphometrically. However, PLGA-coated particles (group I) were more colonized by MSI1-positive osteoblast precursors (P = 0.0001, chi-squared test) and were penetrated by more CD34-positive vascular structures (P = 0.001, chi-squared test) than noncoated particles (group II). CONCLUSIONS PLGA-coated particles are associated with more MSI11-positive cells and more extensive microvascularization than noncoated particles.
Collapse
Affiliation(s)
- Antonio J Flichy-Fernández
- Department of Oral Surgery and Implantology, Valencia University Medical and Dental School, Valencia, Spain
| | - Juan A Blaya-Tárraga
- Department of Oral Surgery and Implantology, Valencia University Medical and Dental School, Valencia, Spain
| | - Francisco O'Valle
- Department of Pathology, School of Medicine & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| | - Miguel Peñarrocha-Diago
- Department of Oral Surgery and Implantology, Valencia University Medical and Dental School, Valencia, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| |
Collapse
|
36
|
Xia L, Ma W, Zhou Y, Gui Z, Yao A, Wang D, Takemura A, Uemura M, Lin K, Xu Y. Stimulatory Effects of Boron Containing Bioactive Glass on Osteogenesis and Angiogenesis of Polycaprolactone: In Vitro Study. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8961409. [PMID: 31011582 PMCID: PMC6442456 DOI: 10.1155/2019/8961409] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/25/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022]
Abstract
Polycaprolactone (PCL) has attracted great attention for bone regeneration attributed to its cost-efficiency, high toughness, and good processability. However, the relatively low elastic modulus, hydrophobic nature, and insufficient bioactivity of pure PCL limited its wider application for bone regeneration. In the present study, the effects of the addition of boron containing bioactive glass (B-BG) materials on the mechanical properties and biological performance of PCL polymer were investigated with different B-BG contents (0, 10, 20, 30, and 40 wt.%), in order to evaluate the potential applications of B-BG/PCL composites for bone regeneration. The results showed that the B-BG/PCL composites possess better tensile strength, human neutral pH value, and fast degradation as compared to pure PCL polymers. Moreover, the incorporation of B-BG could enhance proliferation, osteogenic differentiation, and angiogenic factor expression for rat bone marrow stromal cells (rBMSCs) as compared to pure PCL polymers. Importantly, the B-BG also promoted the angiogenic differentiation for human umbilical vein endothelial cells (HUVECs). These enhanced effects had a concentration dependence of B-BG content, while 30 wt.% B-BG/PCL composites achieved the greatest stimulatory effect. Therefore the 30 wt.% B-BG/PCL composites have potential applications in bone reconstruction fields.
Collapse
Affiliation(s)
- Lunguo Xia
- Department of Orthodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| | - Wudi Ma
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| | - Yuning Zhou
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| | - Zhipeng Gui
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| | - Aihua Yao
- Key Laboratory of the Advanced Civil Engineering Materials, Tongji University, Ministration of Education, China
| | - Deping Wang
- Key Laboratory of the Advanced Civil Engineering Materials, Tongji University, Ministration of Education, China
| | | | - Mamoru Uemura
- Department of Anatomy, Osaka Dental University, Osaka, Japan
| | - Kailin Lin
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| | - Yuanjin Xu
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, China
| |
Collapse
|
37
|
Lin Z, Tang Y, Tan H, Cai D. MicroRNA-92a-1-5p influences osteogenic differentiation of MC3T3-E1 cells by regulating β-catenin. J Bone Miner Metab 2019; 37:264-272. [PMID: 30019248 DOI: 10.1007/s00774-018-0935-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/25/2018] [Indexed: 12/27/2022]
Abstract
Osteoblastic differentiation is a complex process that is critical for proper bone formation. An increasing number of studies have suggested that microRNAs (miRNAs) are pivotal regulators in various physiological and pathological processes, including osteogenesis. Here, we discuss the influence of miRNA-92a-1-5p on osteogenic differentiation. We found that miR-92a-1-5p was obviously downregulated during osteogenic differentiation of MC3T3-E1 cells. Gain-of-function and loss-of-function experiments revealed that miR-92a-1-5p was a negative regulator of osteogenic differentiation. Experimental validation demonstrated that β-catenin, which acts as a positive regulator of osteogenic differentiation, was negatively regulated by miR-92a1-5p. The findings of this study provide new insights into the possibility of miR-92a1-5p being a potential therapeutic target in the management of bone regeneration-related diseases.
Collapse
Affiliation(s)
- Zhiping Lin
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, number 183, Zhong shan Road West, Guangzhou, 510630, Guangdong, People's Republic of China
- Department of Orthopedics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People's Republic of China
| | - Yangyang Tang
- Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Hongchang Tan
- Department of Orthopedics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People's Republic of China
| | - Daozhang Cai
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, number 183, Zhong shan Road West, Guangzhou, 510630, Guangdong, People's Republic of China.
| |
Collapse
|
38
|
Patelli A, Mussano F, Brun P, Genova T, Ambrosi E, Michieli N, Mattei G, Scopece P, Moroni L. Nanoroughness, Surface Chemistry, and Drug Delivery Control by Atmospheric Plasma Jet on Implantable Devices. ACS APPLIED MATERIALS & INTERFACES 2018; 10:39512-39523. [PMID: 30359523 DOI: 10.1021/acsami.8b15886] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Implantable devices need specific tailored surface morphologies and chemistries to interact with the living systems or to actively induce a biological response also by the release of drugs or proteins. These customized requirements foster technologies that can be implemented in additive manufacturing systems. Here, we present a novel approach based on spraying processes that allow to control separately topographic features in the submicron range (∼60 nm to 2 μm), ammine or carboxylic chemistry, and fluorophore release even on temperature-sensitive biodegradable polymers such as polycaprolactone (PCL). We developed a two-steps process with a first deposition of 220 nm silica and poly(lactic- co-glycolide) (PLGA) fluorescent nanoparticles by aerosol followed by the deposition of a fixing layer by an atmospheric pressure plasma jet (APPJ). The nanoparticles can be used to create the nanoroughness and to include active molecule release, while the capping layer ensures stability and the chemical functionalities. The process is enabled by a novel APPJ which allows deposition rates of 10-20 nm·s-1 at temperatures lower than 50 °C using argon as the process gas. This approach was assessed on titanium alloys for dental implants and on PCL films. The surfaces were characterized by Fourier transform infrared, atomic force microscopy, and scanning electron microscopy (SEM). Titanium alloys were tested with the preosteoblast murine cells line, while the PCL film was tested with fibroblasts. Cell behavior was evaluated by viability and adhesion assays, protein adsorption, cell proliferation, focal adhesion formation, and SEM. The release of a fluorophore molecule was assessed in the cell growing media, simulating a drug release. Osteoblast adhesion on the plasma-treated materials increased by 20% with respect to commercial titanium alloy implants. Fibroblast adhesion increased by a 100% compared to smooth PCL substrates. The release of the fluorophore by the dissolution of the PLGA nanoparticles was verified, and the integrity of the encapsulated drug model was confirmed.
Collapse
Affiliation(s)
- Alessandro Patelli
- Department Physics and Astronomy , Padova University , via Marzolo 8 , 35131 Padova , Italy
| | - Federico Mussano
- CIR Dental School, Department Surgical Sciences , Torino University , 10126 Torino , Italy
| | - Paola Brun
- Department Molecular Medicine, Unit of Microbiology , Padova University , 35121 Padova , Italy
| | - Tullio Genova
- CIR Dental School, Department Surgical Sciences , Torino University , 10126 Torino , Italy
- Department Life Sciences and Systems Biology , Torino University , 10124 Torino , Italy
| | - Emmanuele Ambrosi
- Department Molecular Sciences and Nanosystems , Venezia University , 30172 Venezia , Italy
| | - Niccoló Michieli
- Department Physics and Astronomy , Padova University , via Marzolo 8 , 35131 Padova , Italy
| | - Giovanni Mattei
- Department Physics and Astronomy , Padova University , via Marzolo 8 , 35131 Padova , Italy
| | | | - Lorenzo Moroni
- MERLN-Institute for Technology-Inspired Regenerative Medicine , Maastricht University , 6229 ER Maastricht , The Netherlands
| |
Collapse
|
39
|
Abstract
Present bioprinting techniques lack the methodology to print with bioactive materials that retain their biological functionalities. This constraint is due to the fact that extrusion-based printing of synthetic polymers is commonly performed at very high temperatures in order to achieve desired mechanical properties and printing resolutions. Consequently, current methodology prevents printing scaffolds embedded with bioactive molecules, such as growth factors. With the wide use of mesenchymal stem cells (MSCs) in regenerative medicine research, the integration of growth factors into 3D printed scaffolds is critical because it can allow for inducible MSC differentiation. We have successfully incorporated growth factors into extrusion printed poly (lactic-co-glycolic acid) (PLGA) scaffolds by introducing dimethyl sulfoxide (DMSO) for low temperature printing. Mechanical testing results demonstrated significantly different compressive and tensile properties for PLGA scaffold printed with or without DMSO. In particular, the PLGA-DMSO scaffold displayed a highly stretchable feature compared to the regular PLGA scaffold. The cellular response of growth factor introduction was evaluated in vitro using human mesenchymal stem cells (hMSCs). By evaporating the DMSO after printing, we ensured that there was no cytotoxic effect on seeded hMSCs. The addition of lineage specific growth factors led to increased expression of corresponding genetic markers for chondrogenesis, osteogenesis, and adipogenesis. We concluded that the use of DMSO for 3D printed scaffold fabrication with bioactive items is a revolutionary methodology in advancing regenerative medicine. The incorporation of bioactive molecules opens pathways to more therapeutic uses for 3D printing in treating damaged or deteriorating native tissue.
Collapse
|
40
|
Interaction of surfactant and protein at the O/W interface and its effect on colloidal and biological properties of polymeric nanocarriers. Colloids Surf B Biointerfaces 2018; 173:295-302. [PMID: 30308454 DOI: 10.1016/j.colsurfb.2018.09.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022]
Abstract
HYPOTHESIS The use of polymer-based surfactants in the double-emulsion (water/oil/water, W/O/W) solvent-evaporation technique is becoming a widespread strategy for preparing biocompatible and biodegradable polymeric nanoparticles (NPs) loaded with biomolecules of interest in biomedicine, or biotechnology. This approach enhances the stability of the NPs, reduces their size and recognition by the mononuclear phagocytic system, and protects the encapsulated biomolecule against losing biological activity. Different protocols to add the surfactant during the synthesis lead to different NP colloidal properties and biological activity. EXPERIMENTS We develop an in vitro model to mimic the first step of the W/O/W NP synthesis method, which enables us to analyze the surfactant-biomolecule interaction at the O/W interface. We compare the interfacial properties when the surfactant is added from the aqueous or the organic phase, and the effect of pH of the biomolecule solution. We work with a widely used biocompatible surfactant (Pluronic F68), and lysozyme, reported as a protein model. FINDINGS The surfactant, when added from the water phase, displaces the protein from the interface, hence protecting the biomolecule. This could explain the improved colloidal stability of NPs, and the higher biological activity of the lysozyme released from nanoparticles found with the counterpart preparation.
Collapse
|
41
|
Wang X, Wang G, Zingales S, Zhao B. Biomaterials Enabled Cell-Free Strategies for Endogenous Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:463-481. [PMID: 29897021 DOI: 10.1089/ten.teb.2018.0012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repairing bone defects poses a major orthopedic challenge because current treatments are constrained by the limited regenerative capacity of human bone tissue. Novel therapeutic strategies, such as stem cell therapy and tissue engineering, have the potential to enhance bone healing and regeneration, and hence may improve quality of life for millions of people. However, the ex vivo expansion of stem cells and their in vivo delivery pose technical difficulties that hamper clinical translation and commercial development. A promising alternative to cell delivery-based strategies is to stimulate or augment the inherent self-repair mechanisms of the patient to promote endogenous restoration of the lost/damaged bone. There is growing evidence indicating that increasing the endogenous regenerative potency of bone tissues for therapeutics will require the design and development of new generations of biomedical devices that provide key signaling molecules to instruct cell recruitment and manipulate cell fate for in situ tissue regeneration. Currently, a broad range of biomaterial-based deployment technologies are becoming available, which allow for controlled spatial presentation of biological cues required for endogenous bone regeneration. This article aims to explore the proposed concepts and biomaterial-enabled strategies involved in the design of cell-free endogenous techniques in bone regenerative medicine.
Collapse
Affiliation(s)
- Xiaojing Wang
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| | - Guowei Wang
- 3 Department of Stomatology, Laoshan Branch of No. 401 Hospital of the Chinese Navy , Qingdao, Shandong, P.R. China
| | - Sarah Zingales
- 4 Department of Chemistry and Biochemistry, Georgia Southern University , Savannah, Georgia
| | - Baodong Zhao
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
42
|
Martins C, Sousa F, Araújo F, Sarmento B. Functionalizing PLGA and PLGA Derivatives for Drug Delivery and Tissue Regeneration Applications. Adv Healthc Mater 2018; 7. [PMID: 29171928 DOI: 10.1002/adhm.201701035] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/27/2017] [Indexed: 12/16/2022]
Abstract
Poly(lactic-co-glycolic) acid (PLGA) is one of the most versatile biomedical polymers, already approved by regulatory authorities to be used in human research and clinics. Due to its valuable characteristics, PLGA can be tailored to acquire desirable features for control bioactive payload or scaffold matrix. Moreover, its chemical modification with other polymers or bioconjugation with molecules may render PLGA with functional properties that make it the Holy Grail among the synthetic polymers to be applied in the biomedical field. In this review, the physical-chemical properties of PLGA, its synthesis, degradation, and conjugation with other polymers or molecules are revised in detail, as well as its applications in drug delivery and regeneration fields. A particular focus is given to successful examples of products already on the market or at the late stages of trials, reinforcing the potential of this polymer in the biomedical field.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Flávia Sousa
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- ICBAS - Instituto Ciências Biomédicas Abel Salazar; Universidade do Porto; Rua de Jorge Viterbo Ferreira 228 4050-313 Porto Portugal
| | - Francisca Araújo
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde; Rua Central de Gandra 1317 4585-116 Gandra Portugal
| |
Collapse
|
43
|
Vora LK, Donnelly RF, Larrañeta E, González-Vázquez P, Thakur RRS, Vavia PR. Novel bilayer dissolving microneedle arrays with concentrated PLGA nano-microparticles for targeted intradermal delivery: Proof of concept. J Control Release 2017; 265:93-101. [PMID: 29037785 DOI: 10.1016/j.jconrel.2017.10.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/25/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022]
Abstract
Polymeric microneedle (MN) arrays continue to receive growing attention due to their ability to bypass the skin's stratum corneum barrier in a minimally-invasive fashion and achieve enhanced transdermal drug delivery and "targeted" intradermal vaccine administration. In this research work, we fabricated biodegradable bilayer MN arrays containing nano - microparticles for targeted and sustained intradermal drug delivery. For this study, model drug (vitamin D3, VD3)-loaded PLGA nano- and microparticles (NMP) were prepared by a single emulsion solvent evaporation method with 72.8% encapsulation of VD3. The prepared NMP were directly mixed 20% w/v poly(vinyl pyrrolidone) (PVP) gel, with the mixture filled into laser engineered micromoulds by high-speed centrifugation (30min) to concentrate NMP into MN shafts. The particle size of PLGA NMP ranged from 300nm to 3.5μm and they retained their particle size after moulding of bilayer MN arrays. The relatively wide particle size distribution of PLGA NMP was shown to be important in producing a compact structure in bilayer conical, as well as pyramidal, MN, as confirmed by scanning electron microscopy. The drug release profile from PLGA NMP was tri-phasic, being sustained over 5days. The height of bilayer MN arrays was influenced by the weight ratio of NMP and 20% w/v PVP. Good mechanical and insertion profiles (into a skin simulant and excised neonatal porcine skin) were confirmed by texture analysis and optical coherence tomography, respectively. Ex vivo intradermal neonatal porcine skin penetration of VD3 NMP from bilayer MN was quantitatively analysed after cryostatic skin sectioning, with 74.2±9.18% of VD3 loading delivered intradermally. The two-stage novel processing strategy developed here provides a simple and easy method for localising particulate delivery systems into dissolving MN. Such systems may serve as promising means for controlled transdermal delivery and targeted intradermal administration.
Collapse
Affiliation(s)
- Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, University Under Section 3 of UGC Act - 1956, Elite Status and Center of Excellence, Govt. of Maharashtra, Mumbai 400 019, India
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Eneko Larrañeta
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | | | | | - Pradeep R Vavia
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, University Under Section 3 of UGC Act - 1956, Elite Status and Center of Excellence, Govt. of Maharashtra, Mumbai 400 019, India.
| |
Collapse
|
44
|
Wang W, Shahzad KA, Li M, Zhang A, Zhang L, Xu T, Wan X, Shen C. An Antigen-Presenting and Apoptosis-Inducing Polymer Microparticle Prolongs Alloskin Graft Survival by Selectively and Markedly Depleting Alloreactive CD8 + T Cells. Front Immunol 2017. [PMID: 28649247 PMCID: PMC5465244 DOI: 10.3389/fimmu.2017.00657] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Selectively depleting the pathogenic T cells is a fundamental strategy for the treatment of allograft rejection and autoimmune disease since it retains the overall immune function of host. The concept of killer artificial antigen-presenting cells (KaAPCs) has been developed by co-coupling peptide–major histocompatibility complex (pMHC) multimer and anti-Fas monoclonal antibody (mAb) onto the polymeric microparticles (MPs) to induce the apoptosis of antigen-specific T cells. But little information is available about its in vivo therapeutic potential and mechanism. In this study, polyethylenimine (PEI)-coated poly lactic-co-glycolic acid microparticle (PLGA MP) was fabricated as a cell-sized scaffold to covalently co-couple H-2Kb-Ig dimer and anti-Fas mAb for the generation of alloantigen-presenting and apoptosis-inducing MPs. Intravenous infusions of the biodegradable KaAPCs prolonged the alloskin graft survival for 43 days in a single MHC-mismatched murine model, depleted the most of H-2Kb-alloreactive CD8+ T cells in peripheral blood, spleen, and alloskin graft in an antigen-specific manner and anti-Fas-dependent fashion. The cell-sized KaAPCs circulated throughout vasculature into liver, kidney, spleen, lymph nodes, lung, and heart, but few ones into local allograft at early stage, with a retention time up to 36 h in vivo. They colocalized with CD8+ T cells in secondary lymphoid organs while few ones contacted with CD4+ T cells, B cells, macrophage, and dendritic cells, or internalized by phagocytes. Importantly, the KaAPC treatment did not significantly impair the native T cell repertoire or non-pathogenic immune cells, did not obviously suppress the overall immune function of host, and did not lead to visible organ toxicity. Our results strongly document the high potential of PLGA MP-based KaAPCs as a novel antigen-specific immunotherapy for allograft rejection and autoimmune disorder. The in vivo mechanism of alloinhibition, tissue distribution, and biosafety were also initially characterized, which will facilitate its translational studies from bench to bedside.
Collapse
Affiliation(s)
- Wei Wang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Miaochen Li
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Aifeng Zhang
- Department of Pathology, Southeast University Medical School, Nanjing, China
| | - Lei Zhang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Tao Xu
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Xin Wan
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, China
| |
Collapse
|
45
|
Yao Q, Sandhurst ES, Liu Y, Sun H. BBP-Functionalized Biomimetic Nanofibrous Scaffold Can Capture BMP2 and Promote Osteogenic Differentiation. J Mater Chem B 2017; 5:5196-5205. [PMID: 29250330 DOI: 10.1039/c7tb00744b] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bone morphogenetic proteins (BMPs, e.g., BMP2 and 7) are potent mediators for bone repair, however, their clinical use has been limited by their safety and cost-effectiveness. Therefore, innovative strategies that can improve the efficacy of BMPs, and thereby, use a lower dose of exogenous BMPs are highly desired. Inspired by the natural interaction between extracellular matrix (ECM) and growth factors, we hypothesize that bone matrix-mimicking nanofibrous scaffold functionalized with BMP binding moieties can selectively capture and stabilize BMPs, and thereby, promote BMP-induced osteogenic differentiation. To test our hypothesis, a gelatin nanofibrous scaffold was fabricated using thermally induced phase separation together with a porogen leaching technique (TIPS&P) and functionalized by a BMP-binding peptide (BBP) through cross-linking. Our data indicated that BBP decoration largely improved the BMP2 binding and retention capacity of the nanofibrous scaffolds without compromising their macro/microstructure and mechanical properties. Importantly, the BBP-functionalized gelatin scaffolds were able to significantly promote BMP2-induced osteogenic differentiation. Moreover, BBP alone was able to significantly stimulate endogenous BMP2 expression and improve osteogenic differentiation. Compared to other affinity-based drug delivery strategies, e.g., heparin and antibody-mediated growth factor delivering techniques, we expect BBP-functionalized scaffolds will be a safer, more feasible and selective strategy for endogenous BMP stimulating and binding. Therefore, our data suggests a promising application of using the BBP-decorated gelatin nanofibrous scaffold to stimulate/capture BMPs and promote endogenous bone formation in situ in contrast to relying on the administration of high doses of exogenous BMPs and transplantation of cells.
Collapse
Affiliation(s)
- Qingqing Yao
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA.,School of Ophthalmology and Optometry, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Eric S Sandhurst
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| | - Yangxi Liu
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| | - Hongli Sun
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| |
Collapse
|
46
|
van Rijt S, Habibovic P. Enhancing regenerative approaches with nanoparticles. J R Soc Interface 2017; 14:20170093. [PMID: 28404870 PMCID: PMC5414913 DOI: 10.1098/rsif.2017.0093] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022] Open
Abstract
In this review, we discuss recent developments in the field of nanoparticles and their use in tissue regeneration approaches. Owing to their unique chemical properties and flexibility in design, nanoparticles can be used as drug delivery systems, to create novel features within materials or as bioimaging agents, or indeed these properties can be combined to create smart multifunctional structures. This review aims to provide an overview of this research field where the focus will be on nanoparticle-based strategies to stimulate bone regeneration; however, the same principles can be applied for other tissue and organ regeneration strategies. In the first section, nanoparticle-based methods for the delivery of drugs, growth factors and genetic material to promote tissue regeneration are discussed. The second section deals with the addition of nanoparticles to materials to create nanocomposites. Such materials can improve several material properties, including mechanical stability, biocompatibility and biological activity. The third section will deal with the emergence of a relatively new field of research using nanoparticles in advanced cell imaging and stem cell tracking approaches. As the development of nanoparticles continues, incorporation of this technology in the field of regenerative medicine will ultimately lead to new tools that can diagnose, track and stimulate the growth of new tissues and organs.
Collapse
Affiliation(s)
- Sabine van Rijt
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, The Netherlands
| | - Pamela Habibovic
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, The Netherlands
| |
Collapse
|
47
|
Zhong C, Feng J, Lin X, Bao Q. Continuous release of bone morphogenetic protein-2 through nano-graphene oxide-based delivery influences the activation of the NF-κB signal transduction pathway. Int J Nanomedicine 2017; 12:1215-1226. [PMID: 28243085 PMCID: PMC5315217 DOI: 10.2147/ijn.s124040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Graphene oxide (GO) has been used as a delivery vehicle for small molecule drugs and nucleotides. To further investigate GO as a smart biomaterial for the controlled release of cargo molecules, we hypothesized that GO may be an appropriate delivery vehicle because it releases bone morphogenetic protein 2 (BMP2). GO characterization indicated that the size distribution of the GO flakes ranged from 81.1 nm to 45,749.7 nm, with an approximate thickness of 2 nm. After BMP2 adsorption onto GO, Fourier-transformed infrared spectroscopy (FTIR) and thermal gravimetric analysis were performed. Compared to GO, BMP2-GO did not induce significant changes in the characteristics of the materials. GO continuously released BMP2 for at least 40 days. Bone marrow stem cells (BMSCs) and chondrocytes were treated with BMP2-GO in interleukin-1 media and assessed in terms of cell viability, flow cytometric characterization, and expression of particular mRNA. Compared to GO, BMP2-GO did not induce any significant changes in biocompatibility. We treated osteoarthritic rats with BMP2 and BMP2-GO, which showed significant differences in Osteoarthritis Research Society International (OARSI) scores (P<0.05). Quantitative assessment revealed significant differences compared to that using BMP2 and BMP2-GO (P<0.05). These findings indicate that GO may be potentially used to control the release of carrier materials. The combination of BMP2 and GO slowed the progression of NF-κB-activated degenerative changes in osteoarthritis. Therefore, we infer that our BMP2-GO strategy could alleviate the NF-κB pathway by inducing continuous BMP2 release.
Collapse
Affiliation(s)
- Cheng Zhong
- Department of Orthopaedic, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jun Feng
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA
| | - Xiangjin Lin
- Department of Orthopaedic, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qi Bao
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital, School of Medicine; Institute of Gastroenterology, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
48
|
Crecente-Campo J, Borrajo E, Vidal A, Garcia-Fuentes M. New scaffolds encapsulating TGF-β3/BMP-7 combinations driving strong chondrogenic differentiation. Eur J Pharm Biopharm 2017; 114:69-78. [PMID: 28087378 DOI: 10.1016/j.ejpb.2016.12.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 11/28/2022]
Abstract
The regeneration of articular cartilage remains an unresolved question despite the current access to a variety of tissue scaffolds activated with growth factors relevant to this application. Further advances might result from combining more than one of these factors; here, we propose a scaffold composition optimized for the dual delivery of BMP-7 and TGF-β3, two proteins with described chondrogenic activity. First, we tested in a mesenchymal stem cell micromass culture with TGF-β3 whether the exposure to microspheres loaded with BMP-7 would improve cartilage formation. Histology and qRT-PCR data confirmed that the sustained release of BMP-7 cooperates with TGF-β3 towards chondrogenic differentiation. Then, we optimized a scaffold prototype for tissue culture and dual encapsulation of BMP-7 and TGF-β3. The scaffolds were prepared from poly(lactic-co-glycolic acid), and BMP-7/TGF-β3 were loaded as nanocomplexes with heparin and Tetronic 1107. The scaffolds showed the sustained release of both proteins over four weeks, with minimal burst effect. We finally cultured human mesenchymal stem cells on these scaffolds, in the absence of exogenous chondrogenic factor supplementation. The cells cultured on the scaffolds loaded with BMP-7 and TGF-β3 showed clear signs of cartilage formation macroscopically and histologically. RT-PCR studies confirmed a clear upregulation of cartilage markers SOX9 and Aggrecan. In summary, scaffolds encapsulating BMP-7 and TGF-β3 can efficiently deliver a cooperative growth factor combination that drives efficient cartilage formation in human mesenchymal stem cell cultures. These results open attractive perspectives towards in vivo translation of this technology in cartilage regeneration experiments.
Collapse
Affiliation(s)
- Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Erea Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Anxo Vidal
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
49
|
Wei D, Jung J, Yang H, Stout DA, Yang L. Nanotechnology Treatment Options for Osteoporosis and Its Corresponding Consequences. Curr Osteoporos Rep 2016; 14:239-47. [PMID: 27542011 DOI: 10.1007/s11914-016-0324-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Unfortunately, osteoporosis, as a worldwide disease, is challenging human health with treatment only available for the symptoms of osteoporosis without managing the disease itself. Osteoporosis can be linked as the common cause of fractures and increased mortality among post-menopausal women, men, and the elderly. Regrettably, due to osteoporosis, incidents of fractures are more frequent among the presented populations and can be afflictive for carrying out everyday life activities. Current treatments of osteoporosis encompass changing lifestyles, taking orthopedic drugs, and invasive surgeries. However, these treatment options are not long lasting and can lead to complications after post-surgical life. Therefore, to solve this impairment, researchers have turned to nanotechnologies and nanomaterials to create innovative and alternative treatments associated with the consequences of osteoporosis. This review article provides an introduction to osteoporotic compression vertebral fractures (OVCFs) and current clinical treatments, along with the rationale and efficacy of utilizing nanomaterials to modify and improve biomaterials or instruments. The methods of applying bioactive agents (bone morphogenetic protein-2 (BMP-2), parathyroid hormone 1-34 (PTH 1-34)), as well as 3D printing will be presented from an osteoporosis treatment perspective. Additionally, the application of nanoparticles and nanotube arrays onto the current surgical treatments and orthopedic drug administration methods addressed will show that these systems reinforce a better mechanical performance and provide precise and slow-releasing drug delivery for better osseointegration, bone regeneration, and bone strength. In summary, nanomaterials can be seen as an alternative and more effective treatment for individuals with osteoporosis.
Collapse
Affiliation(s)
- Donglei Wei
- Orthopaedic Institute, Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
- International Research Center for Translational Orthopaedics (IRCTO), Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
| | - Jinsuh Jung
- International Research Center for Translational Orthopaedics (IRCTO), Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
- Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Huilin Yang
- Orthopaedic Institute, Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
- International Research Center for Translational Orthopaedics (IRCTO), Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
| | - David A Stout
- International Research Center for Translational Orthopaedics (IRCTO), Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China.
- Department of Mechanical and Aerospace Engineering, California State University, Long Beach, 1250 Bellflower Blvd. ECS-632, Long Beach, CA, 90802, USA.
| | - Lei Yang
- Orthopaedic Institute, Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China.
- International Research Center for Translational Orthopaedics (IRCTO), Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China.
| |
Collapse
|
50
|
Virlan MJR, Miricescu D, Radulescu R, Sabliov CM, Totan A, Calenic B, Greabu M. Organic Nanomaterials and Their Applications in the Treatment of Oral Diseases. Molecules 2016; 21:E207. [PMID: 26867191 PMCID: PMC6273611 DOI: 10.3390/molecules21020207] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/20/2016] [Accepted: 01/28/2016] [Indexed: 12/18/2022] Open
Abstract
There is a growing interest in the development of organic nanomaterials for biomedical applications. An increasing number of studies focus on the uses of nanomaterials with organic structure for regeneration of bone, cartilage, skin or dental tissues. Solid evidence has been found for several advantages of using natural or synthetic organic nanostructures in a wide variety of dental fields, from implantology, endodontics, and periodontics, to regenerative dentistry and wound healing. Most of the research is concentrated on nanoforms of chitosan, silk fibroin, synthetic polymers or their combinations, but new nanocomposites are constantly being developed. The present work reviews in detail current research on organic nanoparticles and their potential applications in the dental field.
Collapse
Affiliation(s)
- Maria Justina Roxana Virlan
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| | - Radu Radulescu
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| | - Cristina M Sabliov
- Agricultural and Biological Engineering Department, Louisiana State University and LSU Ag Center, 149 EB Doran Building, Baton Rouge, LA 70803, USA.
| | - Alexandra Totan
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| | - Bogdan Calenic
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dentistry, University of Medicine and Pharmacy Carol Davila, Blvd. EroiiSanitari, No. 8, RO-050474 Bucharest, Romania.
| |
Collapse
|