1
|
Samari M, Kashanian S, Zinadini S, Derakhshankhah H. Enhanced delivery of azithromycin using asymmetric polyethersulfone membrane modified with KIT-6 mesoporous material: Optimization and mechanistic studies. Eur J Pharm Sci 2025; 207:107038. [PMID: 39933630 DOI: 10.1016/j.ejps.2025.107038] [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: 11/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
This study presents the development of a novel drug delivery system designed for improving the release profile and sustained delivery of azithromycin (AZI), particularly aimed at applications requiring localized infection control and improved tissue compatibility. The system employs an asymmetric polyethersulfone (PES) membrane modified with KIT-6 mesoporous material, offering improved drug release performance and biocompatibility over conventional delivery platforms. Membrane optimization was achieved by systematically varying parameters such as thickness (150-600 µm), drug concentration (500-1500 mg/L), polymer content (13-21 % PES), pore maker percentage (0-4 % polyvinylpyrrolidone), and KIT-6 modifier percentage (0.5-2 %). Characterization included scanning electron microscopy, water contact angle measurements, porosity, tensile strength evaluation, and comprehensive bioactivity testing (cytotoxicity, antimicrobial efficacy, blood compatibility, and a novel tissue integrity assay). The optimized formulation (17 % PES, 2 % PVP, 1 % KIT-6) achieved a controlled and sustained release profile with improved drug availability (464 mg/L) compared to unmodified membranes (252 mg/L), with a sustained release profile governed by the Higuchi model. Additionally, the membrane demonstrated superior biocompatibility (-90 % cell viability, low hemolysis at 1.2 %) and preserved tissue integrity better than unmodified counterparts, as evidenced by in vitro and ex vivo studies. Notably, the system showed robust reusability over prolonged use, indicating its potential as an effective, sustainable, and biocompatible solution for localized AZI delivery. These advantages position this system as a promising alternative for medical applications requiring precise drug release and minimal tissue disruption.
Collapse
Affiliation(s)
- Mahya Samari
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Soheila Kashanian
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Nanobiotechnology Department, Faculty of Innovative Science and Technology, Razi University, Kermanshah, Iran.
| | - Sirus Zinadini
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Environmental Research Center (ERC), Razi University, Kermanshah, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Hourdel L, Lebaz N, Peral F, Ripoll M, Briançon S, Bensaid F, Luthra S, Cogné C. Overview on LNP-mRNA encapsulation unit operation: Mixing technologies, scalability, and influence of formulation & process parameters on physico-chemical characteristics. Int J Pharm 2025; 672:125297. [PMID: 39900125 DOI: 10.1016/j.ijpharm.2025.125297] [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: 10/14/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Abstract
Nanoparticles carrying active drug substances have been used since the 70's and have undergone numerous improvements since then. Nowadays, the latest generation of nanoparticles, called lipid nanoparticles (LNPs), is used for different applications such as vaccines and cancer treatments and offer a versatile approach to delivering genetic materials like RNA. LNPs are non-viral delivery vehicles obtained by the self-assembly of lipids during the rapid mixing of an aqueous phase containing mRNA with an organic phase containing lipids. During this process, mRNA is encapsulated within the LNP due to electrostatic interaction with an ionizable lipid. Different methods to produce LNPs are described in the literature and, as of now, continuous methods are mostly used to produce LNP-encapsulated mRNA (LNP-mRNA). T-shaped mixers are commonly used to produce mRNA-LNPs. This technology can operate at two different scales: microfluidic chips which can range from tens to hundreds of microns in size, and millimetric tubing for production scale up. This review intends to describe LNP-mRNA characteristics and their production modes with a special focus on the challenges related to the mixing quality, especially during scale-up.
Collapse
Affiliation(s)
- Laurine Hourdel
- Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy-l'Etoile, France; Universite Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, 43 boulevard du 11 novembre 1918, F-69100 Villeurbanne, France.
| | - Noureddine Lebaz
- Universite Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, 43 boulevard du 11 novembre 1918, F-69100 Villeurbanne, France
| | - Florent Peral
- Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy-l'Etoile, France
| | - Manon Ripoll
- Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy-l'Etoile, France
| | - Stéphanie Briançon
- Universite Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, 43 boulevard du 11 novembre 1918, F-69100 Villeurbanne, France
| | - Fethi Bensaid
- Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy-l'Etoile, France
| | - Sumit Luthra
- Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy-l'Etoile, France
| | - Claudia Cogné
- Universite Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, 43 boulevard du 11 novembre 1918, F-69100 Villeurbanne, France
| |
Collapse
|
3
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [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: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
4
|
Zhou L, Zhang Y, Yi X, Chen Y, Li Y. Advances in proteins, polysaccharides, and composite biomaterials for enhanced wound healing via microenvironment management: A review. Int J Biol Macromol 2024; 282:136788. [PMID: 39490870 DOI: 10.1016/j.ijbiomac.2024.136788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Wound management is crucial yet imposes substantial social and economic burdens on patients and healthcare systems. The recent rapid advancements in biomaterials and manufacturing technology have created favorable conditions for expediting wound healing. This review examines the latest developments in biomacromolecule-based wound dressings, with a particular focus on proteins and polysaccharides, and their role in modulating the wound microenvironment. The importance of extracellular matrix (ECM)-inspired materials, such as hydrogels and biomimetic dressings, is emphasized. Additionally, this review explores the functionalization of wound dressings, emphasizing properties such as hemostatic capabilities, pain relief, antimicrobial activity, and innovative smart functions like electroceuticals and wound condition monitoring. The study integrates discussions on both the macroscopic healing outcomes and the microscopic pathophysiological mechanisms, highlighting recent advances in managing wound environments to expedite healing. Finally, the review critically assesses the challenges associated with the clinical translation of these wound-healing materials in the future.
Collapse
Affiliation(s)
- Lingyan Zhou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoli Yi
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yining Chen
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Sezer A, Ozalp H, Imge Ucar-Goker B, Gencer A, Ozogul E, Cennet O, Yazici G, Arica Yegin B, Yabanoglu-Ciftci S. Protective role of transforming growth factor-Β3 (TGF-Β3) in the formation of radiation-induced capsular contracture around a breast implant: In vivo experimental study. Int J Pharm 2024; 665:124715. [PMID: 39284424 DOI: 10.1016/j.ijpharm.2024.124715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 09/21/2024]
Abstract
Postmastectomy radiotherapy causes capsular contracture due to fibroproliferation of the capsular tissue around the implant. In fibrosis, unlike normal wound healing, structural and functional disorders are observed in the tissues caused by excessive/irregular accumulation of extracellular matrix proteins. It has been reported that transforming growth factor-β3 (TGF-β3) prevents and reverses fibrosis in various tissues or provides scarless healing with its antifibrotic effect. Additionally, TGF-β3 has been shown to reduce fibrosis in radiotherapy-induced fibrosis syndrome. However, no study in the literature investigates the effects of exogenously applied TGF-β3 on capsular contracture in aesthetic or reconstructive breast implant application. TGF-β3, which has a very short half-life, has low bioavailability with parenteral administration. Within the scope of this study, free TGF-β3 was loaded into the nanoparticles to increase its low bioavailability and extend its duration of action by providing controlled release. The aim of this study is to investigate the preventive/improving effects of radiation induced capsular contracture using chitosan film formulations containing TGF-β3 loaded poly(lactic-co-glycolic acid)-b-poly(ethylene glycol) (PLGA-b-PEG) nanoparticles in implant-based breast reconstruction. In the characterization studies of nanoparticles, the particle size and zeta potential of the TGF-β3-loaded PLGA-b-PEG nanoparticle formulation selected to be used in the treatment group were found to be 123.60 ± 2.09 nm and -34.87 ± 1.42 mV, respectively. The encapsulation efficiency of the formulation was calculated as 99.91 %. A controlled release profile was obtained in in vitro release studies. Chitosan film formulations containing free TGF-β3 or TGF-β3-loaded PLGA-b-PEG nanoparticles were used in in vivo studies. In animal studies, rats were randomly distributed into 6 groups (n = 8) as sham, implant, implant + radiotherapy, implant + radiotherapy + chitosan film containing unloaded nanoparticles, implant + radiotherapy + chitosan film containing free TGF-β3, implant + radiotherapy + chitosan film containing TGF-β3 loaded nanoparticle. In all study groups, a 2 cm incision was made along the posterior axillary line at the thoracic vertebral level in rats to reach the lateral edge of the latissimus dorsi. The fascial attachment to the chest wall was then bluntly dissected to create a pocket for the implants. In the treatment groups, the wound was closed after films were placed on the outer surface of the implants. After administering prophylactic antibiotics, rats were subjected to irradiation with 10 Gy photon beams targeted to each implant site. Each implant and the surrounding excised tissue were subjected to the necessary procedures for histological (capsule thickness, cell density), immunohistochemical, and biochemical (α-SMA, vimentin, collagen type I and type III, TGF-β1 and TGF-β3: expression level/protein level) examinations. It was determined that the levels of TGF-β1 and TGF-β3 collagen type III, which decreased as a result of radiotherapy, were brought to the control level with free TGF-β3 film and TGF-β3 nanoparticle film formulations. Histological analyses, consistent with biochemical analyses, showed that thick collagen and fibrosis, which increased with radiotherapy, were brought to the control level with free TGF-β3 film and TGF-β3 nanoparticle film treatments. In biochemical analyses, the decrease in thick collagen was compatible with the decrease in the collagen type I/type III ratio in the free TGF-β3 film and TGF-β3 nanoparticle film groups. Changes in protein expression show that TGF-β3 loaded nanoparticles are more successful than free TGF-β3 in wound healing. In line with these results and the literature, it is thought that the balance of TGF-β1 and TGF-β3 should be maintained to ensure scarless wound healing with no capsule contracture.
Collapse
Affiliation(s)
- Aysima Sezer
- Hacettepe University, Faculty of Pharmacy, Department of Biochemistry, 06100 Ankara, Turkey
| | - Hulya Ozalp
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, 06100 Ankara, Turkey
| | - Bercis Imge Ucar-Goker
- Kütahya Health Sciences University, Faculty of Medicine, Department of General Surgery, 43000 Kutahya, Turkey
| | - Ayse Gencer
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Ece Ozogul
- Hacettepe University, Department of Pathology, 06100 Ankara, Turkey
| | - Omer Cennet
- Hacettepe University, Faculty of Medicine, Department of General Surgery, 06100 Ankara, Turkey
| | - Gozde Yazici
- Hacettepe University, Faculty of Medicine, Department of Radiation Oncology, 06100 Ankara, Turkey
| | - Betul Arica Yegin
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, 06100 Ankara, Turkey; Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Samiye Yabanoglu-Ciftci
- Hacettepe University, Faculty of Pharmacy, Department of Biochemistry, 06100 Ankara, Turkey; Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, 06100 Ankara, Turkey; Hacettepe University, Institute of Health Sciences, Department of One Health, 06100 Ankara, Turkey.
| |
Collapse
|
6
|
Yin M, Tong X, Feng Y, Zhang Z, Zhu M, Qiu Q, Huang Y, Hao X, Liu Z, Hu X, Gong C. Polyhedrin microcrystals embedded with bFGF promote wound healing. Int J Biol Macromol 2024; 282:136711. [PMID: 39490869 DOI: 10.1016/j.ijbiomac.2024.136711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
Growth factors play a critical role in wound healing, and finding a suitable biosustained-release system has always been a research hotspot. Bombyx mori cypovirus (BmCPV) is an insect virus, which produces polyhedra that encapsulate progeny virions. In this study, we found that the viral structural protein VP7 encoded by the BmCPV genomic dsRNAs S7 segment can interact with polyhedrin (Polh) encoded by the BmCPV genomic dsRNAs S10 segment. We also confirmed that the amino acid sequence at position 331-360 (VP7-tag) of VP7 is needed to interact with Polh. We found that VP7-tag can be used as an immobilization signal to direct the incorporation of foreign proteins into polyhedra. Furthermore, we constructed polyhedra (bFGF-polyhedra) containing basic fibroblast growth factor (bFGF) using a baculovirus expression system co-expressing Polh and bFGF-VP7 (fusion of VP7-tag to C-terminus of bFGF). We found that bFGF-VP7 embedded into polyhedra was difficult to degrade in the natural environment, and bFGF-VP7 was continuously released from the polyhedra, enhancing cell proliferation and migration. The animal model was used to assess the effect of bFGF-polyhedra spray on the healing of full-thickness wounds. bFGF-polyhedra promoted the expression of TGF-β1, α-SMA, and PCNA, inhibited the expression of proinflammatory factors NF-κB and COX-2, promoted the proliferation and differentiation of fibroblasts, enhanced collagen production and epidermal regeneration, and improved wound healing. These results indicated that bFGF-polyhedra has a promising potential for accelerating wound healing.
Collapse
Affiliation(s)
- Mei Yin
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Xinyu Tong
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Yongjie Feng
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Ziyao Zhang
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Min Zhu
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Qunnan Qiu
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Yuqing Huang
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Xinyue Hao
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Zhuo Liu
- School of Life Science, Soochow University, Suzhou 215123, China
| | - Xiaolong Hu
- School of Life Science, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology and Ecological Research, Soochow University, Suzhou 215123, China.
| | - Chengliang Gong
- School of Life Science, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology and Ecological Research, Soochow University, Suzhou 215123, China.
| |
Collapse
|
7
|
Zohri M, Arefian E, Azizi Z, Akbari Javar H, Shadboorestan A, Fatahi Y, Chogan F, Taheri M, Karoobi S, Aghaee-Bakhtiari SH, Bonakdar S, Gazori T, Mohammadi S, Saadatpour F, Ghahremani MH. Activation of the BMP2/SMAD4 signaling pathway for enhancing articular cartilage regeneration of mesenchymal stem cells utilizing chitosan/alginate nanoparticles on 3D extracellular matrix scaffold. Int J Biol Macromol 2024; 277:133995. [PMID: 39038571 DOI: 10.1016/j.ijbiomac.2024.133995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
This study investigated the efficacy of using chitosan/alginate nanoparticles loaded with recombinant human bone morphogenetic-2 (rhBMP-2) and SMAD4 encoding plasmid to enhance the chondrogenesis of human bone marrow mesenchymal stem cells (hBM-MSCs) seeded on an extracellular matrix (ECM). The research treatments included the stem cells treated with the biological cocktail (BC), negative control (NC), hBM-MSCs with chondrogenic medium (MCM), hBM-MSCs with naked rhBMP-2 and chondrogenic medium (NB/C), and hBM-MSCs with naked rhBMP-2 and chondrogenic medium plus SMAD4 encoding plasmid transfected with polyethyleneimine (PEI) (NB/C/S/P). The cartilage differentiation was performed with real-time quantitative PCR analysis and alizarin blue staining. The data indicated that the biological cocktail (BC) exhibited significantly higher expression of cartilage-related genes compared to significant differences with MCM and negative control (NC) on chondrogenesis. In the (NB/C/S/P), the expression levels of SOX9 and COLX were lower than those in the BC group. The expression pattern of the ACAN gene was similar to COL2A1 changes suggesting that it holds promising potential for cartilage regeneration.
Collapse
Affiliation(s)
- Maryam Zohri
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences.
| | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Akbari Javar
- Departments of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Faraz Chogan
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sepideh Karoobi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Center, Mashhad University of Medical Science, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Taraneh Gazori
- Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 1917733831 Tehran, Iran
| | - Saeid Mohammadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Fatemeh Saadatpour
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
8
|
Kinali H, Kalaycioglu GD, Boyacioglu O, Korkusuz P, Aydogan N, Vargel I. Clinic-oriented injectable smart material for the treatment of diabetic wounds: Coordinating the release of GM-CSF and VEGF. Int J Biol Macromol 2024; 276:133661. [PMID: 38992546 DOI: 10.1016/j.ijbiomac.2024.133661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/16/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Chronic wounds are often caused by diabetes and present a challenging clinical problem due to vascular problems leading to ischemia. This inhibits proper wound healing by delaying inflammatory responses and angiogenesis. To address this problem, we have developed injectable particle-loaded hydrogels which sequentially release Granulocyte-macrophage- colony-stimulating-factor (GM-CSF) and Vascular endothelial growth factor (VEGF) encapsulated in polycaprolactone-lecithin-geleol mono-diglyceride hybrid particles. GM-CSF promotes inflammation, while VEGF facilitates angiogenesis. The hybrid particles (200-1000 nm) designed within the scope of the study can encapsulate the model proteins Bovine Serum Albumin 65 ± 5 % and Lysozyme 77 ± 10 % and can release stably for 21 days. In vivo tests and histological findings revealed that in the hydrogels containing GM-CSF/VEGF-loaded hybrid particles, wound depth decreased, inflammation phase increased, and fibrotic scar tissue decreased, while mature granulation tissue was formed on day 10. These findings confirm that the hybrid particles first initiate the inflammation phase by delivering GM-CSF, followed by VEGF, increasing the number of vascularization and thus increasing the healing rate of wounds. We emphasize the importance of multi-component and sequential release in wound healing and propose a unifying therapeutic strategy to sequentially deliver ligands targeting wound healing stages, which is very important in the treatment of the diabetic wounds.
Collapse
Affiliation(s)
- Hurmet Kinali
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara 06800, Turkey
| | - Gokce Dicle Kalaycioglu
- Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara 06800, Turkey
| | - Ozge Boyacioglu
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara 06800, Turkey; Department of Medical Biochemistry, Faculty of Medicine, Atılım University, 06830 Gölbaşı, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, 06100 Sıhhiye, Ankara, Turkey
| | - Nihal Aydogan
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara 06800, Turkey; Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara 06800, Turkey.
| | - Ibrahim Vargel
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara 06800, Turkey; Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hacettepe University, 06560 Ankara, Turkey.
| |
Collapse
|
9
|
Syromiatnikova VY, Kvon AI, Starostina IG, Gomzikova MO. Strategies to enhance the efficacy of FGF2-based therapies for skin wound healing. Arch Dermatol Res 2024; 316:405. [PMID: 38878084 DOI: 10.1007/s00403-024-02953-x] [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: 01/22/2024] [Revised: 01/22/2024] [Accepted: 04/26/2024] [Indexed: 06/23/2024]
Abstract
Basic fibroblast growth factor (FGF2 or bFGF) is critical for optimal wound healing. Experimental studies show that local application of FGF2 is a promising therapeutic approach to stimulate tissue regeneration, including for the treatment of chronic wounds that have a low healing potential or are characterised by a pathologically altered healing process. However, the problem of low efficiency of growth factors application due to their rapid loss of biological activity in the aggressive proteolytic environment of the wound remains. Therefore, ways to preserve the efficacy of FGF2 for wound treatment are being actively developed. This review considers the following strategies to improve the effectiveness of FGF2-based therapy: (1) use of vehicles/carriers for delivery and gradual release of FGF2; (2) chemical modification of FGF2 to increase the stability of the molecule; (3) use of genetic constructs encoding FGF2 for de novo synthesis of protein in the wound. In addition, this review discusses FGF2-based therapeutic strategies that are undergoing clinical trials and demonstrating the efficacy of FGF2 for skin wound healing.
Collapse
Affiliation(s)
- V Y Syromiatnikova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russia
| | - A I Kvon
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russia
| | - I G Starostina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russia
| | - M O Gomzikova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russia.
| |
Collapse
|
10
|
Liu X, Peng S, Pei Y, Huo Y, Zong Y, Ren J, Zhao J. Facile fabrication of chitosan/hyaluronic acid hydrogel-based wound closure material Co-loaded with gold nanoparticles and fibroblast growth factor to improve anti-microbial and healing efficiency in diabetic wound healing and nursing care. Regen Ther 2024; 26:1018-1029. [PMID: 39553541 PMCID: PMC11565426 DOI: 10.1016/j.reth.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Generally, diabetic wounds heal very slowly and inefficiently with an increasing risk of infections. Recent nanotechnology and biomaterial advances elaborate developed multi-functional hydrogels and nanoparticles offer promising solutions to accelerate wound healing for diabetic patients. This research work demonstrates to use of solvent diffusion method to develop hydrogel nanocomposites composed of chitosan (CS), hyaluronic acid (HA), gold (Au), and fibroblast growth factors (FGF). The biological analysis of nanocomposites exhibited enhanced wound healing efficiency by incorporating bioactive molecules like FGF and bioactive Au nanoparticles. In vitro, cell compatibility analysis (MTT assay) of prepared hydrogel nanocomposites was studied on fibroblast cell lines NIH-3T3-L1 and L929 and exhibited greater cell survival ability (>90 %), cell proliferation and migration ability, which demonstrated the suitability of nanocomposite for wound healing treatment. In vitro, anti-bacterial analyses established that FGF-Au@CS/HA has strong antibacterial effectiveness against gram-positive and gram-negative pathogens. The observation of the present research revealed that prepared FGF-Au@CS/HA hydrogel composites could be a suitable biomaterial for diabetic wound care, potentially improving its antibacterial and healing efficacies.
Collapse
Affiliation(s)
- Xin Liu
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Shengwei Peng
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yongju Pei
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yuanyuan Huo
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yadi Zong
- Department of Pediatric Surgery, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jianwei Ren
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jing Zhao
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
11
|
Kumar M, Kumar D, Kumar D, Garg Y, Chopra S, Bhatia A. Therapeutic Potential of Nanocarrier Mediated Delivery of Peptides for Wound Healing: Current Status, Challenges and Future Prospective. AAPS PharmSciTech 2024; 25:108. [PMID: 38730090 DOI: 10.1208/s12249-024-02827-5] [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: 02/07/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Wound healing presents a complex physiological process that involves a sequence of events orchestrated by various cellular and molecular mechanisms. In recent years, there has been growing interest in leveraging nanomaterials and peptides to enhance wound healing outcomes. Nanocarriers offer unique properties such as high surface area-to-volume ratio, tunable physicochemical characteristics, and the ability to deliver therapeutic agents in a controlled manner. Similarly, peptides, with their diverse biological activities and low immunogenicity, hold great promise as therapeutics in wound healing applications. In this review, authors explore the potential of peptides as bioactive components in wound healing formulations, focusing on their antimicrobial, anti-inflammatory, and pro-regenerative properties. Despite the significant progress made in this field, several challenges remain, including the need for standardized characterization methods, optimization of biocompatibility and safety profiles, and translation from bench to bedside. Furthermore, developing multifunctional nanomaterial-peptide hybrid systems represents promising avenues for future research. Overall, the integration of nanomaterials made up of natural or synthetic polymers with peptide-based formulations holds tremendous therapeutic potential in advancing the field of wound healing and improving clinical outcomes for patients with acute and chronic wounds.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Dikshant Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Yogesh Garg
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Shruti Chopra
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India.
| |
Collapse
|
12
|
Liu G, Yang Y, Liu Y, Li Y, Jiang M, Li Y, Meng Z, Zhao Z, Liu Z, Liu J, Ge D, Wang H. Injectable and Thermosensitive Hydrogel with Platelet-Rich Plasma for Enhanced Biotherapy of Skin Wound Healing. Adv Healthc Mater 2024; 13:e2303930. [PMID: 38306618 DOI: 10.1002/adhm.202303930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/23/2024] [Indexed: 02/04/2024]
Abstract
The rapid and effective healing of skin wounds resulted from severe injuries and full-layer skin defects remains a pressing clinical challenge in contemporary medical practice. The reduction of wound infection and rapid healing is helpful to rebuild and repair skin tissue. Here, a thermosensitive chitosan-based wound dressing hydrogel incorporating β-glycerophosphate (GP), hydroxy propyl cellulose (HPC), graphene oxide (GO), and platelet-rich plasma (PRP) is developed, which exhibits the dual functions of antibacterial properties and repair promotion. GP and HPC enhance the mechanical properties through forming hydrogen bonding connection, while GO produces local heat under near-infrared light, leading to improved blood circulation and skin recovery. Notably, antibacterial properties against Pseudomonas aeruginosa, and control-release of growth factors from PRP are also achieved based on the system. In vitro experiments reveal its biocompatibility, and ability to promote cell proliferation and migration. Animal experiments demonstrate that the epithelial repair and collagen deposition can be promoted during skin wound healing in Sprague Dawley rats. Moreover, a reduction in wound inflammation levels and the improvement of wound microenvironment are observed, collectively fostering effective wound healing. Therefore, the composite hydrogel system incorporated with GO and PRP can be a promising dressing for the treatment of skin wounds.
Collapse
Affiliation(s)
- Gengjun Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Yuanming Yang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Yan Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Ying Li
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Mingyuan Jiang
- College of Materials Science and Engineering, Institute for Graphene Applied, Technology Innovation, Qingdao University, Ningxia Road 308, Qingdao, 266071, China
| | - Yaxin Li
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Zifan Meng
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Zhen Zhao
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Zheng Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Jiao Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Dongmei Ge
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, P. R. China
| |
Collapse
|
13
|
Kareem YG, Rachid S, Al-Jaf O. Synthesis and characterization of novel poly cysteine methacrylate nanoparticles and their morphology and size studies. RSC Adv 2024; 14:13474-13481. [PMID: 38665499 PMCID: PMC11044863 DOI: 10.1039/d4ra00067f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Polymer nanoparticles (PNPs) have significantly advanced the field of biomedicine, showcasing the remarkable potential for precise drug delivery, administration of nutraceuticals, diagnostics/imaging applications, and the fabrication of biocompatible materials, among other uses. Despite these promising developments, the invention faces notable challenges related to biodegradability, bioactivity, target-site specificity, particle size, carrier efficiency, and controlled release. Addressing these concerns is essential for optimizing the functionality and impact of PNPs in biomedical applications. Here, new poly cysteine methacrylate nanoparticles (PCMANPs), ca. (200 nm) in size have been synthesized from the cysteine methacrylate (CysMA) monomer using different strategies, including emulsion and inverse emulsion polymerization techniques. The monomer was synthesized using the Michael addition reaction, involving the addition of 3-(acryloyloxy)-2-hydroxypropyl methacrylate to the sulfhydryl group (-SH) of the cysteine (Cys) active site, with the aid of dimethyl phenyl phosphine (DMPP) as a nucleophilic agent as previously reported. To enhance nano-polymerization, a thorough exploration of various initiators, including ammonium persulfate (APS) and 4,4'-azobis (4-cyanovaleric acid) (ACVA), alongside surfactants, such as polyvinyl alcohol (PVA), polyvinyl pyrrolidone (PVP), and sodium dodecyl sulfate (SDS), was conducted. Additionally, critical parameters, such as reaction time, temperature, and solvents, were systematically investigated due to their substantial influence on the shape, size, stability, and morphology of the synthesized polymer nanoparticles. This comprehensive approach aims to optimize the synthesis process, ensuring precise control over the key characteristics of the resulting nanoparticles for enhanced performance in diverse applications. Various characterization techniques, including field emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), nuclear magnetic resonance (NMR), Raman spectroscopy, Fourier-transform infrared spectroscopy (FTIR), zeta potential, and zeta sizer dynamic light scattering (DLS) analysis, were utilized to investigate purity, morphology, and particle size of the PNPs. As a result, a spherical, monodispersed (homogenized), and stable PCMANP with defined size and morphology was achieved. This may exhibit a remarkable achievement in the future of drug delivery systems and therapeutic index.
Collapse
Affiliation(s)
- Yaseen G Kareem
- Charmo Center for Research, Training, and Consultancy, Charmo University Chamchamal, Kurdistan Region 46023 Iraq
- Medical Laboratory Science, Komar University for Science and Technology Sulaymaniah, Kurdistan Region 46001 Iraq
| | - Shwan Rachid
- Department of Medical Laboratory Science, College of Science, Charmo University Chamchamal, Kurdistan Region 46023 Iraq
| | - O Al-Jaf
- Department of Applied Chemistry, College of Science, Charmo University Chamchamal, Kurdistan Region 46023 Iraq
| |
Collapse
|
14
|
Alavi SE, Alavi SZ, Nisa MU, Koohi M, Raza A, Ebrahimi Shahmabadi H. Revolutionizing Wound Healing: Exploring Scarless Solutions through Drug Delivery Innovations. Mol Pharm 2024; 21:1056-1076. [PMID: 38288723 DOI: 10.1021/acs.molpharmaceut.3c01072] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Human skin is the largest organ and outermost surface of the human body, and due to the continuous exposure to various challenges, it is prone to develop injuries, customarily known as wounds. Although various tissue engineering strategies and bioactive wound matrices have been employed to speed up wound healing, scarring remains a significant challenge. The wound environment is harsh due to the presence of degradative enzymes and elevated pH levels, and the physiological processes involved in tissue regeneration operate on distinct time scales. Therefore, there is a need for effective drug delivery systems (DDSs) to address these issues. The objective of this review is to provide a comprehensive exposition of the mechanisms underlying the skin healing process, the factors and materials used in engineering DDSs, and the different DDSs used in wound care. Furthermore, this investigation will delve into the examination of emergent technologies and potential avenues for enhancing the efficacy of wound care devices.
Collapse
Affiliation(s)
- Seyed Ebrahim Alavi
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Seyed Zeinab Alavi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran
| | - Mehr Un Nisa
- Nishtar Medical University and Hospital, Multan 60000, Pakistan
| | - Maedeh Koohi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran
| | - Aun Raza
- School of Pharmacy, Jiangsu University, Zhenjiang 202013, PR China
| | - Hasan Ebrahimi Shahmabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran
| |
Collapse
|
15
|
Ruiz-González N, Esporrín-Ubieto D, Hortelao AC, Fraire JC, Bakenecker AC, Guri-Canals M, Cugat R, Carrillo JM, Garcia-Batlletbó M, Laiz P, Patiño T, Sánchez S. Swarms of Enzyme-Powered Nanomotors Enhance the Diffusion of Macromolecules in Viscous Media. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309387. [PMID: 38200672 DOI: 10.1002/smll.202309387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Indexed: 01/12/2024]
Abstract
Over the past decades, the development of nanoparticles (NPs) to increase the efficiency of clinical treatments has been subject of intense research. Yet, most NPs have been reported to possess low efficacy as their actuation is hindered by biological barriers. For instance, synovial fluid (SF) present in the joints is mainly composed of hyaluronic acid (HA). These viscous media pose a challenge for many applications in nanomedicine, as passive NPs tend to become trapped in complex networks, which reduces their ability to reach the target location. This problem can be addressed by using active NPs (nanomotors, NMs) that are self-propelled by enzymatic reactions, although the development of enzyme-powered NMs, capable of navigating these viscous environments, remains a considerable challenge. Here, the synergistic effects of two NMs troops, namely hyaluronidase NMs (HyaNMs, Troop 1) and urease NMs (UrNMs, Troop 2) are demonstrated. Troop 1 interacts with the SF by reducing its viscosity, thus allowing Troop 2 to swim more easily through the SF. Through their collective motion, Troop 2 increases the diffusion of macromolecules. These results pave the way for more widespread use of enzyme-powered NMs, e.g., for treating joint injuries and improving therapeutic effectiveness compared with traditional methods.
Collapse
Affiliation(s)
- Noelia Ruiz-González
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - David Esporrín-Ubieto
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ana C Hortelao
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Juan C Fraire
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Anna C Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Marta Guri-Canals
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ramón Cugat
- Mutualidad de Futbolistas - Delegación Catalana, Federación Española de Fútbol, Barcelona, 08010, Spain
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - José María Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain. García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, 46115, Spain
| | | | - Patricia Laiz
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - Tania Patiño
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, 5612 AZ, The Netherlands
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
- Institució Catalana de Recerca i Estudies Avancats (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
16
|
Zhu S, Wang J, Suo M, Huang H, Liu X, Wang J, Li Z. Can extracellular vesicles be considered as a potential frontier in the treatment of intervertebral disc disease? Ageing Res Rev 2023; 92:102094. [PMID: 37863436 DOI: 10.1016/j.arr.2023.102094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
As a global public health problem, low back pain (LBP) caused by intervertebral disc degeneration (IDD) seriously affects patients' quality of life. In addition, the prevalence of IDD tends to be younger, which brings a huge burden to individuals and society economically. Current treatments do not delay or reverse the progression of IDD. The emergence of biologic therapies has brought new hope for the treatment of IDD. Among them, extracellular vesicles (EVs), as nanoscale bioactive substances that mediate cellular communication, have now produced many surprising results in the research of the treatment of IDD. This article reviews the mechanisms and roles of EVs in delaying IDD and describes the prospects and challenges of EVs.
Collapse
Affiliation(s)
- Shengxu Zhu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Junlin Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, the People's Republic of China; Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, the People's Republic of China.
| |
Collapse
|
17
|
Beheshtizadeh N, Gharibshahian M, Bayati M, Maleki R, Strachan H, Doughty S, Tayebi L. Vascular endothelial growth factor (VEGF) delivery approaches in regenerative medicine. Biomed Pharmacother 2023; 166:115301. [PMID: 37562236 DOI: 10.1016/j.biopha.2023.115301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023] Open
Abstract
The utilization of growth factors in the process of tissue regeneration has garnered significant interest and has been the subject of extensive research. However, despite the fervent efforts invested in recent clinical trials, a considerable number of these studies have produced outcomes that are deemed unsatisfactory. It is noteworthy that the trials that have yielded the most satisfactory outcomes have exhibited a shared characteristic, namely, the existence of a mechanism for the regulated administration of growth factors. Despite the extensive exploration of drug delivery vehicles and their efficacy in delivering certain growth factors, the development of a reliable predictive approach for the delivery of delicate growth factors like Vascular Endothelial Growth Factor (VEGF) remains elusive. VEGF plays a crucial role in promoting angiogenesis; however, the administration of VEGF demands a meticulous approach as it necessitates precise localization and transportation to a specific target tissue. This process requires prolonged and sustained exposure to a low concentration of VEGF. Inaccurate administration of drugs, either through off-target effects or inadequate delivery, may heighten the risk of adverse reactions and potentially result in tumorigenesis. At present, there is a scarcity of technologies available for the accurate encapsulation of VEGF and its subsequent sustained and controlled release. The objective of this review is to present and assess diverse categories of VEGF administration mechanisms. This paper examines various systems, including polymeric, liposomal, hydrogel, inorganic, polyplexes, and microfluidic, and evaluates the appropriate dosage of VEGF for multiple applications.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Bayati
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Reza Maleki
- Department of Chemical Technologies, Iranian Research Organization for Science and Technology (IROST), P.O. Box 33535111, Tehran, Iran.
| | - Hannah Strachan
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Sarah Doughty
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
18
|
Saberi A, Kouhjani M, Mohammadi M, Hosta-Rigau L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: a cutting-edge approach. J Nanobiotechnology 2023; 21:351. [PMID: 37770928 PMCID: PMC10536787 DOI: 10.1186/s12951-023-02115-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Despite the recent advances in the development of bone graft substitutes, treatment of critical size bone defects continues to be a significant challenge, especially in the elderly population. A current approach to overcome this challenge involves the creation of bone-mimicking scaffolds that can simultaneously promote osteogenesis and angiogenesis. In this context, incorporating multiple bioactive agents like growth factors, genes, and small molecules into these scaffolds has emerged as a promising strategy. To incorporate such agents, researchers have developed scaffolds incorporating nanoparticles, including nanoparticulate carriers, inorganic nanoparticles, and exosomes. Current paper provides a summary of the latest advancements in using various bioactive agents, drugs, and cells to synergistically promote osteogenesis and angiogenesis in bone-mimetic scaffolds. It also discusses scaffold design properties aimed at maximizing the synergistic effects of osteogenesis and angiogenesis, various innovative fabrication strategies, and ongoing clinical studies.
Collapse
Affiliation(s)
- Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Kouhjani
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
19
|
Abbasi R, Shineh G, Mobaraki M, Doughty S, Tayebi L. Structural parameters of nanoparticles affecting their toxicity for biomedical applications: a review. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2023; 25:43. [PMID: 36875184 PMCID: PMC9970140 DOI: 10.1007/s11051-023-05690-w] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Rapidly growing interest in using nanoparticles (NPs) for biomedical applications has increased concerns about their safety and toxicity. In comparison with bulk materials, NPs are more chemically active and toxic due to the greater surface area and small size. Understanding the NPs' mechanism of toxicity, together with the factors influencing their behavior in biological environments, can help researchers to design NPs with reduced side effects and improved performance. After overviewing the classification and properties of NPs, this review article discusses their biomedical applications in molecular imaging and cell therapy, gene transfer, tissue engineering, targeted drug delivery, Anti-SARS-CoV-2 vaccines, cancer treatment, wound healing, and anti-bacterial applications. There are different mechanisms of toxicity of NPs, and their toxicity and behaviors depend on various factors, which are elaborated on in this article. More specifically, the mechanism of toxicity and their interactions with living components are discussed by considering the impact of different physiochemical parameters such as size, shape, structure, agglomeration state, surface charge, wettability, dose, and substance type. The toxicity of polymeric, silica-based, carbon-based, and metallic-based NPs (including plasmonic alloy NPs) have been considered separately.
Collapse
Affiliation(s)
- Reza Abbasi
- Department of Bioengineering, McGill University, Montreal, QC Canada
| | - Ghazal Shineh
- Biomaterial Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran, 15916-34311 Iran
| | - Mohammadmahdi Mobaraki
- Biomaterial Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran, 15916-34311 Iran
| | - Sarah Doughty
- Marquette University School of Dentistry, Milwaukee, WI USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI USA
| |
Collapse
|
20
|
Song H, Kim DI, Abbasi SA, Latifi Gharamaleki N, Kim E, Jin C, Kim S, Hwang J, Kim JY, Chen XZ, Nelson BJ, Pané S, Choi H. Multi-target cell therapy using a magnetoelectric microscale biorobot for targeted delivery and selective differentiation of SH-SY5Y cells via magnetically driven cell stamping. MATERIALS HORIZONS 2022; 9:3031-3038. [PMID: 36129054 PMCID: PMC9704487 DOI: 10.1039/d2mh00693f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cell therapy refers to a treatment that involves the delivery of cells or cellular material by means of injection, grafting, or implantation in order to replace damaged tissue and restore its function, or to aid the body in fighting disease. However, limitations include poor targeting delivery and low therapeutic efficacy due to low cell survival. Hence, novel approaches are required to increase cell delivery efficiency and enhance therapeutic efficacy via selective cell differentiation at target areas. Here, we present a stamping magnetoelectric microscale biorobot (SMMB) consisting of neuron-like cell spheroids loaded with magnetoelectric nanoparticles. The SMMB enables not only effective targeted delivery of cells to multiple target areas (via minimally invasive stamping employing magnetic actuation) but also facilitates selective neuronal differentiation via magnetoelectric (ME) stimulation. This ensures rapid colonization and enhances efficacy. SMMBs were fabricated using SH-SY5Y cells. Magnetoelectric nanoparticles for ME stimulation responded to an alternating magnetic field that ensured targeted cell differentiation. Multi-target cell therapy facilitated the targeted delivery and selective differentiation of SH-SY5Y cells to multiple regions using a single SMMB with rotating and alternating magnetic fields for delivery and ME stimulation. This promising tool may overcome the limitations of existing cell therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Hyunseok Song
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Dong-In Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sarmad Ahmad Abbasi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Nader Latifi Gharamaleki
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Eunhee Kim
- IMsystem Co., Ltd., Daegu, Republic of Korea
| | - Chaewon Jin
- Department of Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Samhwan Kim
- Brain Science Department, Daegu Gyeongbuk Institute of Science and Technology (DGIST) 711-873, Daegu, South Korea
| | - Junsun Hwang
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Jin-Young Kim
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
- IMsystem Co., Ltd., Daegu, Republic of Korea
| | - Xiang-Zhong Chen
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, CH-8092, Switzerland.
| | - Bradley J Nelson
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, CH-8092, Switzerland.
| | - Salvador Pané
- Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, CH-8092, Switzerland.
| | - Hongsoo Choi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- DGIST-ETH Microrobotics Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
- Robotics Research Center, DGIST, Daegu, Republic of Korea
| |
Collapse
|
21
|
Kasowanjete P, Houreld NN, Abrahamse H. The effect of photomodulation on fibroblast growth factor and the Ras/MAPK signalling pathway: a review. J Wound Care 2022; 31:832-845. [DOI: 10.12968/jowc.2022.31.10.832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Objective: Current therapies and technologies used to treat hard-to-heal diabetic wounds are limited to a 50% healing rate. The rise in the percentage of lower limb non-traumatic amputations in patients with diabetes has caused an increased demand for alternative, effective and safe treatment modalities. Photobiomodulation therapy (PBMT) utilises light to induce physiological changes and provide therapeutic benefits and has been shown to increase the healing of hard-to-heal wounds through the release of growth factors. The aim of this narrative review is to investigate the effect of photobiomodulation (PBM) on fibroblast growth factor (FGF) and the role of the Ras/MAPK signalling pathway in diabetic wound healing. Method: Relevant journal articles were obtained through PubMed and Google Scholar. Results: Experimental and clinical findings from the review show that PBM can stimulate the release of growth factors, including FGF, an essential cytokine in wound healing, and one which is present at lower concentrations in diabetic wounds. There is also activation of the Ras/MAPK signalling pathway. Conclusion: One mechanism through which healing may be stimulated by PBM is via the FGF-Ras/MAPK signalling pathway, although strong evidence under hyperglycaemic conditions is lacking.
Collapse
Affiliation(s)
| | - Nicolette N Houreld
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
22
|
The Bridging Effect of Controlled-Release Glial Cell-Derived Neurotrophic Factor Microcapsules within Nerve Conduits on Rat Facial Nerve Regeneration. DISEASE MARKERS 2022; 2022:8942985. [PMID: 35774850 PMCID: PMC9239766 DOI: 10.1155/2022/8942985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022]
Abstract
Objectives The study is aimed at exploring the effect of the controlled release of the glial-derived neurotrophic factor (GDNF) on nerve regeneration. Methods The PLGA/chitosan composite nerve conduit was used to bridge the dissected trunk of the rat facial nerve. GDNF microcapsules were loaded into the nerve conduit. Nine weeks after surgery, the facial nerve zygomatic and buccal branches were labeled with fluorescent indicators. The incorrectly grown facial neurons were reversed and counted. The facial nerve functional recovery was assessed by measuring the maximum evoked potential. Results The nerve conduit was filled with different regenerating factors, such as the GDNF, GDNF microcapsules, or saline (control). The number of incorrectly regenerated neurons was lower with the nerve conduits filled with GDNF microcapsules than with those supplied with just the GDNF. However, neither the GDNF nor GDNF microcapsules affected the number of regenerated neurons. The functional recovery of the facial nerve was the best, with the nerve conduit filled with GDNF microcapsules closest to the healthy uncut facial nerve. Conclusion The stable slow-release GNDF microcapsule inside the biodegradable nerve conduit can reduce the extent of incorrect growth of the facial nerve neuron when bridging the dissected rat facial nerve trunk. The technique has a good effect on functional nerve recovery.
Collapse
|
23
|
Barisón MJ, Nogoceke R, Josino R, Horinouchi CDDS, Marcon BH, Correa A, Stimamiglio MA, Robert AW. Functionalized Hydrogels for Cartilage Repair: The Value of Secretome-Instructive Signaling. Int J Mol Sci 2022; 23:ijms23116010. [PMID: 35682690 PMCID: PMC9181449 DOI: 10.3390/ijms23116010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
Cartilage repair has been a challenge in the medical field for many years. Although treatments that alleviate pain and injury are available, none can effectively regenerate the cartilage. Currently, regenerative medicine and tissue engineering are among the developed strategies to treat cartilage injury. The use of stem cells, associated or not with scaffolds, has shown potential in cartilage regeneration. However, it is currently known that the effect of stem cells occurs mainly through the secretion of paracrine factors that act on local cells. In this review, we will address the use of the secretome—a set of bioactive factors (soluble factors and extracellular vesicles) secreted by the cells—of mesenchymal stem cells as a treatment for cartilage regeneration. We will also discuss methodologies for priming the secretome to enhance the chondroregenerative potential. In addition, considering the difficulty of delivering therapies to the injured cartilage site, we will address works that use hydrogels functionalized with growth factors and secretome components. We aim to show that secretome-functionalized hydrogels can be an exciting approach to cell-free cartilage repair therapy.
Collapse
|
24
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
25
|
Bousalis D, McCrary MW, Vaughn N, Hlavac N, Evering A, Kolli S, Song YH, Morley C, Angelini T, Schmidt CE. Decellularized peripheral nerve as an injectable delivery vehicle for neural applications. J Biomed Mater Res A 2022; 110:595-611. [PMID: 34590403 PMCID: PMC8742792 DOI: 10.1002/jbm.a.37312] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/17/2022]
Abstract
Damage to the nervous system can result in loss of sensory and motor function, paralysis, or even death. To facilitate neural regeneration and functional recovery, researchers have employed biomaterials strategies to address both peripheral and central nervous system injuries. Injectable hydrogels that recapitulate native nerve extracellular matrix are especially promising for neural tissue engineering because they offer more flexibility for minimally invasive applications and provide a growth-permissive substrate for neural cell types. Here, we explore the development of injectable hydrogels derived from decellularized rat peripheral nerves (referred to as "injectable peripheral nerve [iPN] hydrogels"), which are processed using a newly developed sodium deoxycholate and DNase (SDD) decellularization method. We assess the gelation kinetics, mechanical properties, cell bioactivity, and drug release kinetics of the iPN hydrogels. The iPN hydrogels thermally gel when exposed to 37°C in under 20 min and have mechanical properties similar to neural tissue. The hydrogels demonstrate in vitro biocompatibility through support of Schwann cell viability and metabolic activity. Additionally, iPN hydrogels promote greater astrocyte spreading compared to collagen I hydrogels. Finally, the iPN is a promising delivery vehicle of drug-loaded microparticles for a combinatorial approach to neural injury therapies.
Collapse
Affiliation(s)
- Deanna Bousalis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Michaela W. McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Natalie Vaughn
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Nora Hlavac
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Ashley Evering
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Shruti Kolli
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL,Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR
| | - Cameron Morley
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL
| | - Thomas Angelini
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL
| | - Christine E. Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| |
Collapse
|
26
|
Pillai MM, Dandia H, Checker R, Rokade S, Sharma D, Tayalia P. Novel combination of bioactive agents in bilayered dermal patches provides superior wound healing. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102495. [PMID: 34838992 DOI: 10.1016/j.nano.2021.102495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/07/2021] [Indexed: 10/19/2022]
Abstract
In present study, multifunctional bilayered dermal patches with antibacterial, antioxidant and anti-inflammatory properties were developed using solvent casting or electrospinning methods and compared for performance. Top layer was made up of polycaprolactone (PCL) and chitosan (CS) while bottom layer was made of polyvinyl alcohol (PVA) with curcumin nanoparticles and soluble eggshell membrane protein (SESM) as the wound healing agents. Curcumin nanoparticles showed reduction in the production of reactive oxygen species (ROS) and inflammatory cytokines and markers in mice T cells or human macrophages, confirming their antioxidant and anti-inflammatory properties while SESM improved migration of human adult dermal fibroblasts, suggesting its contribution to wound healing. The dermal patches were hemocompatible and antibacterial and also provided adequate absorption of wound exudates, support and components required for recruitment of cells and deposition of extracellular matrix to enable superior wound healing than its commercial counterpart in a full thickness excision wound model in rats.
Collapse
Affiliation(s)
- Mamatha M Pillai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay
| | - Hiren Dandia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Sushama Rokade
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay.
| |
Collapse
|
27
|
Zhou S, Wang Q, Huang A, Fan H, Yan S, Zhang Q. Advances in Skin Wound and Scar Repair by Polymer Scaffolds. Molecules 2021; 26:6110. [PMID: 34684690 PMCID: PMC8541489 DOI: 10.3390/molecules26206110] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/25/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022] Open
Abstract
Scars, as the result of abnormal wound-healing response after skin injury, may lead to loss of aesthetics and physical dysfunction. Current clinical strategies, such as surgical excision, laser treatment, and drug application, provide late remedies for scarring, yet it is difficult to eliminate scars. In this review, the functions, roles of multiple polymer scaffolds in wound healing and scar inhibition are explored. Polysaccharide and protein scaffolds, an analog of extracellular matrix, act as templates for cell adhesion and migration, differentiation to facilitate wound reconstruction and limit scarring. Stem cell-seeded scaffolds and growth factors-loaded scaffolds offer significant bioactive substances to improve the wound healing process. Special emphasis is placed on scaffolds that continuously release oxygen, which greatly accelerates the vascularization process and ensures graft survival, providing convincing theoretical support and great promise for scarless healing.
Collapse
Affiliation(s)
| | | | | | | | - Shuqin Yan
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (S.Z.); (Q.W.); (A.H.); (H.F.)
| | - Qiang Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (S.Z.); (Q.W.); (A.H.); (H.F.)
| |
Collapse
|
28
|
Hosseini M, Shafiee A. Engineering Bioactive Scaffolds for Skin Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101384. [PMID: 34313003 DOI: 10.1002/smll.202101384] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Indexed: 06/13/2023]
Abstract
Large skin wounds pose a major clinical challenge. Scarcity of donor site and postsurgical scarring contribute to the incomplete or partial loss of function and aesthetic concerns in skin wound patients. Currently, a wide variety of skin grafts are being applied in clinical settings. Scaffolds are used to overcome the issues related to the misaligned architecture of the repaired skin tissues. The current review summarizes the contribution of biomaterials to wound healing and skin regeneration and addresses the existing limitations in skin grafting. Then, the clinically approved biologic and synthetic skin substitutes are extensively reviewed. Next, the techniques for modification of skin grafts aiming for enhanced tissue regeneration are outlined, and a summary of different growth factor delivery systems using biomaterials is presented. Considering the significant progress in biomaterial science and manufacturing technologies, the idea of biomaterial-based skin grafts with the ability for scarless wound healing and reconstructing full skin organ is more achievable than ever.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
29
|
Girotra P, Behl T, Sehgal A, Singh S, Bungau S. Investigation of the Molecular Role of Brain-Derived Neurotrophic Factor in Alzheimer's Disease. J Mol Neurosci 2021; 72:173-186. [PMID: 34424488 DOI: 10.1007/s12031-021-01824-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), or abrineurin, is a member of the neurotrophin family of growth factors that acts on both the central and peripheral nervous systems. BDNF is also well known for its cardinal role in normal neural maturation. It binds to at least two receptors at the cell surface known as tyrosine kinase B (TrkB) and p75NTR. Additional neurotrophins that are anatomically linked with BDNF include neurotrophin-3 (NT-3), neurotrophin-4 (NT-4), and nerve growth factor (NGF). It is evident that BDNF levels in patients with Alzheimer's disease (AD) are altered. AD is a progressive disorder and a form of dementia, where the mental function of an elderly person is disrupted. It is associated with a progressive decline in cognitive function, which mainly targets the thinking, memory, and behavior of the person. The degeneration of neurons occurs in the cerebral cortex region of brain. The two major sources responsible for neuronal degeneration are protein fragment amyloid-beta (Aβ), which builds up in the spaces between the nerve cells, known as plaques, disrupting the neuron signaling pathway and leading to dementia, and neurofibrillary tangles (NFTs), which are the twisted fibers of proteins that build up inside the cells. AD is highly prevalent, with recent data indicating nearly 5.8 million Americans aged 65 and older with AD in 2020, and with 80% of patients 75 and older. AD is recognized as the sixth leading cause of death in the USA, and its prevalence is predicted to increase exponentially in the coming years. As AD worsens over time, it becomes increasingly important to understand the exact pathophysiology, biomarkers, and treatment. In this article, we focus primarily on the controversial aspect of BDNF in AD, including its influence on various other proteins and enzymes and the current treatments associated with BDNF, along with future perspectives.
Collapse
Affiliation(s)
- Pragya Girotra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
30
|
Heparin-Eluting Tissue-Engineered Bioabsorbable Vascular Grafts. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11104563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The creation of small-diameter tissue-engineered vascular grafts using biodegradable materials has the potential to change the quality of cardiovascular surgery in the future. The implantation of these tissue-engineered arterial grafts has yet to reach clinical application. One of the reasons for this is thrombus occlusion of the graft in the acute phase. In this paper, we first describe the causes of accelerated thrombus formation and discuss the drugs that are thought to inhibit thrombus formation. We then review the latest research on methods to locally bind the anticoagulant heparin to biodegradable materials and methods to extend the duration of sustained heparin release. We also discuss the results of studies using large animal models and the challenges that need to be overcome for future clinical applications.
Collapse
|
31
|
Pop NL, Nan A, Urda-Cimpean AE, Florea A, Toma VA, Moldovan R, Decea N, Mitrea DR, Orasan R. Chitosan Functionalized Magnetic Nanoparticles to Provide Neural Regeneration and Recovery after Experimental Model Induced Peripheral Nerve Injury. Biomolecules 2021; 11:676. [PMID: 33946445 PMCID: PMC8147170 DOI: 10.3390/biom11050676] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Peripheral nerve injuries have a great impact on a patient's quality of life and a generally poor outcome regarding functional recovery. Lately, studies have focused on different types of nanoparticles and various natural substances for the treatment of peripheral nerve injuries. This is the case of chitosan, a natural compound from the crustaceans' exoskeleton. The present study proposes to combine chitosan benefic properties to the nanoparticles' ability to transport different substances to specific locations and evaluate the effects of magnetic nanoparticles functionalized with chitosan (CMNPs) on peripheral nerve injuries' rehabilitation by using an in vivo experimental model. (2) Methods: CMNPs treatment was administrated daily, orally, for 21 days to rats subjected to right sciatic nerve lesion and compared to the control group (no treatment) by analyzing the sciatic functional index, pain level, body weight, serum nerve growth factor levels and histology, TEM and EDX analysis at different times during the study. (3) Results: Animals treated with CMNPs had a statistically significant functional outcome compared to the control group regarding: sciatic functional index, pain-like behavior, total body weight, which were confirmed by the histological and TEM images. (4) Conclusions: The results of the study suggest that CMNPs appear to be a promising treatment method for peripheral nerve injuries.
Collapse
Affiliation(s)
- Nadina Liana Pop
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street No. 1-3, 400006 Cluj-Napoca, Cluj County, Romania; (N.L.P.); (R.M.); (N.D.); (R.O.)
| | - Alexandrina Nan
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donath Street No. 67-103, 400293 Cluj-Napoca, Cluj County, Romania;
| | - Andrada Elena Urda-Cimpean
- Department of Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Pasteur Street No. 4-6, 400349 Cluj-Napoca, Cluj County, Romania;
| | - Adrian Florea
- Department of Cell and Molecular Biology, Iuliu Haţieganu University of Medicine and Pharmacy, Pasteur Street No. 4-6, 400349 Cluj-Napoca, Cluj County, Romania;
| | - Vlad Alexandru Toma
- Department of Molecular Biology and Biotechnologies, Babeș-Bolyai University, Clinicilor Street No. 4-6, 400000 Cluj-Napoca, Cluj County, Romania;
- Institute of Biological Research, Republicii Street No. 48, 400015 Cluj-Napoca, Cluj County, Romania
| | - Remus Moldovan
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street No. 1-3, 400006 Cluj-Napoca, Cluj County, Romania; (N.L.P.); (R.M.); (N.D.); (R.O.)
| | - Nicoleta Decea
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street No. 1-3, 400006 Cluj-Napoca, Cluj County, Romania; (N.L.P.); (R.M.); (N.D.); (R.O.)
| | - Daniela Rodica Mitrea
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street No. 1-3, 400006 Cluj-Napoca, Cluj County, Romania; (N.L.P.); (R.M.); (N.D.); (R.O.)
| | - Remus Orasan
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street No. 1-3, 400006 Cluj-Napoca, Cluj County, Romania; (N.L.P.); (R.M.); (N.D.); (R.O.)
| |
Collapse
|
32
|
Eskens O, Amin S. Challenges and effective routes for formulating and delivery of epidermal growth factors in skin care. Int J Cosmet Sci 2021; 43:123-130. [PMID: 33354795 DOI: 10.1111/ics.12685] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/10/2020] [Accepted: 12/18/2020] [Indexed: 01/08/2023]
Abstract
Epidermal growth factors are important morphogenetic proteins that instruct cell behaviour. They have many inferred applications within regenerative medicine and have shown promising results in skincare treatments. Many growth factors are known to have skin anti-ageing benefits along with further potential in resolving scarring, acne and other skin disorders. Incorporation of these biologics into skincare formulations has been greatly hindered by low transdermal delivery efficacy, intricate material interactions and protein instability - especially within common cosmetic emulsions. This review explores the cosmeceutical capability of growth factors in skin care, current understandings of constituent interactions and advantageous delivery approaches for more effective topical delivery. The first section highlights the influences growth factor product formulation has on stability and synergy. Current understandings of growth factor formulating techniques in cosmetic products is limited, and the performance of other protein structures is an adequate point of reference. The second section examines emerging drug delivery systems to overcome the challenges of topical growth factor treatment. It is important to consider the coaction and durability of all components in a formulation simultaneously: active ingredients, product format and delivery vehicle, in order to engineer an optimal cosmeceutical product.
Collapse
Affiliation(s)
- Olivia Eskens
- Chemical Engineering Department, Manhattan College, Riverdale, NY, USA
| | - Samiul Amin
- Chemical Engineering Department, Manhattan College, Riverdale, NY, USA
| |
Collapse
|
33
|
Sun M, Lee J, Chen Y, Hoshino K. Studies of nanoparticle delivery with in vitro bio-engineered microtissues. Bioact Mater 2020; 5:924-937. [PMID: 32637755 PMCID: PMC7330434 DOI: 10.1016/j.bioactmat.2020.06.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 01/04/2023] Open
Abstract
A variety of engineered nanoparticles, including lipid nanoparticles, polymer nanoparticles, gold nanoparticles, and biomimetic nanoparticles, have been studied as delivery vehicles for biomedical applications. When assessing the efficacy of a nanoparticle-based delivery system, in vitro testing with a model delivery system is crucial because it allows for real-time, in situ quantitative transport analysis, which is often difficult with in vivo animal models. The advent of tissue engineering has offered methods to create experimental models that can closely mimic the 3D microenvironment in the human body. This review paper overviews the types of nanoparticle vehicles, their application areas, and the design strategies to improve delivery efficiency, followed by the uses of engineered microtissues and methods of analysis. In particular, this review highlights studies on multicellular spheroids and other 3D tissue engineering approaches for cancer drug development. The use of bio-engineered tissues can potentially provide low-cost, high-throughput, and quantitative experimental platforms for the development of nanoparticle-based delivery systems.
Collapse
Affiliation(s)
- Mingze Sun
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Jinhyung Lee
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Kazunori Hoshino
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| |
Collapse
|
34
|
Kudo TA, Tominami K, Izumi S, Hayashi Y, Noguchi T, Matsuzawa A, Hong G, Nakai J. Characterization of PC12 Cell Subclones with Different Sensitivities to Programmed Thermal Stimulation. Int J Mol Sci 2020; 21:ijms21218356. [PMID: 33171774 PMCID: PMC7664380 DOI: 10.3390/ijms21218356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 11/16/2022] Open
Abstract
Neuritogenesis is the process underling nervous system regeneration; however, optimal extracellular signals that can promote neuronal regenerative activities require further investigation. Previously, we developed a novel method for inducing neuronal differentiation in rat PC12 cells using temperature-controlled repeated thermal stimulation (TRTS) with a heating plate. Based on neurogenic sensitivity to TRTS, PC12 cells were classified as either hyper- or hyposensitive. In this study, we aimed to investigate the mechanism of hyposensitivity by establishing two PC12-derived subclones according to TRTS sensitivity during differentiation: PC12-P1F1, a hypersensitive subclone, and PC12-P1D10, a hyposensitive subclone. To characterize these subclones, cell size and neuritogenesis were evaluated in subclones treated with nerve growth factor (NGF), bone morphogenetic protein (BMP), or various TRTS. No significant differences in cell size were observed among the parental cells and subclones. BMP4- or TRTS-induced neuritogenesis was increased in PC12-P1F1 cells compared to that in the parental cells, while no neuritogenesis was observed in PC12-P1D10 cells. In contrast, NGF-induced neuritogenesis was observed in all three cell lines. Furthermore, a BMP inhibitor, LDN-193189, considerably inhibited TRTS-induced neuritogenesis. These results suggest that the BMP pathway might be required for TRTS-induced neuritogenesis, demonstrating the useful aspects of these novel subclones for TRTS research.
Collapse
Affiliation(s)
- Tada-aki Kudo
- Division of Oral Physiology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; (K.T.); (S.I.); (J.N.)
- Correspondence: ; Tel./Fax: +81-22-717-8293
| | - Kanako Tominami
- Division of Oral Physiology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; (K.T.); (S.I.); (J.N.)
| | - Satoshi Izumi
- Division of Oral Physiology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; (K.T.); (S.I.); (J.N.)
| | - Yohei Hayashi
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan;
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (T.N.); (A.M.)
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (T.N.); (A.M.)
| | - Guang Hong
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan;
| | - Junichi Nakai
- Division of Oral Physiology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; (K.T.); (S.I.); (J.N.)
| |
Collapse
|
35
|
Mehrabi T, Mesgar AS, Mohammadi Z. Bioactive Glasses: A Promising Therapeutic Ion Release Strategy for Enhancing Wound Healing. ACS Biomater Sci Eng 2020; 6:5399-5430. [PMID: 33320556 DOI: 10.1021/acsbiomaterials.0c00528] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The morbidity, mortality, and burden of burn victims and patients with severe diabetic wounds are still high, which leads to an extensively growing demand for novel treatments with high clinical efficacy. Biomaterial-based wound treatment approaches have progressed over time from simple cotton wool dressings to advanced skin substitutes containing cells and growth factors; however, no wound care approach is yet completely satisfying. Bioactive glasses are materials with potential in many areas that exhibit unique features in biomedical applications. Today, bioactive glasses are not only amorphous solid structures that can be used as a substitute in hard tissue but also are promising materials for soft tissue regeneration and wound healing applications. Biologically active elements such as Ag, B, Ca, Ce, Co, Cu, Ga, Mg, Se, Sr, and Zn can be incorporated in glass networks; hence, the superiority of these multifunctional materials over current materials results from their ability to release multiple therapeutic ions in the wound environment, which target different stages of the wound healing process. Bioactive glasses and their dissolution products have high potency for inducing angiogenesis and exerting several biological impacts on cell functions, which are involved in wound healing and some other features that are valuable in wound healing applications, namely hemostatic and antibacterial properties. In this review, we focus on skin structure, the dynamic process of wound healing in injured skin, and existing wound care approaches. The basic concepts of bioactive glasses are reviewed to better understand the relationship between glass structure and its properties. We illustrate the active role of bioactive glasses in wound repair and regeneration. Finally, research studies that have used bioactive glasses in wound healing applications are summarized and the future trends in this field are elaborated.
Collapse
Affiliation(s)
- Tina Mehrabi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Abdorreza S Mesgar
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Zahra Mohammadi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| |
Collapse
|
36
|
Budama-Kilinc Y, Kecel-Gunduz S, Ozdemir B, Bicak B, Akman G, Arvas B, Aydogan F, Yolacan C. New nanodrug design for cancer therapy: Its synthesis, formulation, in vitro and in silico evaluations. Arch Pharm (Weinheim) 2020; 353:e2000137. [PMID: 32757360 DOI: 10.1002/ardp.202000137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/22/2020] [Accepted: 07/11/2020] [Indexed: 11/07/2022]
Abstract
The aim of this study was to develop a novel nanosize drug candidate for cancer therapy. For this purpose, (S)-methyl 2-[(7-hydroxy-2-oxo-4-phenyl-2H-chromen-8-yl)methyleneamino]-3-(1H-indol-3-yl)propanoate (ND3) was synthesized by the condensation reaction of 8-formyl-7-hydroxy-4-phenylcoumarin with l-tryptophan methyl ester. Its controlled release formulation was prepared and characterized by different spectroscopic and imaging methods. The cytotoxic effects of ND3 and its controlled release formulation were evaluated against MCF-7 and A549 cancer cell lines, and it was found that both of them have a toxic effect on cancer cells. For drug design and process development, the molecular docking analysis technique helps to clarify the effects of some DNA-targeted anticancer drugs to determine the interaction mechanisms of these drugs on DNA in a shorter time and at a lower cost. By using the molecular docking analysis and DNA binding assays, the interaction between the synthesized compound and DNA was elucidated and non-binding interactions were also determined. To predict the pharmacokinetics, and thereby accelerate drug discovery, the absorption, distribution, metabolism, excretion and toxicity values of the synthesized compound were determined by in silico methods.
Collapse
Affiliation(s)
- Yasemin Budama-Kilinc
- Department of Bioengineering, Yildiz Technical University, Davutpasa Campus, Istanbul, Turkey
| | - Serda Kecel-Gunduz
- Department of Physics, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Burak Ozdemir
- Department of Bioengineering, Yildiz Technical University, Davutpasa Campus, Istanbul, Turkey
| | - Bilge Bicak
- Department of Physics, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Gizem Akman
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Busra Arvas
- Department of Chemistry, Yildiz Technical University, Davutpasa Campus, Istanbul, Turkey
| | - Feray Aydogan
- Department of Chemistry, Yildiz Technical University, Davutpasa Campus, Istanbul, Turkey
| | - Cigdem Yolacan
- Department of Chemistry, Yildiz Technical University, Davutpasa Campus, Istanbul, Turkey
| |
Collapse
|
37
|
Abstract
INTRODUCTION Bioactive molecule carrier systems (BACS) are biomaterial-based substrates that facilitate the delivery of active signaling molecules for different biologically based therapeutic applications, which include regenerative endodontic procedures. Tissue regeneration or organized repair in regenerative endodontic procedures is governed by the dynamic orchestration of interactions between stem/progenitor cells, bioactive molecules, and extracellular matrix. BACS aid in mimicking some of the complex physiological processes, overcoming some of the challenges faced in the clinical translation of regenerative endodontic procedures. AREAS COVERED This narrative review addresses the role of BACS in stem/progenitor cell proliferation, migration, and differentiation with the application for dentin-pulp tissue engineering both in vitro and in vivo. BACS shield the bioactivity of the immobilized molecules against environmental factors, while its design allows the pre-programmed release of bioactive molecules in a spatial and temporal-controlled manner. The polymeric and non-polymeric materials used to synthesize micro and nanoscale-based BACS are reviewed. EXPERT OPINION Comprehensive characterization of well-designed and customized BACS is necessary to be able to deliver multiple bioactive molecules in spatiotemporally controlled manner and to address the release kinetics required for potential in vivo application. This warrants further laboratory-based experiments and rigorous clinical investigations to enable their clinical translation for regenerative endodontic procedures.
Collapse
Affiliation(s)
- Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto , Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto , Toronto, ON, Canada.,School of Graduate Studies, University of Toronto , Toronto, ON, Canada.,Department of Dentistry, Mount Sinai Health System, Mount Sinai Hospital , Toronto, ON, Canada
| | - Hebatullah Hussein
- The Kishen Lab, Dental Research Institute, University of Toronto , Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto , Toronto, ON, Canada
| |
Collapse
|
38
|
Thapa RK, Margolis DJ, Kiick KL, Sullivan MO. Enhanced wound healing via collagen-turnover-driven transfer of PDGF-BB gene in a murine wound model. ACS APPLIED BIO MATERIALS 2020; 3:3500-3517. [PMID: 32656505 DOI: 10.1021/acsabm.9b01147] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Wound healing is a complex biological process that requires coordinated cell proliferation, migration, and extracellular matrix production/remodeling, all of which are inhibited/delayed in chronic wounds. In this study, a formulation was developed that marries a fibrin-based, provisional-like matrix with collagen mimetic peptide (CMP)/PDGF gene-modified collagens, leading to the formation of robust gels that supported temporally controlled PDGF expression and facile application within the wound bed. Analysis employing in vitro co-gel scaffolds confirmed sustained and temporally controlled gene release based on matrix metalloproteinase (MMP) activity, with ~30% higher PDGF expression in MMP producing fibroblasts as-compared with non-MMP-expressing cells. The integration of fibrin with the gene-modified collagens resulted in co-gels that strongly supported both fibroblast cell recruitment/invasion as well as multiple aspects of the longer-term healing process. The excisional wound healing studies in mice established faster wound closure using CMP-modified PDGF polyplex-loaded co-gels, which exhibited up to 24% more wound closure (achieved with ~2 orders of magnitude lower growth factor dosing) after 9 days as compared to PDGF-loaded co-gels, and 19% more wound closure after 9 days as compared to CMP-free polyplex loaded co-gels. Moreover, minimal scar formation as well as improved collagen production, myofibroblast activity, and collagen orientation was observed following CMP-modified PDGF polyplex-loaded co-gel application on wounds. Taken together, the combined properties of the co-gels, including their stability and capacity to control both cell recruitment and cell phenotype within the murine wound bed, strongly supports the potential of the co-gel scaffolds for improved treatment of chronic non-healing wounds.
Collapse
Affiliation(s)
- Raj Kumar Thapa
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - David J Margolis
- Perelman School of Medicine, Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
39
|
Buie T, McCune J, Cosgriff-Hernandez E. Gelatin Matrices for Growth Factor Sequestration. Trends Biotechnol 2020; 38:546-557. [PMID: 31954527 PMCID: PMC11577444 DOI: 10.1016/j.tibtech.2019.12.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/15/2019] [Accepted: 12/06/2019] [Indexed: 01/07/2023]
Abstract
Gelatin is used in a broad range of tissue engineering applications because of its bioactivity, mild processing conditions, and ease of modification, which have increased interest in its use as a growth factor delivery vehicle. Traditional methods to control growth factor sequestration and delivery have relied on controlling hydrogel mesh size via chemical crosslinking with corollary changes to the physical properties of the hydrogel. To decouple growth factor release from scaffold properties, affinity sequestration modalities have been developed to preserve the bioactivity of the growth factor through interactions with the modified gelatin. This review provides a summary of these mechanisms, highlights current gelatin growth factor delivery systems, and addresses the future perspective of gelatin matrices for growth factor delivery in tissue engineering.
Collapse
Affiliation(s)
- Taneidra Buie
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Joshua McCune
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | | |
Collapse
|
40
|
Li S, Tang Q, Xu H, Huang Q, Wen Z, Liu Y, Peng C. Improved stability of KGF by conjugation with gold nanoparticles for diabetic wound therapy. Nanomedicine (Lond) 2019; 14:2909-2923. [PMID: 31791171 DOI: 10.2217/nnm-2018-0487] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: Diabetic wound healing is seriously interrupted, and administration of KGF for wound treatment is restricted by its inherent instability. We aim to develop an ideal way toward KGF stabilization, thus improving diabetic wound healing. Materials & methods: We conjugated KGF with gold nanoparticles (GNPs) and determined the stability and binding affinity. Biological effects of conjugates (KGF-GNPs) were evaluated in vitro and in an animal model. Results: KGF-GNPs revealed high stability under hostile circumstances because of the preserved secondary structure and possessed elevated binding affinity to KGF receptor. Moreover, application of KGF-GNPs contributed to accelerated wound recovery in diabetic rats, including re-epithelialization and contraction. Conclusion: KGF-GNPs were promising for future clinical application for diabetic wound therapy.
Collapse
Affiliation(s)
- Shuaihua Li
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China.,Department of Cosmetic & Plastic Surgery, The First People's Hospital of Chenzhou, Chenzhou 423000, Hunan, PR China
| | - Qiyu Tang
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China.,Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, PR China
| | - Hongbo Xu
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| | - Qiangru Huang
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| | - Zi Wen
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| | - Yawei Liu
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| | - Cheng Peng
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, PR China
| |
Collapse
|
41
|
Polley P, Gupta S, Singh R, Pradhan A, Basu SM, V. R, Yadava SK, Giri J. Protein–Sugar-Glass Nanoparticle Platform for the Development of Sustained-Release Protein Depots by Overcoming Protein Delivery Challenges. Mol Pharm 2019; 17:284-300. [DOI: 10.1021/acs.molpharmaceut.9b01022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Poulomi Polley
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Shivam Gupta
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Ruby Singh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Arpan Pradhan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, Maharashtra, India
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Remya V.
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Sunil Kumar Yadava
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| |
Collapse
|
42
|
Qi H, Yang L, Li X, Sun X, Zhao J, Hou X, Li Z, Yuan X, Cui Z, Yang X. Systemic administration of enzyme-responsive growth factor nanocapsules for promoting bone repair. Biomater Sci 2019; 7:1675-1685. [PMID: 30742145 DOI: 10.1039/c8bm01632a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Accelerating the healing of bone fractures by local delivery of growth factors possessing osteoinductive activity has been extensively demonstrated. Unfortunately, for some complex clinical fractures, such as osteoporotic vertebral compression fracture, it is not possible to adopt such a strategy because of access restrictions. Systemic administration of growth factors is considered to be an appropriate alternative method due to its easy operability and precise spatiotemporal compatibility at fracture sites. But this therapy method was hampered by the poor in vivo stability, inefficient distribution at the fracture site and potential side effects of growth factors. To address these challenges, we conceived a systemic delivery platform of growth factors based on nanocapsules, taking advantage of the unique physiological character of bone fracture, i.e., the malformed blood vessels and the over-expression of matrix metalloproteinases (MMPs). In this work, bone morphogenetic protein-2 (BMP-2), 2-(methacryloyloxy)ethyl phosphorylcholine (MPC) and the bisacryloylated VPLGVRTK peptide were respectively used as the model growth factor, monomer, and MMP-cleavable crosslinker. Nanocapsules were formed by in situ free radical polymerization on the surface of BMP-2 with MPC and peptides. The structure and function of BMP-2 were well maintained during the preparation process. BMP-2 nanocapsules (n(BMP-2)) were of uniform small size (∼30 nm) possessing a long circulation time (half-life is ∼48 h) and could be passively targeted to the fracture site through malformed blood vessels after systemic administration. Once accumulated at the fracture site, the shells of nanocapsules could be degraded by MMP and thus BMP-2 was released. Animal experiments proved that n(BMP-2) showed a better ability of bone repair than native BMP-2. In addition, n(BMP-2) showed a much lower inflammatory irritation. The results demonstrated that the systemic administration of growth factor nanocapsules could enhance their in vivo stability and fracture site delivery efficiency, realizing the efficient repair of a bone fracture. This rational delivery system may expand the bone repair types which can be administered with growth factors. Furthermore, the concept of taking advantage of the malformed vascular structure to deliver drugs potentially inspires researchers for the therapy of other diseases, especially sudden disease (such as cerebral hemorrhage).
Collapse
Affiliation(s)
- Hongzhao Qi
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mabrouk M, Rajendran R, Soliman IE, Ashour MM, Beherei HH, Tohamy KM, Thomas S, Kalarikkal N, Arthanareeswaran G, Das DB. Nanoparticle- and Nanoporous-Membrane-Mediated Delivery of Therapeutics. Pharmaceutics 2019; 11:E294. [PMID: 31234394 PMCID: PMC6631283 DOI: 10.3390/pharmaceutics11060294] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Pharmaceutical particulates and membranes possess promising prospects for delivering drugs and bioactive molecules with the potential to improve drug delivery strategies like sustained and controlled release. For example, inorganic-based nanoparticles such as silica-, titanium-, zirconia-, calcium-, and carbon-based nanomaterials with dimensions smaller than 100 nm have been extensively developed for biomedical applications. Furthermore, inorganic nanoparticles possess magnetic, optical, and electrical properties, which make them suitable for various therapeutic applications including targeting, diagnosis, and drug delivery. Their properties may also be tuned by controlling different parameters, e.g., particle size, shape, surface functionalization, and interactions among them. In a similar fashion, membranes have several functions which are useful in sensing, sorting, imaging, separating, and releasing bioactive or drug molecules. Engineered membranes have been developed for their usage in controlled drug delivery devices. The latest advancement in the technology is therefore made possible to regulate the physico-chemical properties of the membrane pores, which enables the control of drug delivery. The current review aims to highlight the role of both pharmaceutical particulates and membranes over the last fifteen years based on their preparation method, size, shape, surface functionalization, and drug delivery potential.
Collapse
Affiliation(s)
- Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St (former EL Tahrirst)-Dokki, Giza 12622, Egypt.
| | - Rajakumari Rajendran
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560, India.
| | - Islam E Soliman
- Biophysics Branch, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt.
| | | | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St (former EL Tahrirst)-Dokki, Giza 12622, Egypt.
| | - Khairy M Tohamy
- Biophysics Branch, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt.
| | - Sabu Thomas
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560, India.
| | - Nandakumar Kalarikkal
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560, India.
| | | | - Diganta B Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK.
| |
Collapse
|
44
|
Controlling the Release Profile Through Phase Control of Calcium Phosphate-Alginate Core-shell Nanoparticles in Gene Delivery. Macromol Res 2019. [DOI: 10.1007/s13233-019-7106-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
45
|
Yamakawa S, Hayashida K. Advances in surgical applications of growth factors for wound healing. BURNS & TRAUMA 2019; 7:10. [PMID: 30993143 PMCID: PMC6450003 DOI: 10.1186/s41038-019-0148-1] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/13/2019] [Indexed: 12/15/2022]
Abstract
Growth factors have recently gained clinical importance for wound management. Application of recombinant growth factors has been shown to mimic cell migration, proliferation, and differentiation in vivo, allowing for external modulation of the healing process. Perioperative drug delivery systems can enhance the biological activity of these growth factors, which have a very short in vivo half-life after topical administration. Although the basic mechanisms of these growth factors are well understood, most have yet to demonstrate a significant impact in animal studies or small-sized clinical trials. In this review, we emphasized currently approved growth factor therapies, including a sustained release system for growth factors, emerging therapies, and future research possibilities combined with surgical procedures. Approaches seeking to understand wound healing at a systemic level are currently ongoing. However, further research and consideration in surgery will be needed to provide definitive confirmation of the efficacy of growth factor therapies for intractable wounds.
Collapse
Affiliation(s)
- Sho Yamakawa
- Division of Plastic and Reconstructive Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501 Japan
| | - Kenji Hayashida
- Division of Plastic and Reconstructive Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501 Japan
| |
Collapse
|
46
|
Subbiah R, Guldberg RE. Materials Science and Design Principles of Growth Factor Delivery Systems in Tissue Engineering and Regenerative Medicine. Adv Healthc Mater 2019; 8:e1801000. [PMID: 30398700 DOI: 10.1002/adhm.201801000] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/13/2018] [Indexed: 01/22/2023]
Abstract
Growth factors (GFs) are signaling molecules that direct cell development by providing biochemical cues for stem cell proliferation, migration, and differentiation. GFs play a key role in tissue regeneration, but one major limitation of GF-based therapies is dosage-related adverse effects. Additionally, the clinical applications and efficacy of GFs are significantly affected by the efficiency of delivery systems and other pharmacokinetic factors. Hence, it is crucial to design delivery systems that provide optimal activity, stability, and tunable delivery for GFs. Understanding the physicochemical properties of the GFs and the biomaterials utilized for the development of biomimetic GF delivery systems is critical for GF-based regeneration. Many different delivery systems have been developed to achieve tunable delivery kinetics for single or multiple GFs. The identification of ideal biomaterials with tunable properties for spatiotemporal delivery of GFs is still challenging. This review characterizes the types, properties, and functions of GFs, the materials science of widely used biomaterials, and various GF loading strategies to comprehensively summarize the current delivery systems for tunable spatiotemporal delivery of GFs aimed for tissue regeneration applications. This review concludes by discussing fundamental design principles for GF delivery vehicles based on the interactive physicochemical properties of the proteins and biomaterials.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact; 6231 University of Oregon; Eugene OR 97403 USA
| |
Collapse
|
47
|
Rasouli R, Barhoum A, Uludag H. A review of nanostructured surfaces and materials for dental implants: surface coating, patterning and functionalization for improved performance. Biomater Sci 2018; 6:1312-1338. [PMID: 29744496 DOI: 10.1039/c8bm00021b] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emerging field of nanostructured implants has enormous scope in the areas of medical science and dental implants. Surface nanofeatures provide significant potential solutions to medical problems by the introduction of better biomaterials, improved implant design, and surface engineering techniques such as coating, patterning, functionalization and molecular grafting at the nanoscale. This review is of an interdisciplinary nature, addressing the history and development of dental implants and the emerging area of nanotechnology in dental implants. After a brief introduction to nanotechnology in dental implants and the main classes of dental implants, an overview of different types of nanomaterials (i.e. metals, metal oxides, ceramics, polymers and hydrides) used in dental implant together with their unique properties, the influence of elemental compositions, and surface morphologies and possible applications are presented from a chemical point of view. In the core of this review, the dental implant materials, physical and chemical fabrication techniques and the role of nanotechnology in achieving ideal dental implants have been discussed. Finally, the critical parameters in dental implant design and available data on the current dental implant surfaces that use nanotopography in clinical dentistry have been discussed.
Collapse
Affiliation(s)
- Rahimeh Rasouli
- Department of Medical Nanotechnology, International Campus, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
48
|
Rahman SU, Nagrath M, Ponnusamy S, Arany PR. Nanoscale and Macroscale Scaffolds with Controlled-Release Polymeric Systems for Dental Craniomaxillofacial Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E1478. [PMID: 30127246 PMCID: PMC6120038 DOI: 10.3390/ma11081478] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Tremendous progress in stem cell biology has resulted in a major current focus on effective modalities to promote directed cellular behavior for clinical therapy. The fundamental principles of tissue engineering are aimed at providing soluble and insoluble biological cues to promote these directed biological responses. Better understanding of extracellular matrix functions is ensuring optimal adhesive substrates to promote cell mobility and a suitable physical niche to direct stem cell responses. Further, appreciation of the roles of matrix constituents as morphogen cues, termed matrikines or matricryptins, are also now being directly exploited in biomaterial design. These insoluble topological cues can be presented at both micro- and nanoscales with specific fabrication techniques. Progress in development and molecular biology has described key roles for a range of biological molecules, such as proteins, lipids, and nucleic acids, to serve as morphogens promoting directed behavior in stem cells. Controlled-release systems involving encapsulation of bioactive agents within polymeric carriers are enabling utilization of soluble cues. Using our efforts at dental craniofacial tissue engineering, this narrative review focuses on outlining specific biomaterial fabrication techniques, such as electrospinning, gas foaming, and 3D printing used in combination with polymeric nano- or microspheres. These avenues are providing unprecedented therapeutic opportunities for precision bioengineering for regenerative applications.
Collapse
Affiliation(s)
- Saeed Ur Rahman
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan.
| | - Malvika Nagrath
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Department of Biomedical Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada.
| | - Sasikumar Ponnusamy
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| | - Praveen R Arany
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
49
|
Young DA, Pimentel MB, Lima LD, Custodio AF, Lo WC, Chen SC, Teymour F, Papavasiliou G. Design and characterization of hydrogel nanoparticles with tunable network characteristics for sustained release of a VEGF-mimetic peptide. Biomater Sci 2018; 5:2079-2092. [PMID: 28744527 DOI: 10.1039/c7bm00359e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Peptides that mimic the bioactivity of growth factors are rapidly emerging as therapeutics for a variety of drug delivery applications including therapeutic neovascularization. Neovascularization requires controlled and sustained delivery of proangiogenic factors to stimulate reperfusion of ischemic tissues. To this end, hydrogel nanoparticles were designed to provide sustained and tunable diffusion-based release of a pro-angiogenic peptide, QK. Inverse phase mini-emulsion polymerization (IPMP) was used to generate crosslinked poly(ethylene) glycol (PEG) hydrogel nanoparticles entrapped with the QK peptide. Peptide release kinetics were tuned through adjustments in nanoparticle crosslink density. This was achieved by altering the mole fraction of the crosslinking agent which allowed for the synthesis of low crosslink density (0.754 mmol cm-3) and high crosslink density (0.810 mmol cm-3) nanoparticles. Nanoparticle tracking analysis revealed narrow particle size distributions and similar particle sizes regardless of crosslink density (225 ± 75 nm and 233 ± 73 nm, for low and high crosslink density nanoparticles, respectively). The zeta potential was found to be -26 mV for blank nanoparticles and +4 mV in the case of QK-loaded nanoparticles. The resulting nanoparticle crosslink density impacted both peptide loading as well as release kinetics. In terms of cumulative fractional release and weight of peptide released per mass of nanoparticle, higher crosslink density nanoparticles resulted in slower peptide release kinetics. The IPMP process preserved the QK secondary structure and its bioactivity as confirmed using circular dichroism spectroscopy and a Matrigel tubulogenesis assay, respectively, with released peptide. The presented nanoparticles hold great potential for use as drug delivery carriers for stimulation of therapeutic neovascularization of ischemic tissues.
Collapse
Affiliation(s)
- Daniel A Young
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Shamloo A, Sarmadi M, Aghababaie Z, Vossoughi M. Accelerated full-thickness wound healing via sustained bFGF delivery based on a PVA/chitosan/gelatin hydrogel incorporating PCL microspheres. Int J Pharm 2018; 537:278-289. [DOI: 10.1016/j.ijpharm.2017.12.045] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/25/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022]
|