1
|
L J, Kamaraj S, Kandasamy R, Alagarsamy S. Electrospinning: A New Frontier in Peptide Therapeutics. AAPS PharmSciTech 2025; 26:69. [PMID: 40011310 DOI: 10.1208/s12249-025-03054-2] [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: 09/29/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
The nanofiber technology has recently undergone an unprecedented transformation, finding widespread utilities across diverse scientific disciplines. It is noteworthy that electrospinning approaches have emerged as an adaptable and successful approach to generate fibers ranging in rapidly as a class of therapeutic agents with a high level of target specificity. Peptides encounter several challenges as drugs, including swift breakdown by the body, rapid elimination from the bloodstream, inadequate stability, and restricted ability to cross cell membranes. This renders it challenging to employ them as drugs. However, electrospun nanofibers might address these problems. This review explores the promising potential of electrospinning nanofibers for peptide delivery. We delve into recent advancements in this technique, highlighting its effectiveness in overcoming challenges associated with peptide drug delivery. It provides an analysis of the trends identified in the use of the electrospinning technique and its role in peptide drug delivery systems, based on a review of data collected over a period of five to seven years.
Collapse
Affiliation(s)
- Jeyanthi L
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Sivadharshini Kamaraj
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Ruckmani Kandasamy
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Shanmugarathinam Alagarsamy
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|
2
|
Mullin JA, Rahmani E, Kiick KL, Sullivan MO. Growth factors and growth factor gene therapies for treating chronic wounds. Bioeng Transl Med 2024; 9:e10642. [PMID: 38818118 PMCID: PMC11135157 DOI: 10.1002/btm2.10642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Chronic wounds are an unmet clinical need affecting millions of patients globally, and current standards of care fail to consistently promote complete wound closure and prevent recurrence. Disruptions in growth factor signaling, a hallmark of chronic wounds, have led researchers to pursue growth factor therapies as potential supplements to standards of care. Initial studies delivering growth factors in protein form showed promise, with a few formulations reaching clinical trials and one obtaining clinical approval. However, protein-form growth factors are limited by instability and off-target effects. Gene therapy offers an alternative approach to deliver growth factors to the chronic wound environment, but safety concerns surrounding gene therapy as well as efficacy challenges in the gene delivery process have prevented clinical translation. Current growth factor delivery and gene therapy approaches have primarily used single growth factor formulations, but recent efforts have aimed to develop multi-growth factor approaches that are better suited to address growth factor insufficiencies in the chronic wound environment, and these strategies have demonstrated improved efficacy in preclinical studies. This review provides an overview of chronic wound healing, emphasizing the need and potential for growth factor therapies. It includes a summary of current standards of care, recent advances in growth factor, cell-based, and gene therapy approaches, and future perspectives for multi-growth factor therapeutics.
Collapse
Affiliation(s)
- James A. Mullin
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Erfan Rahmani
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Kristi L. Kiick
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Materials Science and EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| |
Collapse
|
3
|
Sharda D, Kaur P, Choudhury D. Protein-modified nanomaterials: emerging trends in skin wound healing. DISCOVER NANO 2023; 18:127. [PMID: 37843732 PMCID: PMC10579214 DOI: 10.1186/s11671-023-03903-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/23/2023] [Indexed: 10/17/2023]
Abstract
Prolonged inflammation can impede wound healing, which is regulated by several proteins and cytokines, including IL-4, IL-10, IL-13, and TGF-β. Concentration-dependent effects of these molecules at the target site have been investigated by researchers to develop them as wound-healing agents by regulating signaling strength. Nanotechnology has provided a promising approach to achieve tissue-targeted delivery and increased effective concentration by developing protein-functionalized nanoparticles with growth factors (EGF, IGF, FGF, PDGF, TGF-β, TNF-α, and VEGF), antidiabetic wound-healing agents (insulin), and extracellular proteins (keratin, heparin, and silk fibroin). These molecules play critical roles in promoting cell proliferation, migration, ECM production, angiogenesis, and inflammation regulation. Therefore, protein-functionalized nanoparticles have emerged as a potential strategy for improving wound healing in delayed or impaired healing cases. This review summarizes the preparation and applications of these nanoparticles for normal or diabetic wound healing and highlights their potential to enhance wound healing.
Collapse
Affiliation(s)
- Deepinder Sharda
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Pawandeep Kaur
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Thapar Institute of Engineering and Technology-Virginia Tech Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
4
|
Amnieh YA, Ghadirian S, Mohammadi N, Shadkhast M, Karbasi S. Evaluation of the effects of chitosan nanoparticles on polyhydroxy butyrate electrospun scaffolds for cartilage tissue engineering applications. Int J Biol Macromol 2023; 249:126064. [PMID: 37524286 DOI: 10.1016/j.ijbiomac.2023.126064] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
In this study, we synthesized and incorporated chitosan nanoparticles (Cs) into polyhydroxy butyrate (PHB) electrospun scaffolds for cartilage tissue engineering. The Cs nanoparticles were synthesized via an ionic gel interaction between Cs powder and tripolyphosphate (TPP). The mechanical properties, hydrophilicity, and fiber diameter of the PHB scaffolds with varying concentrations of Cs nanoparticles (1-5 wt%) were evaluated. The results of these evaluations showed that the scaffold containing 1 wt% Cs nanoparticles (P1Cs) was the optimum scaffold, with increased ultimate strength from 2.6 to 5.2 MPa and elongation at break from 5.31 % to 12.6 %. Crystallinity, degradation, and cell compatibility were also evaluated. The addition of Cs nanoparticles decreased crystallinity and accelerated hydrolytic degradation. MTT assay results showed that the proliferation of chondrocytes on the scaffold containing 1 wt% Cs nanoparticles were significantly higher than that on pure PHB after 7 days of cultivation. These findings suggest that the electrospun P1Cs scaffold has promising potential as a substrate for cartilage tissue engineering applications. This combination offers a promising approach for the fabrication of biomimetic scaffolds with enhanced mechanical properties, hydrophilicity, and cell compatibility for tissue engineering applications.
Collapse
Affiliation(s)
- Yasamin Alikhasi Amnieh
- Department of Veterinary Histology, School of Veterinary, Shahrekord University, Shahrekord, Iran
| | - Sepideh Ghadirian
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nayereh Mohammadi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Shadkhast
- Basic Science of Veterinary Faculty, Shahrekord University, Shahrekord, Iran
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Dental Implants Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
Born LJ, Bengali S, Hsu ATW, Abadchi SN, Chang KH, Lay F, Matsangos A, Johnson C, Jay SM, Harmon JW. Chitosan Particles Complexed with CA5-HIF-1α Plasmids Increase Angiogenesis and Improve Wound Healing. Int J Mol Sci 2023; 24:14095. [PMID: 37762397 PMCID: PMC10531456 DOI: 10.3390/ijms241814095] [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: 08/18/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Wound therapies involving gene delivery to the skin have significant potential due to the advantage and ease of local treatment. However, choosing the appropriate vector to enable successful gene expression while also ensuring that the treatment's immediate material components are conducive to healing itself is critical. In this study, we utilized a particulate formulation of the polymer chitosan (chitosan particles, CPs) as a non-viral vector for the delivery of a plasmid encoding human CA5-HIF-1α, a degradation resistant form of HIF-1α, to enhance wound healing. We also compared the angiogenic potential of our treatment (HIF/CPs) to that of chitosan particles containing only the plasmid backbone (bb/CPs) and the chitosan particle vector alone (CPs). Our results indicate that chitosan particles exert angiogenic effects that are enhanced with the human CA5-HIF-1α-encoded plasmid. Moreover, HIF/CPs enhanced wound healing in diabetic db/db mice (p < 0.01), and healed tissue was found to contain a significantly increased number of blood vessels compared to bb/CPs (p < 0.01), CPs (p < 0.05) and no-treatment groups (p < 0.01). Thus, this study represents a method of gene delivery to the skin that utilizes an inherently pro-wound-healing polymer as a vector for plasmid DNA that has broad application for the expression of other therapeutic genes.
Collapse
Affiliation(s)
- Louis J. Born
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sameer Bengali
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Angela Ting Wei Hsu
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sanaz Nourmohammadi Abadchi
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kai-Hua Chang
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Frank Lay
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aerielle Matsangos
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher Johnson
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Steven M. Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA
| | - John W. Harmon
- Hendrix Burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Rybak D, Su YC, Li Y, Ding B, Lv X, Li Z, Yeh YC, Nakielski P, Rinoldi C, Pierini F, Dodda JM. Evolution of nanostructured skin patches towards multifunctional wearable platforms for biomedical applications. NANOSCALE 2023; 15:8044-8083. [PMID: 37070933 DOI: 10.1039/d3nr00807j] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Recent advances in the field of skin patches have promoted the development of wearable and implantable bioelectronics for long-term, continuous healthcare management and targeted therapy. However, the design of electronic skin (e-skin) patches with stretchable components is still challenging and requires an in-depth understanding of the skin-attachable substrate layer, functional biomaterials and advanced self-powered electronics. In this comprehensive review, we present the evolution of skin patches from functional nanostructured materials to multi-functional and stimuli-responsive patches towards flexible substrates and emerging biomaterials for e-skin patches, including the material selection, structure design and promising applications. Stretchable sensors and self-powered e-skin patches are also discussed, ranging from electrical stimulation for clinical procedures to continuous health monitoring and integrated systems for comprehensive healthcare management. Moreover, an integrated energy harvester with bioelectronics enables the fabrication of self-powered electronic skin patches, which can effectively solve the energy supply and overcome the drawbacks induced by bulky battery-driven devices. However, to realize the full potential offered by these advancements, several challenges must be addressed for next-generation e-skin patches. Finally, future opportunities and positive outlooks are presented on the future directions of bioelectronics. It is believed that innovative material design, structure engineering, and in-depth study of fundamental principles can foster the rapid evolution of electronic skin patches, and eventually enable self-powered close-looped bioelectronic systems to benefit mankind.
Collapse
Affiliation(s)
- Daniel Rybak
- Institute of Fundamental Technological Research, Polish Academy of Science, 02-106 Warsaw, Poland.
| | - Yu-Chia Su
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Yang Li
- College of Electronic and Optical Engineering & College of Microelectronics, Institute of Flexible Electronics (Future Technology), Nanjing University of Posts & Telecommunications (NJUPT), Nanjing, 210023, China
| | - Bin Ding
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, China.
| | - Xiaoshuang Lv
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Zhaoling Li
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Yi-Cheun Yeh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Pawel Nakielski
- Institute of Fundamental Technological Research, Polish Academy of Science, 02-106 Warsaw, Poland.
| | - Chiara Rinoldi
- Institute of Fundamental Technological Research, Polish Academy of Science, 02-106 Warsaw, Poland.
| | - Filippo Pierini
- Institute of Fundamental Technological Research, Polish Academy of Science, 02-106 Warsaw, Poland.
| | - Jagan Mohan Dodda
- New Technologies - Research Centre (NTC), University of West Bohemia, Univerzitní 8, 301 00 Pilsen, Czech Republic.
| |
Collapse
|
7
|
Tang X, Yang F, Chu G, Li X, Fu Q, Zou M, Zhao P, Lu G. Characterizing the inherent activity of urinary bladder matrix for adhesion, migration, and activation of fibroblasts as compared with collagen-based synthetic scaffold. J Biomater Appl 2023; 37:1446-1457. [PMID: 36177498 DOI: 10.1177/08853282221130883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The mechanism of action underlying the intriguing prominent bioactivity of urinary bladder matrix (UBM) for in situ tissue regeneration of soft tissue defects remains to be elucidated. It is speculated that the activity of UBM for cell adhesion, migration, and activation is inherent. The bioactivity of UBM for in situ tissue regeneration and its relation with the structure and intact soluble components of UBM were investigated in comparison to a collagen-based scaffold, PELNAC (PEL). We isolated the soluble component of the two materials with urea buffer, and evaluated the respective effect of these soluble components on the in vitro adhesion and migration of L929 fibroblasts. The spatiotemporal pattern of endogenous-cell ingrowth into the scaffolds and cell activation were investigated using a model of murine subcutaneous implantation. UBM is more capable of promoting the adhesion, migration, and proliferation of fibroblasts than PEL in a serum-independent manner. In vivo, as compared with PEL, UBM exhibits significantly enhanced activity for fast endogenous cell ingrowth and produces a more prominent pro-regenerative and pro-remodeling microenvironment by inducing the expression of TGF-β1, VEGF, MMP-9, and murine type I collagen. Overall, our results suggest the prominent bioactivity of UBM for in situ tissue regeneration is inherent.
Collapse
Affiliation(s)
- Xiaoyu Tang
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Guoping Chu
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoxiao Li
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiuyan Fu
- 66374Jiangnan University, Wuxi, China
| | - Mingli Zou
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Zhao
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Guozhong Lu
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
8
|
González-Torres M, Vargas-Muñoz S, Leyva-Gómez G, Méndez-Padilla MG, Cortés H, Nuñez-Rojas E, González-Mendoza O, Pérez-Díaz MA, Ruvalcaba-Paredes EK, Lima E, Brena AM, Rodríguez-Talavera R, Pineda C. Discovering the effect of solvents on poly(2-aminoethyl methacrylate) grafting onto chitosan for an in vitro skin model. Carbohydr Polym 2022; 295:119864. [DOI: 10.1016/j.carbpol.2022.119864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022]
|
9
|
Iliou K, Kikionis S, Ioannou E, Roussis V. Marine Biopolymers as Bioactive Functional Ingredients of Electrospun Nanofibrous Scaffolds for Biomedical Applications. Mar Drugs 2022; 20:md20050314. [PMID: 35621965 PMCID: PMC9143254 DOI: 10.3390/md20050314] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 02/01/2023] Open
Abstract
Marine biopolymers, abundantly present in seaweeds and marine animals, feature diverse structures and functionalities, and possess a wide range of beneficial biological activities. Characterized by high biocompatibility and biodegradability, as well as unique physicochemical properties, marine biopolymers are attracting a constantly increasing interest for the development of advanced systems for applications in the biomedical field. The development of electrospinning offers an innovative technological platform for the production of nonwoven nanofibrous scaffolds with increased surface area, high encapsulation efficacy, intrinsic interconnectivity, and structural analogy to the natural extracellular matrix. Marine biopolymer-based electrospun nanofibrous scaffolds with multifunctional characteristics and tunable mechanical properties now attract significant attention for biomedical applications, such as tissue engineering, drug delivery, and wound healing. The present review, covering the literature up to the end of 2021, highlights the advancements in the development of marine biopolymer-based electrospun nanofibers for their utilization as cell proliferation scaffolds, bioadhesives, release modifiers, and wound dressings.
Collapse
|
10
|
Liu C, Wang C, Yang F, Lu Y, Du P, Hu K, Yin X, Zhao P, Lu G. The conditioned medium from mesenchymal stromal cells pretreated with proinflammatory cytokines promote fibroblasts migration and activation. PLoS One 2022; 17:e0265049. [PMID: 35404961 PMCID: PMC9000110 DOI: 10.1371/journal.pone.0265049] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/22/2022] [Indexed: 11/19/2022] Open
Abstract
Human dermal fibroblasts (HDFs) play important roles in all stages of wound healing. However, in nonhealing wounds, fibroblasts are prone to aging, resulting in insufficient migration, proliferation and secretion functions. Recent studies have suggested that mesenchymal stromal cells (MSCs) are conducive to wound healing and cell growth through paracrine cytokine signaling. In our studies, we found that conditioned medium of MSCs pretreated with IFN-γ and TNF-α (IT MSC-CM) has abundant growth factors associated with wound repair. Our in vitro results showed that the effects of IT MSC-CM on promoting cell migration, proliferation and activation in HDFs were better than those of conditioned medium from mesenchymal stromal cells (MSC-CM). Moreover, we embedded a scaffold material containing IT MSC-CM and reconfirmed that cell migration and activation were superior to that in the presence of MSC-CM in vivo. Generally, PDGF-BB is perceived as a promoter of the migration and proliferation of HDFs. Moreover, a high level of PDGF-BB in IT MSC-CM was detected, according to which we guess that the effect on HDFs may be mediated by the upregulation of PDGF-BB. These studies all showed the potential of IT MSC-CM to promote rapid and effective wound healing.
Collapse
Affiliation(s)
- Chenyang Liu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China
| | - Chengchun Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Pan Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Kai Hu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China
| | - Xinyao Yin
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Peng Zhao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
- * E-mail: (GL); (PZ)
| | - Guozhong Lu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
- * E-mail: (GL); (PZ)
| |
Collapse
|
11
|
Alturki AM. Rationally design of electrospun polysaccharides polymeric nanofiber webs by various tools for biomedical applications: A review. Int J Biol Macromol 2021; 184:648-665. [PMID: 34102239 DOI: 10.1016/j.ijbiomac.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/30/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Nanofibers have a particular benefit when delivering a spectrum of therapeutic drugs for diverse biomedical applications. Nanofibers are easily fabricated from cellulose acetate, chitosan, polycaprolactone, and other polymers with regulated morphology and release profiles due to nanotechnology's recent advancement. This review will provide the latest approaches to the fabrication of electrospun nanofibers containing herbal extracts, antimicrobial peptides, and antibiotics for wound-healing potential. Besides, synthesis and evaluation of nanofibrous mats, including conducting polymer and evaluate their possibility for wound healing. In addition, nanofibers are loaded with some drugs for skin cancer treatment and contain growth factors for tissue regeneration. Also, the current two-dimensional nanofibers limitations and the various techniques for convert two-dimensional to three-dimension nanofibers to avoid these drawbacks. Moreover, the future direction in improving the three-dimensional structure and functionality has been including.
Collapse
Affiliation(s)
- Asma M Alturki
- Department of Chemistry, Faculty of Science, University of Tabuk, Saudi Arabia.
| |
Collapse
|
12
|
Quantitative measurement of Bax and Bcl2 genes and protein expression in MCF7 cell-line when treated by Aloe Vera extract. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
The platelet derived growth factor BB promotes osteogenic differentiation of periodontal ligament stem cells via the Wnt/β-catenin signaling pathway. Arch Oral Biol 2021; 129:105162. [PMID: 34087681 DOI: 10.1016/j.archoralbio.2021.105162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To determine the role of platelet derived growth factor BB in the regulation of cell cycle, migration and differentiation of stem cells. DESIGN The gene was overexpressed in periodontal ligament stem cells using lentiviral vectors. Normal stem cells and empty lentiviral vectors-transfected were used as controls. Real time-PCR, western blotting, Cell Counting Kit-8 assay, flow cytometry, cell scratch test, Alkaline phosphatase activity assay, cell cycle analyses were conducted to assess the biological properties of stem cells. In addition, the effect of platelet derived growth factor BB on the Wnt/β-catenin signaling pathway were assessed by western blotting and immunofluorescent staining. RESULTS The gene was successfully overexpressed in periodontal ligament stem cells. The Cell Counting Kit-8, scratch test and cell cycle experiments proved that platelet derived growth factor BB promoted stem cells proliferation, migration and cell cycle progression. The Real time-PCR results showed that the Osterix (OSX) and Bone Morphogenetic Protein 2 (BMP2) genes in the overexpression group were significantly higher than those in the control group, but the Peroxisome Proliferators-activated Receptors (PPARγ) and Glycogen Synthase Kinase-3β (GSK-3β) gene were lower than that in the control group. Western blotting results also indicated that the Collagen Type 1 (COL-1), BMP2, Wnt1 and β-catenin proteins were increased in the overexpression group. In addition, the expression level of β-catenin protein in the cell nuclei was higher than that in the control group. CONCLUSIONS In conclusion, overexpression of platelet derived growth factor BB promoted cell proliferation, migration, cell cycle progression and decreased adipogenic differentiation. Furthermore, platelet derived growth factor BB regulated osteogenic differentiation of stem cells through the Wnt/β-catenin signaling pathway.
Collapse
|
14
|
Catanzano O, Quaglia F, Boateng JS. Wound dressings as growth factor delivery platforms for chronic wound healing. Expert Opin Drug Deliv 2021; 18:737-759. [PMID: 33338386 DOI: 10.1080/17425247.2021.1867096] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Years of tissue engineering research have clearly demonstrated the potential of integrating growth factors (GFs) into scaffolds for tissue regeneration, a concept that has recently been applied to wound dressings. The old concept of wound dressings that only take a passive role in wound healing has now been overtaken, and advanced dressings which can take an active part in wound healing, are of current research interest.Areas covered: In this review we will focus on the recent strategies for the delivery of GFs to wound sites with an emphasis on the different approaches used to achieve fine tuning of spatial and temporal concentrations to achieve therapeutic efficacy.Expert opinion: The use of GFs to accelerate wound healing and reduce scar formation is now considered a feasible therapeutic approach in patients with a high risk of infections and complications. The integration of micro - and nanotechnologies into wound dressings could be the key to overcome the inherent instability of GFs and offer adequate control over the release rate. Many investigations have led to encouraging outcomes in various in vitro and in vivo wound models, and it is expected that some of these technologies will satisfy clinical needs and will enter commercialization.
Collapse
Affiliation(s)
- Ovidio Catanzano
- Institute for Polymers Composites and Biomaterials (IPCB) - CNR, Pozzuoli, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Joshua S Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Avenue, Chatham Maritime, Kent, UK
| |
Collapse
|
15
|
Bizeau J, Mertz D. Design and applications of protein delivery systems in nanomedicine and tissue engineering. Adv Colloid Interface Sci 2021; 287:102334. [PMID: 33341459 DOI: 10.1016/j.cis.2020.102334] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Proteins are biological macromolecules involved in a wide range of biological functions, which makes them very appealing as therapeutics agents. Indeed, compared to small molecule drugs, their endogenous nature ensures their biocompatibility and biodegradability, they can be used in a large range of applications and present a higher specificity and activity. However, they suffer from unfolding, enzymatic degradation, short half-life and poor membrane permeability. To overcome such drawbacks, the development of protein delivery systems to protect, carry and deliver them in a controlled way have emerged importantly these last years. In this review, the formulation of a wide panel of protein delivery systems either in the form of polymer or inorganic nanoengineered colloids and scaffolds are presented and the protein loading and release mechanisms are addressed. A section is also dedicated to the detection of proteins and the characterization methods of their release. Then, the main protein delivery systems developed these last three years for anticancer, tissue engineering or diabetes applications are presented, as well as the major in vivo models used to test them. The last part of this review aims at presenting the perspectives of the field such as the use of protein-rich material or the sequestration of proteins. This part will also deal with less common applications and gene therapy as an indirect method to deliver protein.
Collapse
|
16
|
Paek SC, Min SK, Park JB. Effects of platelet-derived growth factor-BB on cellular morphology and cellular viability of stem cell spheroids composed of bone-marrow-derived stem cells. Biomed Rep 2020; 13:59. [PMID: 33123373 PMCID: PMC7583700 DOI: 10.3892/br.2020.1366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Platelet-derived growth factor-BB (PDGF-BB) is a potent mitogenic, angiogenic and chemoattractant, and is one of the most abundant growth factors in platelet-derived products. The goal of the present study was to examine the effects of PDGF-BB on cellular morphology and cellular viability using 3D stem cell cultures. On day 1, spheroids formed well in silicon-elastomer-based concave microwells. The addition of 10 or 100 ng/ml PDGF-BB did not affect the morphology of the cell spheroids. During longer periods of incubation, the cell spheroids maintained their shape without noticeable alterations. The majority of cells in the spheroids exhibited green fluorescence when analyzed using a live/dead assay, indicative of live cells. On day 1, the Cell Counting Kit-8 (CCK-8) assay values for PDGF-BB at 0, 10 and 100 ng/ml were 0.241±0.003, 0.227±0.001 and 0.241±0.004, respectively; on day 3, the CCK-8 assay values for PDGF-BB were 0.233±0.005, 0.278±0.001 and 0.194±0.003, respectively; and on day 7, they were 0.248±0.014, 0.293±0.031 and 0.346±0.034, respectively. The 100 ng/ml group showed significantly higher values compared with the control group on day 7. Together, the results of the present study showed that the addition of 10 and 100 ng/ml PDGF-BB increased cellular viability, suggesting that PDGF-BB may be usable in cell therapy.
Collapse
Affiliation(s)
- Soung-Chu Paek
- Department of Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sae Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jun-Beom Park
- Department of Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
17
|
Moreira A, Lawson D, Onyekuru L, Dziemidowicz K, Angkawinitwong U, Costa PF, Radacsi N, Williams GR. Protein encapsulation by electrospinning and electrospraying. J Control Release 2020; 329:1172-1197. [PMID: 33127450 DOI: 10.1016/j.jconrel.2020.10.046] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
Given the increasing interest in the use of peptide- and protein-based agents in therapeutic strategies, it is fundamental to develop delivery systems capable of preserving the biological activity of these molecules upon administration, and which can provide tuneable release profiles. Electrohydrodynamic (EHD) techniques, encompassing electrospinning and electrospraying, allow the generation of fibres and particles with high surface area-to-volume ratios, versatile architectures, and highly controllable release profiles. This review is focused on exploring the potential of different EHD methods (including blend, emulsion, and co-/multi-axial electrospinning and electrospraying) for the development of peptide and protein delivery systems. An overview of the principles of each technique is first presented, followed by a survey of the literature on the encapsulation of enzymes, growth factors, antibodies, hormones, and vaccine antigens using EHD approaches. The possibility for localised delivery using stimuli-responsive systems is also explored. Finally, the advantages and challenges with each EHD method are summarised, and the necessary steps for clinical translation and scaled-up production of electrospun and electrosprayed protein delivery systems are discussed.
Collapse
Affiliation(s)
| | - Dan Lawson
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, Edinburgh EH9 3FB, UK
| | - Lesley Onyekuru
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Karolina Dziemidowicz
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Ukrit Angkawinitwong
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal.
| | - Norbert Radacsi
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, Edinburgh EH9 3FB, UK.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
18
|
An alginate/poly(N-isopropylacrylamide)-based composite hydrogel dressing with stepwise delivery of drug and growth factor for wound repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111123. [DOI: 10.1016/j.msec.2020.111123] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/24/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023]
|
19
|
Zhang F, Qiao S, Li C, Wu B, Reischl S, Neumann PA. The immunologic changes during different phases of intestinal anastomotic healing. J Clin Lab Anal 2020; 34:e23493. [PMID: 32692419 PMCID: PMC7676198 DOI: 10.1002/jcla.23493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 01/06/2023] Open
Abstract
Intestinal anatomosis is a complex and multicellular process that involving three overlapped phases: exudative phase, proliferative phase, and reparative phase. Undisturbed anastomotic healings are crucial for the recovery of patients after operations but unsuccessful healings are linked with a considerable mortality. This time, we concentrate on the immunologic changes during different phases of intestinal anastomotic healing and select several major immune cells and cytokines of each phase to get a better understanding of these immunologic changes in different phases, which will be significant for more precise therapy strategies in anastomoses.
Collapse
Affiliation(s)
- Feng Zhang
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China.,Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Song Qiao
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China
| | - Chunqiao Li
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Bo Wu
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China
| | - Stefan Reischl
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Philipp-Alexander Neumann
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| |
Collapse
|
20
|
Li R, Xu Z, Jiang Q, Zheng Y, Chen Z, Chen X. Characterization and biological evaluation of a novel silver nanoparticle-loaded collagen-chitosan dressing. Regen Biomater 2020; 7:371-380. [PMID: 32793382 PMCID: PMC7414998 DOI: 10.1093/rb/rbaa008] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/15/2020] [Accepted: 02/23/2020] [Indexed: 12/19/2022] Open
Abstract
Effective coverage and protection is a priority in wound treatment. Collagen and chitosan have been widely used for wound dressings due to their excellent biological activity and biocompatibility. Silver nanoparticles (AgNPs) have a powerful antibacterial effect. In this study, a macromolecular and small-molecular collagen mixed solution, a macromolecular and small-molecular chitosan mixed solution were prepared, and a silver nanoparticle-loaded collagen-chitosan dressing (AgNP-CCD) has been proposed. First, the effects of a collagen-chitosan mixed solution on the proliferation of human umbilical vein endothelial cells and the secretion of cytokines were evaluated. Then, the characteristics and antibacterial effects of the AgNP-CCD were tested, and the effects on wound healing and the influence of wound cytokine expression were investigated via a deep second-degree burn wound model. The results showed that at the proper proportion and concentration, the collagen-chitosan mixed solution effectively promoted cell proliferation and regulated the levels of growth factors (vascular endothelial growth factor [VEGF], epidermal growth factor [EGF], platelet-derived growth factor [PDGF], transforming growth factor [TGF-β1], basic fibroblastic growth factor [bFGF]) and inflammatory factors (TNF-α, IL-1β, IL-6, IL-8). Moreover, AgNP solutions at lower concentrations exerted limited inhibitory effects on cell proliferation and had no effect on cytokine secretion. The AgNP-CCD demonstrated satisfactory morphological and physical properties as well as efficient antibacterial activities. An in vivo evaluation indicated that AgNP-CCD could accelerate the healing process of deep second-degree burn wounds and played an important role in the regulation of growth and inflammatory factors, including VEGF, EGFL-7, TGF-β1, bFGF, TNF-α and IL-1β. This AgNP-CCD exerted excellent biological effects on wound healing promotion and cytokine expression regulation.
Collapse
Affiliation(s)
- Rongfu Li
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Department of Critical Care Medicine, Quanzhou First Hospital Affiliated Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Zhaorong Xu
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Qiong Jiang
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Yunquan Zheng
- Institute of Pharmaceutical Biotechnology and Engineering, Fuzhou University, Fuzhou, Fujian 350001, China
| | - Zhaohong Chen
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Xiaodong Chen
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| |
Collapse
|
21
|
Sabra S, Ragab DM, Agwa MM, Rohani S. Recent advances in electrospun nanofibers for some biomedical applications. Eur J Pharm Sci 2020; 144:105224. [DOI: 10.1016/j.ejps.2020.105224] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
|
22
|
Evrova O, Kellenberger D, Calcagni M, Vogel V, Buschmann J. Supporting Cell-Based Tendon Therapy: Effect of PDGF-BB and Ascorbic Acid on Rabbit Achilles Tenocytes in Vitro. Int J Mol Sci 2020; 21:ijms21020458. [PMID: 31936891 PMCID: PMC7014238 DOI: 10.3390/ijms21020458] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/21/2022] Open
Abstract
Cell-based tendon therapies with tenocytes as a cell source need effective tenocyte in vitro expansion before application for tendinopathies and tendon injuries. Supplementation of tenocyte culture with biomolecules that can boost proliferation and matrix synthesis is one viable option for supporting cell expansion. In this in vitro study, the impacts of ascorbic acid or PDGF-BB supplementation on rabbit Achilles tenocyte culture were studied. Namely, cell proliferation, changes in gene expression of several ECM and tendon markers (collagen I, collagen III, fibronectin, aggrecan, biglycan, decorin, ki67, tenascin-C, tenomodulin, Mohawk, α-SMA, MMP-2, MMP-9, TIMP1, and TIMP2) and ECM deposition (collagen I and fibronectin) were assessed. Ascorbic acid and PDGF-BB enhanced tenocyte proliferation, while ascorbic acid significantly accelerated the deposition of collagen I. Both biomolecules led to different changes in the gene expression profile of the cultured tenocytes, where upregulation of collagen I, Mohawk, decorin, MMP-2, and TIMP-2 was observed with ascorbic acid, while these markers were downregulated by PDGF-BB supplementation. Vice versa, there was an upregulation of fibronectin, biglycan and tenascin-C by PDGF-BB supplementation, while ascorbic acid led to a downregulation of these markers. However, both biomolecules are promising candidates for improving and accelerating the in vitro expansion of tenocytes, which is vital for various tendon tissue engineering approaches or cell-based tendon therapy.
Collapse
Affiliation(s)
- Olivera Evrova
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Damian Kellenberger
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Maurizio Calcagni
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Johanna Buschmann
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
- Correspondence: ; Tel.: +41-44-255-9895
| |
Collapse
|
23
|
Tiffany AS, Dewey MJ, Harley BAC. Sequential sequestrations increase the incorporation and retention of multiple growth factors in mineralized collagen scaffolds. RSC Adv 2020; 10:26982-26996. [PMID: 33767853 PMCID: PMC7990239 DOI: 10.1039/d0ra03872e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Trauma induced injuries of the mouth, jaw, face, and related structures present unique clinical challenges due to their large size and complex geometry. Growth factor signaling coordinates the behavior of multiple cell types following an injury, and effective coordination of growth factor availability within a biomaterial can be critical for accelerating bone healing. Mineralized collagen scaffolds are a class of degradable biomaterial whose biophysical and compositional parameters can be adjusted to facilitate cell invasion and tissue remodeling. Here we describe the use of modified simulated body fluid treatments to enable sequential sequestration of bone morphogenic protein 2 and vascular endothelial growth factor into mineralized collagen scaffolds for bone repair. We report the capability of these scaffolds to sequester 60–90% of growth factor from solution without additional crosslinking treatments and show high levels of retention for individual (>94%) and multiple growth factors (>88%) that can be layered into the material via sequential sequestration steps. Sequentially sequestering growth factors allows prolonged release of growth factors in vitro (>94%) and suggests the potential to improve healing of large-scale bone injury models in vivo. Future work will utilize this sequestration method to induce cellular activities critical to bone healing such as vessel formation and cell migration. Trauma induced injuries of the mouth, jaw, face, and related structures present unique clinical challenges due to their large size and complex geometry.![]()
Collapse
Affiliation(s)
- Aleczandria S Tiffany
- Dept. Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Marley J Dewey
- Dept. Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
24
|
Cardiac cell differentiation of muscle satellite cells on aligned composite electrospun polyurethane with reduced graphene oxide. JOURNAL OF POLYMER RESEARCH 2019. [DOI: 10.1007/s10965-019-1936-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Bhattacharya D, Ghosh B, Mukhopadhyay M. Development of nanotechnology for advancement and application in wound healing: a review. IET Nanobiotechnol 2019; 13:778-785. [PMID: 31625517 PMCID: PMC8676206 DOI: 10.1049/iet-nbt.2018.5312] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 06/03/2019] [Accepted: 06/28/2019] [Indexed: 12/21/2022] Open
Abstract
Wound healing is a series of different dynamic and complex phenomena. Many studies have been carried out based on the type and severity of wounds. However, to recover wounds faster there are no suitable drugs available, which are highly stable, less expensive as well as has no side effects. Nanomaterials have been proven to be the most promising agent for faster wound healing among all the other wound healing materials. This review briefly discusses the recent developments of wound healing by nanotechnology, their applicability and advantages. Nanomaterials have unique physicochemical, optical, and biological properties. Some of them can be directly applied for wound healing or some of them can be incorporated into scaffolds to create hydrogel matrix or nanocomposites, which promote wound healing through their antimicrobial, as well as selective anti- and pro-inflammatory, and proangiogenic properties. Owing to their high surface area to volume ratio, nanomaterials have not only been used for drug delivery vectors but also can affect wound healing by influencing collagen deposition and realignment and provide approaches for skin tissue regeneration.
Collapse
Affiliation(s)
- Debalina Bhattacharya
- Department of Biotechnology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| | - Biva Ghosh
- Department of Biotechnology, JIS University, 81 Nilgunj Road, Kolkata 700109, West Bengal, India
| | - Mainak Mukhopadhyay
- Department of Biotechnology, JIS University, 81 Nilgunj Road, Kolkata 700109, West Bengal, India
| |
Collapse
|
26
|
Piran M, Shiri M, Soufi Zomorrod M, Esmaeili E, Soufi Zomorrod M, Vazifeh Shiran N, Mahboudi H, Daneshpazhouh H, Dehghani N, Hosseinzadeh S. Electrospun triple-layered PLLA/gelatin. PRGF/PLLA scaffold induces fibroblast migration. J Cell Biochem 2019; 120:11441-11453. [PMID: 30746766 DOI: 10.1002/jcb.28422] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
The function of fibroblast cells in wounded areas results in reconstruction of the extra cellular matrix and consequently resolution of granulation tissue. It is suggested that the use of platelet-rich plasma can accelerate the healing process in nonhealing or slow-healing wounds. In this study, a simple and novel method has been used to fabricate an electrospun three-layered scaffold containing plasma rich in growth factor with the aim of increasing the proliferation and migration of fibroblast cells in vitro. First, plasma rich in growth factor was derived from platelet rich plasma, and then a three-layered scaffold was fabricated using PLLA nanofibers as the outer layers and plasma rich in growth factor-containing gelatin fibers as the internal layer. The growth morphology of cells seeded on this scaffold was compared to those seeded on one layered PLLA scaffold. The study of the cell growth rate on different substrates and the migration of cells in response to the drug release of multilayered scaffold was investigated by the cell quantification assay and a modified under agarose assay. Scanning electron microscopy and fluorescence images showed that cells seeded on multilayered scaffold were completely oriented 72 hours after seeding compared to those seeded on PLLA scaffold. The cell quantification assay also indicated significant increase in proliferation rate of cells seeded on three-layered scaffold compared to those seeded on PLLA scaffold and finally, monitoring cell migration proved that cells migrate significantly toward the three-layered scaffold up to 48 to 72 hours and afterwards start to show a diminished migration rate toward this scaffold.
Collapse
Affiliation(s)
- Mehrdad Piran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran.,Department of Tissue Engineering and Nanotechnology, Stem Cell Technology Research Center, Tehran, Iran
| | - Mahdi Shiri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical sciences research center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Soufi Zomorrod
- Department of Tissue Engineering and Nanotechnology, Stem Cell Technology Research Center, Tehran, Iran
| | - Elaheh Esmaeili
- Department of Tissue Engineering and Nanotechnology, Stem Cell Technology Research Center, Tehran, Iran.,Department of Hematology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Mina Soufi Zomorrod
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Nader Vazifeh Shiran
- Department of Hematology and blood banking, Paramedical Faculty, Shahid Beheshti university of Medical Sciences, Tehran, Iran
| | - Hossein Mahboudi
- Dietary Supplements and Probiotic Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Biotechnology, School of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | | | | | - Simzar Hosseinzadeh
- Department of Tissue engineering and Applied Cell Sciences, School Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|