1
|
Dini C, Borges MHR, Malheiros SS, Piazza RD, van den Beucken JJJP, de Avila ED, Souza JGS, Barão VAR. Progress in Designing Therapeutic Antimicrobial Hydrogels Targeting Implant-associated Infections: Paving the Way for a Sustainable Platform Applied to Biomedical Devices. Adv Healthc Mater 2025; 14:e2402926. [PMID: 39440583 DOI: 10.1002/adhm.202402926] [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: 08/06/2024] [Revised: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Implantable biomedical devices have found widespread use in restoring lost functions or structures within the human body, but they face a significant challenge from microbial-related infections, which often lead to implant failure. In this context, antimicrobial hydrogels emerge as a promising strategy for treating implant-associated infections owing to their tunable physicochemical properties. However, the literature lacks a comprehensive analysis of antimicrobial hydrogels, encompassing their development, mechanisms, and effect on implant-associated infections, mainly in light of existing in vitro, in vivo, and clinical evidence. Thus, this review addresses the strategies employed by existing studies to tailor hydrogel properties to meet the specific needs of each application. Furthermore, this comprehensive review critically appraises the development of antimicrobial hydrogels, with a particular focus on solving infections related to metallic orthopedic or dental implants. Then, preclinical and clinical studies centering on providing quantitative microbiological results associated with the application of antimicrobial hydrogels are systematically summarized. Overall, antimicrobial hydrogels benefit from the tunable properties of polymers and hold promise as an effective strategy for the local treatment of implant-associated infections. However, future clinical investigations, grounded on robust evidence from in vitro and preclinical studies, are required to explore and validate new antimicrobial hydrogels for clinical use.
Collapse
Affiliation(s)
- Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Maria Helena Rossy Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Samuel Santana Malheiros
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Rodolfo Debone Piazza
- Physical Chemistry Department, Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, 14800-900, Brazil
| | | | - Erica Dorigatti de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, São Paulo, 16015-050, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Guarulhos University (UNG), Guarulhos, São Paulo, 07023-070, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| |
Collapse
|
2
|
Murugan S, Ha CH, Appana Dalavi P, Anil S, Venkatesan J, Seong GH. Enhanced Bioactivity of Chitosan-Alginate-Riboflavin Liquid-Exfoliated Molybdenum Disulfide Nanosheets for Bone Tissue Engineering Applications. ACS OMEGA 2024; 9:46904-46915. [PMID: 39619530 PMCID: PMC11603203 DOI: 10.1021/acsomega.4c06049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 12/16/2024]
Abstract
Bone tissue engineering is a growing field that provides solutions for the treatment of bone deformities, injuries, diseases, and anomalies by replacing autograft and allograft procedures. Various scaffolding materials have been used for the construction of bone tissue, including metals, ceramics, and polymers. This study investigates an innovative liquid exfoliation approach for the production of molybdenum disulfide (MoS2) nanosheets using riboflavin (RF-MoS2) as an exfoliation agent and subsequently analytically characterized for the development of bone scaffolding system. UV analysis of RF-MoS2 shows the absorbance spectra at 610 and 668 nm and the particle size was 123 ± 4 nm and a surface charge of -16.1 ± 2 mV. Further, alginate-chitosan (Alg-Chi) and alginate-chitosan-riboflavin-MoS2 (Alg-Chi-RF-MoS2) nanocomposite scaffolds were developed. The morphology of the Alg-Chi-RF-MoS2 scaffold was studied using scanning electron microscopy and pore size was found to be 210 ± 10 μm. Alg-Chi-RF-MoS2 scaffolds generate calcium phosphate biominerals when immersed in a simulated body fluid. Alg-Chi and Alg-Chi-RF-MoS2 scaffolds were biocompatible with C3H10T1/2 cells, and scaffolds showed a significant increase in alkaline phosphatase and mineralization. Thus, the developed Alg-Chi-RF-MoS2 scaffold proved to be an appropriate artificial graft for bone graft substitution.
Collapse
Affiliation(s)
- Sesha
Subramanian Murugan
- Department
of Bionano Engineering, Center for Bionano Intelligence Education
and Research, Hanyang University, Ansan 426-791, South Korea
- Biomaterials
Research Laboratory, Yenepoya Research Centre, Yenepoya Deemed to be University, Deralakatte, Mangaluru 575018, India
| | - Chang Hyeon Ha
- Department
of Bionano Engineering, Center for Bionano Intelligence Education
and Research, Hanyang University, Ansan 426-791, South Korea
| | - Pandurang Appana Dalavi
- Biomaterials
Research Laboratory, Yenepoya Research Centre, Yenepoya Deemed to be University, Deralakatte, Mangaluru 575018, India
| | - Sukumaran Anil
- Oral
Health Institute, Hamad Medical Corporation, Doha 3050, Qatar
- College
of Dental Medicine, Qatar University, Doha 2713, Qatar
| | - Jayachandran Venkatesan
- Department
of Bionano Engineering, Center for Bionano Intelligence Education
and Research, Hanyang University, Ansan 426-791, South Korea
- Biomaterials
Research Laboratory, Yenepoya Research Centre, Yenepoya Deemed to be University, Deralakatte, Mangaluru 575018, India
| | - Gi Hun Seong
- Department
of Bionano Engineering, Center for Bionano Intelligence Education
and Research, Hanyang University, Ansan 426-791, South Korea
| |
Collapse
|
3
|
Nelogi S, Kumarpatil A, Chowdhary R, Roy R. Optimising titanium implant stability and infection resistance through iron nanoparticle coatings: A preclinical investigation. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2024; 126:102155. [PMID: 39551183 DOI: 10.1016/j.jormas.2024.102155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Attaining adequate osseointegration and mitigating infections are paramount issues in implantology, especially within dental and orthopaedic domains. Titanium implants have been utilised for their biocompatibility and mechanical strength; yet, problems such as peri‑implant infections and inadequate bone integration may undermine their efficacy. Coating titanium implants with iron nanoparticles (FeNp) has surfaced as a promising approach to improve osseointegration and antibacterial characteristics. FeNp's distinctive capacity to react to magnetic fields and produce reactive oxygen species (ROS) has the potential to enhance implant results. OBJECTIVE To assess the influence of FeNp-coated titanium implants on osseointegration, mechanical stability, osteogenesis, and antibacterial effectiveness against prevalent implant-associated infections, Staphylococcus aureus and Escherichia coli. MATERIALS AND METHODS In vivo investigations were performed on animal models to evaluate implant stability by resonance frequency analysis (RFA) and removal torque measurements at 6 and 12 weeks post-implantation. Histopathological assessment was conducted to analyze the osseous formation and vascularization surrounding the implants. Furthermore, in vitro experiments were employed to assess the antibacterial efficacy of magnetized FeNp against S. aureus and E. coli. RESULTS At 6 weeks, no substantial change was detected in (RFA) or removal torque between the control group (GROUP A) and the test group (GROUP B). However, by 12 weeks, GROUP B demonstrated significantly higher RFA scores (75.02 ± 5.11) compared to GROUP A (67.41 ± 9.85), indicating improved implant stability (p < 0.05). Removal torque values were also significantly higher in GROUP B at 12 weeks (76.30 ± 14.20) compared to GROUP A (46.10 ± 9.25), suggesting enhanced mechanical integration (p < 0.01). Histopathological analysis revealed greater new bone formation, increased osteoblast activity, and improved vascularization around FeNp-coated implants in GROUP B. Additionally, in vitro antibacterial testing demonstrated that FeNp coatings effectively inhibited the growth of Staphylococcus aureus and E. coli, providing further evidence of its antimicrobial effect CONCLUSION: FeNp-coated implants have dual advantages: improved osseointegration and antibacterial defence. The findings indicate that FeNp coatings might substantially enhance implant longevity and diminish the likelihood of infection, offering a potential approach for clinical applications, especially for patients at elevated risk of implant failure. Subsequent research should concentrate on enhancing the application of FeNp coatings in clinical environments and further examining their long-term biocompatibility and effectiveness.
Collapse
Affiliation(s)
- Santosh Nelogi
- KLEVK Institute of Dental Science, KLE Academy of Higher Education and Research, Belgavi, Karnataka, India.
| | - Anand Kumarpatil
- Department of Prosthodontics, KLEVK Institute of Dental Science, KLE Academy of Higher Education and Research, Belgavi, Karnataka, India
| | - Ramesh Chowdhary
- Department of Prosthodontics, Siddhartha Institute of Dental Science, Karnataka, India
| | - Richa Roy
- Cochin Implant Centre Kalamassery, Cochin, India
| |
Collapse
|
4
|
Alashi S, Alkhouri I, Alghoraibi I, Kochaji N, Houri A, Karkoutly M. Evaluating various properties of nanohydroxyapatite synthesized from eggshells and dual-doped with Si 4+ and Zn 2+: An in vitro study. Heliyon 2024; 10:e35907. [PMID: 39224256 PMCID: PMC11366878 DOI: 10.1016/j.heliyon.2024.e35907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Background This study aimed to evaluate morphological, chemical and biocompatible properties of nanohydroxyapatite (N-HA) synthesized from eggshells and dual-doped with Si4+ and Zn2+. Methods In the current study, N-HA was synthesized from chicken eggshells using the wet chemical precipitation method and doped with Si4+ and Zn2+. The physical assessment was carried out using field emission scanning electron microscopy (FE-SEM), energy dispersive X-ray (EDX) analysis, and X-ray diffraction (XRD) analysis. Crystal size was calculated using the Scherrer equation. Cytotoxicity was studied in vitro using the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) cytotoxicity assay. The optical density (OD) of each well was obtained and recorded at 570 nm for 24 h (t1), 48 h (t2), 72 h (t3), and 5 days (t4) using a microplate reader. Results The results of Si-Zn-doped HA showed a high specific surface area with an irregular nano-sized spherical particle structure. The atomic percentage provided the ratio of calcium to phosphate; for non-doped HA, the atomic Ca/P ratio was 1.6, but for Si-Zn-doped HA, where Zn+2 Ca and Si + replaced 4 substituted P, the atomic ratio (Ca + Zn)/(P + Si) was 1.76. The average crystal size of Si-Zn-doped HA was 46 nm, while for non-doped HA it was 61 nm. both samples were non-toxic and statistically significantly less viable than the control group After 5 days, the mean cell viability of Si-Zn-doped HA (79.17 ± 2.18) was higher than that of non-doped HA (76.26 ± 1.71) (P = 0.091). Conclusions The MTT assay results showed that Si-Zn-doped HA is biocompatible. In addition, it showed characteristic physiochemical properties of a large surface area with interconnected porosity.
Collapse
Affiliation(s)
- Shaza Alashi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Isam Alkhouri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Ibrahim Alghoraibi
- Department of Physics, Faculty of Science, Damascus University, Damascus, Syrian Arab Republic
| | - Nabil Kochaji
- Department of Oral Pathology, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Abdullah Houri
- Department of Physics, Faculty of Science, Damascus University, Damascus, Syrian Arab Republic
| | - Mawia Karkoutly
- Department of Pediatric Dentistry, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| |
Collapse
|
5
|
Shin HE, Han JH, Shin S, Bae GH, Son B, Kim TH, Park HH, Park CG, Park W. M1-polarized macrophage-derived cellular nanovesicle-coated lipid nanoparticles for enhanced cancer treatment through hybridization of gene therapy and cancer immunotherapy. Acta Pharm Sin B 2024; 14:3169-3183. [PMID: 39027257 PMCID: PMC11252390 DOI: 10.1016/j.apsb.2024.03.004] [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: 01/05/2024] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 07/20/2024] Open
Abstract
Optimum genetic delivery for modulating target genes to diseased tissue is a major obstacle for profitable gene therapy. Lipid nanoparticles (LNPs), considered a prospective vehicle for nucleic acid delivery, have demonstrated efficacy in human use during the COVID-19 pandemic. This study introduces a novel biomaterial-based platform, M1-polarized macrophage-derived cellular nanovesicle-coated LNPs (M1-C-LNPs), specifically engineered for a combined gene-immunotherapy approach against solid tumor. The dual-function system of M1-C-LNPs encapsulates Bcl2-targeting siRNA within LNPs and immune-modulating cytokines within M1 macrophage-derived cellular nanovesicles (M1-NVs), effectively facilitating apoptosis in cancer cells without impacting T and NK cells, which activate the intratumoral immune response to promote granule-mediating killing for solid tumor eradication. Enhanced retention within tumor was observed upon intratumoral administration of M1-C-LNPs, owing to the presence of adhesion molecules on M1-NVs, thereby contributing to superior tumor growth inhibition. These findings represent a promising strategy for the development of targeted and effective nanoparticle-based cancer genetic-immunotherapy, with significant implications for advancing biomaterial use in cancer therapeutics.
Collapse
Affiliation(s)
- Ha Eun Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seungyong Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Ga-Hyun Bae
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Boram Son
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Tae-Hyung Kim
- Department of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Chun Gwon Park
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Biomedical Engineering, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
6
|
Yoon S, Fuwad A, Jeong S, Cho H, Jeon TJ, Kim SM. Surface Deformation of Biocompatible Materials: Recent Advances in Biological Applications. Biomimetics (Basel) 2024; 9:395. [PMID: 39056836 PMCID: PMC11274418 DOI: 10.3390/biomimetics9070395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
The surface topography of substrates is a crucial factor that determines the interaction with biological materials in bioengineering research. Therefore, it is important to appropriately modify the surface topography according to the research purpose. Surface topography can be fabricated in various forms, such as wrinkles, creases, and ridges using surface deformation techniques, which can contribute to the performance enhancement of cell chips, organ chips, and biosensors. This review provides a comprehensive overview of the characteristics of soft, hard, and hybrid substrates used in the bioengineering field and the surface deformation techniques applied to the substrates. Furthermore, this review summarizes the cases of cell-based research and other applications, such as biosensor research, that utilize surface deformation techniques. In cell-based research, various studies have reported optimized cell behavior and differentiation through surface deformation, while, in the biosensor and biofilm fields, performance improvement cases due to surface deformation have been reported. Through these studies, we confirm the contribution of surface deformation techniques to the advancement of the bioengineering field. In the future, it is expected that the application of surface deformation techniques to the real-time interaction analysis between biological materials and dynamically deformable substrates will increase the utilization and importance of these techniques in various fields, including cell research and biosensors.
Collapse
Affiliation(s)
- Sunhee Yoon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Industry-Academia Interactive R&E Center for Bioprocess Innovation (BK21), Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Ahmed Fuwad
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
| | - Seorin Jeong
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
| | - Hyeran Cho
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Industry-Academia Interactive R&E Center for Bioprocess Innovation (BK21), Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Sun Min Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
7
|
Qin D, Zhao Y, Cheng R, Liu Y, Guo S, Sun L, Guo Y, Hao F, Zhao B. Mussel-inspired immunomodulatory and osteoinductive dual-functional hydroxyapatite nanoplatform for promoting bone regeneration. J Nanobiotechnology 2024; 22:320. [PMID: 38849820 PMCID: PMC11162024 DOI: 10.1186/s12951-024-02593-3] [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: 03/16/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Simultaneously modulating the inflammatory microenvironment and promoting local bone regeneration is one of the main challenges in treating bone defects. In recent years, osteoimmunology has revealed that the immune system plays an essential regulatory role in bone regeneration and that macrophages are critical components. In this work, a mussel-inspired immunomodulatory and osteoinductive dual-functional hydroxyapatite nano platform (Gold/hydroxyapatite nanocomposites functionalized with polydopamine - PDA@Au-HA) is developed to accelerate bone tissues regeneration by regulating the immune microenvironment. PDA coating endows nanomaterials with the ability to scavenge reactive oxygen species (ROS) and anti-inflammatory properties, and it also exhibits an immunomodulatory ability to inhibit M1 macrophage polarization and activate M2 macrophage secretion of osteogenesis-related cytokines. Most importantly, this nano platform promotes the polarization of M2 macrophages and regulates the crosstalk between macrophages and pre-osteoblast cells to achieve bone regeneration. Au-HA can synergistically promote vascularized bone regeneration through sustained release of Ca and P particles and gold nanoparticles (NPs). This nano platform has a synergistic effect of good compatibility, scavenging of ROS, and anti-inflammatory and immunomodulatory capability to accelerate the bone repair process. Thus, our research offers a possible therapeutic approach by exploring PDA@Au-HA nanocomposites as a bifunctional platform for tissue regeneration.
Collapse
Affiliation(s)
- Danlei Qin
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yifan Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
| | - Rui Cheng
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yingyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
| | - Susu Guo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
| | - Lingxiang Sun
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
| | - Yanqin Guo
- Department of Ultrasound, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Fengxiang Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China
| | - Bin Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
8
|
Gharatape A, Sadeghi-Abandansari H, Seifalian A, Faridi-Majidi R, Basiri M. Nanocarrier-based gene delivery for immune cell engineering. J Mater Chem B 2024; 12:3356-3375. [PMID: 38505950 DOI: 10.1039/d3tb02279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Clinical advances in genetically modified immune cell therapies, such as chimeric antigen receptor T cell therapies, have raised hope for cancer treatment. The majority of these biotechnologies are based on viral methods for ex vivo genetic modification of the immune cells, while the non-viral methods are still in the developmental phase. Nanocarriers have been emerging as materials of choice for gene delivery to immune cells. This is due to their versatile physicochemical properties such as large surface area and size that can be optimized to overcome several practical barriers to successful gene delivery. The in vivo nanocarrier-based gene delivery can revolutionize cell-based cancer immunotherapies by replacing the current expensive autologous cell manufacturing with an off-the-shelf biomaterial-based platform. The aim of this research is to review current advances and strategies to overcome the challenges in nanoparticle-based gene delivery and their impact on the efficiency, safety, and specificity of the process. The main focus is on polymeric and lipid-based nanocarriers, and their recent preclinical applications for cancer immunotherapy.
Collapse
Affiliation(s)
- Alireza Gharatape
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hamid Sadeghi-Abandansari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd, Nanoloom Ltd, & Liberum Health Ltd), London BioScience Innovation Centre, London, UK
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
9
|
Mohammadrezaei D, Podina L, Silva JD, Kohandel M. Cell viability prediction and optimization in extrusion-based bioprinting via neural network-based Bayesian optimization models. Biofabrication 2024; 16:025016. [PMID: 38128119 DOI: 10.1088/1758-5090/ad17cf] [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: 06/27/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
The fields of regenerative medicine and cancer modeling have witnessed tremendous growth in the application of 3D bioprinting. Maintaining high cell viability throughout the bioprinting process is crucial for the success of this technology, as it directly affects the accuracy of the 3D bioprinted models, the validity of experimental results, and the discovery of new therapeutic approaches. Therefore, optimizing bioprinting conditions, which include numerous variables influencing cell viability during and after the procedure, is of utmost importance to achieve desirable results. So far, these optimizations have been accomplished primarily through trial and error and repeating multiple time-consuming and costly experiments. To address this challenge, we initiated the process by creating a dataset of these parameters for gelatin and alginate-based bioinks and the corresponding cell viability by integrating data obtained in our laboratory and those derived from the literature. Then, we developed machine learning models to predict cell viability based on different bioprinting variables. The trained neural network yielded regressionR2value of 0.71 and classification accuracy of 0.86. Compared to models that have been developed so far, the performance of our models is superior and shows great prediction results. The study further introduces a novel optimization strategy that employs the Bayesian optimization model in combination with the developed regression neural network to determine the optimal combination of the selected bioprinting parameters to maximize cell viability and eliminate trial-and-error experiments. Finally, we experimentally validated the optimization model's performance.
Collapse
Affiliation(s)
- Dorsa Mohammadrezaei
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Lena Podina
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Johanna De Silva
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
10
|
Sobhani-Nasab A, Banafshe HR, Atapour A, Khaksary Mahabady M, Akbari M, Daraei A, Mansoori Y, Moradi Hasan-Abad A. The use of nanoparticles in the treatment of infectious diseases and cancer, dental applications and tissue regeneration: a review. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 5:1330007. [PMID: 38323112 PMCID: PMC10844477 DOI: 10.3389/fmedt.2023.1330007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024] Open
Abstract
The emergence of nanotechnology as a field of study can be traced back to the 1980s, at which point the means to artificially produce, control, and observe matter on a nanometer level was made viable. Recent advancements in technology have enabled us to extend our reach to the nanoscale, which has presented an unparalleled opportunity to directly target biomolecular interactions. As a result of these developments, there is a drive to arise intelligent nanostructures capable of overcoming the obstacles that have impeded the progress of conventional pharmacological methodologies. After four decades, the gradual amalgamation of bio- and nanotechnologies is initiating a revolution in the realm of disease detection, treatment, and monitoring, as well as unsolved medical predicaments. Although a significant portion of research in the field is still confined to laboratories, the initial application of nanotechnology as treatments, vaccines, pharmaceuticals, and diagnostic equipment has now obtained endorsement for commercialization and clinical practice. The current issue presents an overview of the latest progress in nanomedical strategies towards alleviating antibiotic resistance, diagnosing and treating cancer, addressing neurodegenerative disorders, and an array of applications, encompassing dentistry and tuberculosis treatment. The current investigation also scrutinizes the deployment of sophisticated smart nanostructured materials in fields of application such as regenerative medicine, as well as the management of targeted and sustained release of pharmaceuticals and therapeutic interventions. The aforementioned concept exhibits the potential for revolutionary advancements within the field of immunotherapy, as it introduces the utilization of implanted vaccine technology to consistently regulate and augment immune functions. Concurrently with the endeavor to attain the advantages of nanomedical intervention, it is essential to enhance the unceasing emphasis on nanotoxicological research and the regulation of nanomedications' safety. This initiative is crucial in achieving the advancement in medicine that currently lies within our reach.
Collapse
Affiliation(s)
- Ali Sobhani-Nasab
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Reza Banafshe
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Akbari
- Department of Surgery, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
11
|
Lizarazo-Fonseca L, Correa-Araujo L, Prieto-Abello L, Camacho-Rodríguez B, Silva-Cote I. In vitro and in vivo evaluation of electrospun poly (ε-caprolactone)/collagen scaffolds and Wharton's jelly mesenchymal stromal cells (hWJ-MSCs) constructs as potential alternative for skin tissue engineering. Regen Ther 2023; 24:11-24. [PMID: 37284730 PMCID: PMC10239703 DOI: 10.1016/j.reth.2023.05.005] [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: 01/06/2023] [Revised: 03/08/2023] [Accepted: 05/20/2023] [Indexed: 06/08/2023] Open
Abstract
Dermal substitutes bear a high clinical demand because of their ability to promote the healing process of cutaneous wounds by reducing the healing time the appearance and improving the functionality of the repaired tissue. Despite the increasing development of dermal substitutes, most of them are only composed of biological or biosynthetic matrices. This demonstrates the need for new developments focused on using scaffolds with cells (tissue construct) that promote the production of factors for biological signaling, wound coverage, and general support of the tissue repair process. Here, we fabricate by electrospinning two scaffolds: poly(ε-caprolactone) (PCL) as a control and poly(ε-caprolactone)/collagen type I (PCol) in a ratio lower collagen than previously reported, 19:1, respectively. Then, characterize their physicochemical and mechanical properties. As we bear in mind the creation of a biologically functional construct, we characterize and assess in vitro the implications of seeding human Wharton's jelly mesenchymal stromal cells (hWJ-MSCs) on both scaffolds. Finally, to determine the potential functionality of the constructs in vivo, their efficiency was evaluated in a porcine biomodel. Our findings demonstrated that collagen incorporation in the scaffolds produces fibers with similar diameters to those in the human native extracellular matrix, increases wettability, and enhances the presence of nitrogen on the scaffold surface, improving cell adhesion and proliferation. These synthetic scaffolds improved the secretion of factors by hWJ-MSCs involved in skin repair processes such as b-FGF and Angiopoietin I and induced its differentiation towards epithelial lineage, as shown by the increased expression of Involucrin and JUP. In vivo experiments confirmed that lesions treated with the PCol/hWJ-MSCs constructs might reproduce a morphological organization that seems relatively equivalent to normal skin. These results suggest that the PCol/hWJ-MSCs construct is a promising alternative for skin lesions repair in the clinic.
Collapse
Affiliation(s)
| | | | | | | | - Ingrid Silva-Cote
- Corresponding author. Secretaría Distrital de Salud, Carrera 32 # 12-81, Bogotá, Colombia
| |
Collapse
|
12
|
Selvam A, Majood M, Chaurasia R, Rupesh, Singh A, Dey T, Agrawal O, Verma YK, Mukherjee M. Injectable organo-hydrogels influenced by click chemistry as a paramount stratagem in the conveyor belt of pharmaceutical revolution. J Mater Chem B 2023; 11:10761-10777. [PMID: 37807713 DOI: 10.1039/d3tb01674a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The field of injectable hydrogels has demonstrated a paramount headway in the myriad of biomedical applications and paved a path toward clinical advancements. The innate superiority of hydrogels emerging from organic constitution has exhibited dominance in overcoming the bottlenecks associated with inorganic-based hydrogels in the biological milieu. Inorganic hydrogels demonstrate various disadvantages, including limited biocompatibility, degradability, a cumbersome synthesis process, high cost, and ecotoxicity. The excellent biocompatibility, eco-friendliness, and manufacturing convenience of organo-hydrogels have demonstrated to be promising in therapizing biomedical complexities with low toxicity and augmented bioavailability. This report manifests the realization of biomimetic organo-hydrogels with the development of bioresponsive and self-healing injectable organo-hydrogels in the emerging pharmaceutical revolution. Furthermore, the influence of click chemistry in this regime as a backbone in the pharmaceutical conveyor belt has been suggested to scale up production. Moreover, we propose an avant-garde design stratagem of developing a hyaluronic acid (HA)-based injectable organo-hydrogel via click chemistry to be realized for its pharmaceutical edge. Ultimately, injectable organo-hydrogels that materialize from academia or industry are required to follow the standard set of rules established by global governing bodies, which has been delineated to comprehend their marketability. Thence, this perspective narrates the development of injectable organo-hydrogels via click chemistry as a prospective elixir to have in the arsenal of pharmaceuticals.
Collapse
Affiliation(s)
- Abhyavartin Selvam
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, 201313, India
| | - Misba Majood
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Radhika Chaurasia
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Rupesh
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Akanksha Singh
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Tapan Dey
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Omnarayan Agrawal
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Yogesh Kumar Verma
- Stem Cell & Tissue Engineering Research Group, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, New Delhi, 110054, India
| | - Monalisa Mukherjee
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida, 201313, India.
| |
Collapse
|
13
|
Guo J, Cao G, Wei S, Han Y, Xu P. Progress in the application of graphene and its derivatives to osteogenesis. Heliyon 2023; 9:e21872. [PMID: 38034743 PMCID: PMC10682167 DOI: 10.1016/j.heliyon.2023.e21872] [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: 05/19/2023] [Revised: 09/13/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
As bone and joint injuries from various causes become increasingly prominent, how to effectively reconstruct and repair bone defects presents a difficult problem for clinicians and researchers. In recent years, graphene and its derivatives have been the subject of growing body of research and have been found to promote the proliferation and osteogenic differentiation of stem cells. This provides a new idea for solving the clinical problem of bone defects. However, as as numerous articles address various aspects and have not been fully systematized, there is an urgent need to classify and summarize them. In this paper, for the first time, the effects of graphene and its derivatives on stem cells in solution, in 2D and 3D structures and in vivo and their possible mechanisms are reviewed, and the cytotoxic effects of graphene and its derivatives were summarized and analyzed. The toxicity of graphene and its derivatives is further reviewed. In addition, we suggest possible future development directions of graphene and its derivatives in bone tissue engineering applications to provide a reference for further clinical application.
Collapse
Affiliation(s)
- Jianbin Guo
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Guihua Cao
- Department of Geriatrics, The First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Song Wei
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yisheng Han
- Department of Orthopedics, The First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Peng Xu
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Fan Z, Liu H, Ding Z, Xiao L, Lu Q, Kaplan DL. Simulation of Cortical and Cancellous Bone to Accelerate Tissue Regeneration. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2301839. [PMID: 37601745 PMCID: PMC10437128 DOI: 10.1002/adfm.202301839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 08/22/2023]
Abstract
Different tissues have complex anisotropic structures to support biological functions. Mimicking these complex structures in vitro remains a challenge in biomaterials designs in support of tissue regeneration. Here, inspired by different types of silk nanofibers, a composite materials strategy was pursued towards this challenge. A combination of fabrication methods was utilized to achieve separate control of amorphous and beta-sheet rich silk nanofibers in the same solution. Aqueous solutions containing these two structural types of silk nanofibers were then simultaneously treated with an electric field and with ethylene glycol diglycidyl ether (EGDE). Under these conditions, the beta-sheet rich silk nanofibers in the mixture responded to the electric field while the amorphous nanofibers were active in the crosslinking process with the EGDE. As a result, cryogels with anisotropic structures were prepared, including mimics for cortical- and cancellous-like bone biomaterials as a complex osteoinductive niche. In vitro studies revealed that mechanical cues of the cryogels induced osteodifferentiation of stem cells while the anisotropy inside the cryogels influenced immune reactions of macrophages. These bioactive cryogels also stimulated improved bone regeneration in vivo through modulation of inflammation, angiogenesis and osteogenesis responses, suggesting an effective strategy to develop bioactive matrices with complex anisotropic structures beneficial to tissue regeneration.
Collapse
Affiliation(s)
- Zhihai Fan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215000, People’s Republic of China
| | - Hongxiang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215000, People’s Republic of China
| | - Zhaozhao Ding
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People’s Republic of China
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - Liying Xiao
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People’s Republic of China
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - Qiang Lu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People’s Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
15
|
Bashiri Z, Rajabi Fomeshi M, Ghasemi Hamidabadi H, Jafari D, Alizadeh S, Nazm Bojnordi M, Orive G, Dolatshahi-Pirouz A, Zahiri M, Reis RL, Kundu SC, Gholipourmalekabadi M. 3D-printed placental-derived bioinks for skin tissue regeneration with improved angiogenesis and wound healing properties. Mater Today Bio 2023; 20:100666. [PMID: 37273796 PMCID: PMC10239019 DOI: 10.1016/j.mtbio.2023.100666] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 06/06/2023] Open
Abstract
Extracellular matrix (ECM)-based bioinks has attracted much attention in recent years for 3D printing of native-like tissue constructs. Due to organ unavailability, human placental ECM can be an alternative source for the construction of 3D print composite scaffolds for the treatment of deep wounds. In this study, we use different concentrations (1.5%, 3% and 5%w/v) of ECM derived from the placenta, sodium-alginate and gelatin to prepare a printable bioink biomimicking natural skin. The printed hydrogels' morphology, physical structure, mechanical behavior, biocompatibility, and angiogenic property are investigated. The optimized ECM (5%w/v) 3D printed scaffold is applied on full-thickness wounds created in a mouse model. Due to their unique native-like structure, the ECM-based scaffolds provide a non-cytotoxic microenvironment for cell adhesion, infiltration, angiogenesis, and proliferation. In contrast, they do not show any sign of immune response to the host. Notably, the biodegradation, swelling rate, mechanical property, cell adhesion and angiogenesis properties increase with the increase of ECM concentrations in the construct. The ECM 3D printed scaffold implanted into deep wounds increases granulation tissue formation, angiogenesis, and re-epithelialization due to the presence of ECM components in the construct, when compared with printed scaffold with no ECM and no treatment wound. Overall, our findings demonstrate that the 5% ECM 3D scaffold supports the best deep wound regeneration in vivo, produces a skin replacement with a cellular structure comparable to native skin.
Collapse
Affiliation(s)
- Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Motahareh Rajabi Fomeshi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sanaz Alizadeh
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Nazm Bojnordi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01009, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029, Madrid, Spain
- University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007, Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore, 169856, Singapore
| | | | - Maria Zahiri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Department of Anatomical Sciences, School of Medical Sciences, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, Portugal
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Adam H, Gopinath SCB, Md Arshad MK, Adam T, Parmin NA, Husein I, Hashim U. An update on pathogenesis and clinical scenario for Parkinson's disease: diagnosis and treatment. 3 Biotech 2023; 13:142. [PMID: 37124989 PMCID: PMC10134733 DOI: 10.1007/s13205-023-03553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
In severe cases, Parkinson's disease causes uncontrolled movements known as motor symptoms such as dystonia, rigidity, bradykinesia, and tremors. Parkinson's disease also causes non-motor symptoms such as insomnia, constipation, depression and hysteria. Disruption of dopaminergic and non-dopaminergic neural networks in the substantia nigra pars compacta is a major cause of motor symptoms in Parkinson's disease. Furthermore, due to the difficulty of clinical diagnosis of Parkinson's disease, it is often misdiagnosed, highlighting the need for better methods of detection. Treatment of Parkinson's disease is also complicated due to the difficulties of medications passing across the blood-brain barrier. Moreover, the conventional methods fail to solve the aforementioned issues. As a result, new methods are needed to detect and treat Parkinson's disease. Improved diagnosis and treatment of Parkinson's disease can help avoid some of its devastating symptoms. This review explores how nanotechnology platforms, such as nanobiosensors and nanomedicine, have improved Parkinson's disease detection and treatment. Nanobiosensors integrate science and engineering principles to detect Parkinson's disease. The main advantages are their low cost, portability, and quick and precise analysis. Moreover, nanotechnology can transport medications in the form of nanoparticles across the blood-brain barrier. However, because nanobiosensors are a novel technology, their use in biological systems is limited. Nanobiosensors have the potential to disrupt cell metabolism and homeostasis, changing cellular molecular profiles and making it difficult to distinguish sensor-induced artifacts from fundamental biological phenomena. In the treatment of Parkinson's disease, nanoparticles, on the other hand, produce neurotoxicity, which is a challenge in the treatment of Parkinson's disease. Techniques must be developed to distinguish sensor-induced artifacts from fundamental biological phenomena and to reduce the neurotoxicity caused by nanoparticles.
Collapse
Affiliation(s)
- Hussaini Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
| | - Subash C. B. Gopinath
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Arau, 02600 Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, Arau, 02600 Perlis, Malaysia
| | - M. K. Md Arshad
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, Arau, 02600 Perlis, Malaysia
| | - Tijjani Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, Arau, 02600 Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, Arau, 02600 Perlis, Malaysia
| | - N. A. Parmin
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
| | - Irzaman Husein
- Department of Physics, Faculty of Mathematics and Natural Sciences, IPB University, Bogor-Indonesia, Indonesia
| | - Uda Hashim
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, 01000 Perlis, Malaysia
| |
Collapse
|
17
|
Almeida GHDR, Iglesia RP, Rinaldi JDC, Murai MK, Calomeno CVAQ, da Silva Junior LN, Horvath-Pereira BDO, Pinho LBM, Miglino MA, Carreira ACO. Current Trends on Bioengineering Approaches for Ovarian Microenvironment Reconstruction. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36355603 DOI: 10.1089/ten.teb.2022.0171] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian tissue has a unique microarchitecture and a complex cellular and molecular dynamics that are essential for follicular survival and development. Due to this great complexity, several factors may lead to ovarian insufficiency, and therefore to systemic metabolic disorders and female infertility. Techniques currently used in the reproductive clinic such as oocyte cryopreservation or even ovarian tissue transplant, although effective, have several limitations, which impair their wide application. In this scenario, mimetic ovarian tissue reconstruction comes as an innovative alternative to develop new methodologies for germ cells preservation and ovarian functions restoration. The ovarian extracellular matrix (ECM) is crucial for oocyte viability maintenance, once it acts actively in folliculogenesis. One of the key components of ovarian bioengineering is biomaterials application that mimics ECM and provides conditions for cell anchorage, proliferation, and differentiation. Therefore, this review aims at describing ovarian tissue engineering approaches and listing the main limitations of current methods for preservation and reestablishment of ovarian fertility. In addition, we describe the main elements that structure this study field, highlighting the main advances and the challenges to overcome to develop innovative methodologies to be applied in reproductive medicine. Impact Statement This review presents the main advances in the application of tissue bioengineering in the ovarian tissue reconstruction to develop innovative solutions for ovarian fertility reestablishment.
Collapse
Affiliation(s)
| | - Rebeca Piatniczka Iglesia
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mikaelly Kiemy Murai
- Department of Morphological Sciences, State University of Maringa, Maringá, Brazil
| | | | | | | | - Letícia Beatriz Mazo Pinho
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Center of Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
| |
Collapse
|
18
|
Rouleau N, Murugan NJ, Kaplan DL. Functional bioengineered models of the central nervous system. NATURE REVIEWS BIOENGINEERING 2023; 1:252-270. [PMID: 37064657 PMCID: PMC9903289 DOI: 10.1038/s44222-023-00027-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/10/2023]
Abstract
The functional complexity of the central nervous system (CNS) is unparalleled in living organisms. Its nested cells, circuits and networks encode memories, move bodies and generate experiences. Neural tissues can be engineered to assemble model systems that recapitulate essential features of the CNS and to investigate neurodevelopment, delineate pathophysiology, improve regeneration and accelerate drug discovery. In this Review, we discuss essential structure-function relationships of the CNS and examine materials and design considerations, including composition, scale, complexity and maturation, of cell biology-based and engineering-based CNS models. We highlight region-specific CNS models that can emulate functions of the cerebral cortex, hippocampus, spinal cord, neural-X interfaces and other regions, and investigate a range of applications for CNS models, including fundamental and clinical research. We conclude with an outlook to future possibilities of CNS models, highlighting the engineering challenges that remain to be overcome.
Collapse
Affiliation(s)
- Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario Canada
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| | - Nirosha J. Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario Canada
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| |
Collapse
|
19
|
Wang Q, Li M, Cui T, Wu R, Guo F, Fu M, Zhu Y, Yang C, Chen B, Sun G. A Novel Zwitterionic Hydrogel Incorporated with Graphene Oxide for Bone Tissue Engineering: Synthesis, Characterization, and Promotion of Osteogenic Differentiation of Bone Mesenchymal Stem Cells. Int J Mol Sci 2023; 24:ijms24032691. [PMID: 36769013 PMCID: PMC9916718 DOI: 10.3390/ijms24032691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Zwitterionic materials are widely applied in the biomedical field due to their excellent antimicrobial, non-cytotoxicity, and antifouling properties but have never been applied in bone tissue engineering. In this study, we synthesized a novel zwitterionic hydrogel incorporated with graphene oxide (GO) using maleic anhydride (MA) as a cross-linking agent by grafted L-cysteine (L-Cys) as the zwitterionic material on maleilated chitosan via click chemistry. The composition and each reaction procedure of the novel zwitterionic hydrogel were characterized via X-ray diffraction (XRD) and Fourier transformed infrared spectroscopy (FT-IR), while the morphology was imaged by scanning electron microscope (SEM). In vitro cell studies, CCK-8 and live/dead assay, alkaline phosphatase activity, W-B, and qRT-CR tests showed zwitterionic hydrogel incorporated with GO remarkably enhanced the osteogenic differentiation of bone mesenchymal stem cells (BMSCs); it is dose-dependent, and 2 mg/mL GO is the optimum concentration. In vivo tests also indicated the same results. Hence, these results suggested the novel zwitterionic hydrogel exhibited porous characteristics similar to natural bone tissue. In conclusion, the zwitterionic scaffold has highly biocompatible and mechanical properties. When GO was incorporated in this zwitterionic scaffold, the zwitterionic scaffold slows down the release rate and reduces the cytotoxicity of GO. Zwitterions and GO synergistically promote the proliferation and osteogenic differentiation of rBMSCs in vivo and in vitro. The optimal concentration is 2 mg/mL GO.
Collapse
Affiliation(s)
- Qidong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Meng Li
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Tianming Cui
- Shanghai Research Institute for Intelligent Autonomous Systems, Tongji University, Shanghai 200092, China
| | - Rui Wu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Fangfang Guo
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Mei Fu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Yuqian Zhu
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chensong Yang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Bingdi Chen
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
- Correspondence: (B.C.); (G.S.)
| | - Guixin Sun
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Correspondence: (B.C.); (G.S.)
| |
Collapse
|
20
|
Cheng H, Yu Q, Chen Q, Feng L, Zhao W, Zhao C. Biomass-derived ultrafast cross-linked hydrogels with double dynamic bonds for hemostasis and wound healing. Biomater Sci 2023; 11:931-948. [PMID: 36537166 DOI: 10.1039/d2bm00907b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Developing novel hemostatic materials with accelerating wound healing functions has raised widespread attention recently. To adapt to irregular and incompressible wounds, we fabricated a series of biomass-derived ultrafast cross-linked adhesive hydrogels with adjustable gelation time and injectable properties through Schiff-base and ionic coordinate bonds among catechol-conjugated gelatin (GelDA), dialdehyde cellulose nanocrystals (DACNCs), calcium ions (Ca2+) and ferric iron (Fe3+). The fast-gelling hydrogels possess adjustable gelation time and mechanical properties by altering the contents of DACNCs and Fe3+. With double-dynamic-bond crosslinking, the hydrogels are endowed with the desired self-healing and injectable performance compared to gelatin-based hydrogels without DACNCs. Additionally, the hydrogels present enhanced adhesiveness, NIR responsiveness and antibacterial activity with the introduction of catechol groups and the formation of catechol-Fe complexes. Both in vitro and in vivo hemostatic assays and degradation experiments confirm that the hydrogels achieve rapid hemostasis and display fantastic biodegradability. As demonstrated by a rat full-thickness skin defect model, the hydrogels with multifunctionality remarkably accelerate the regeneration of wound tissues. Thus, the ultrafast cross-linked hydrogels are potentially valuable as hemostatic materials for wound healing applications in the biomedical field.
Collapse
Affiliation(s)
- Huitong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China.
| | - Qiao Yu
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu 610207, China.,Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Qin Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China.
| | - Lan Feng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China.
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China. .,Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China. .,College of Chemical Engineering, Sichuan University, Chengdu, 610065, China.,Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
21
|
Ban G, Hou Y, Shen Z, Jia J, Chai L, Ma C. Potential Biomedical Limitations of Graphene Nanomaterials. Int J Nanomedicine 2023; 18:1695-1708. [PMID: 37020689 PMCID: PMC10069520 DOI: 10.2147/ijn.s402954] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Graphene-family nanomaterials (GFNs) possess mechanical stiffness, optical properties, and biocompatibility making them promising materials for biomedical applications. However, to realize the potential of graphene in biomedicine, it must overcome several challenges that arise when it enters the body's circulatory system. Current research focuses on the development of tumor-targeting devices using graphene, but GFNs accumulated in different tissues and cells through different pathways, which can cause toxic reactions leading to cell apoptosis and body dysfunction when the accumulated amount exceeds a certain limit. In addition, as a foreign substance, graphene can induce complex inflammatory reactions with immune cells and inflammatory factors, potentially enhancing or impairing the body's immune function. This review discusses the biomedical applications of graphene, the effects of graphene materials on human immune function, and the biotoxicity of graphene materials.
Collapse
Affiliation(s)
- Ge Ban
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
- Correspondence: Ge Ban, Email
| | - Yingze Hou
- Clinical Medical College, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Zhean Shen
- Department of Biomedical Research, Research and Innovation Center, Xinjiang Institute of Technology, Xinjiang, 843100, People’s Republic of China
| | - Jingjing Jia
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Lei Chai
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Chongyang Ma
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| |
Collapse
|
22
|
Li Y, Yang L, Hou Y, Zhang Z, Chen M, Wang M, Liu J, Wang J, Zhao Z, Xie C, Lu X. Polydopamine-mediated graphene oxide and nanohydroxyapatite-incorporated conductive scaffold with an immunomodulatory ability accelerates periodontal bone regeneration in diabetes. Bioact Mater 2022; 18:213-227. [PMID: 35387166 PMCID: PMC8961429 DOI: 10.1016/j.bioactmat.2022.03.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022] Open
Abstract
Regenerating periodontal bone tissues in the aggravated inflammatory periodontal microenvironment under diabetic conditions is a great challenge. Here, a polydopamine-mediated graphene oxide (PGO) and hydroxyapatite nanoparticle (PHA)-incorporated conductive alginate/gelatin (AG) scaffold is developed to accelerate periodontal bone regeneration by modulating the diabetic inflammatory microenvironment. PHA confers the scaffold with osteoinductivity and PGO provides a conductive pathway for the scaffold. The conductive scaffold promotes bone regeneration by transferring endogenous electrical signals to cells and activating Ca2+ channels. Moreover, the scaffold with polydopamine-mediated nanomaterials has a reactive oxygen species (ROS)-scavenging ability and anti-inflammatory activity. It also exhibits an immunomodulatory ability that suppresses M1 macrophage polarization and activates M2 macrophages to secrete osteogenesis-related cytokines by mediating glycolytic and RhoA/ROCK pathways in macrophages. The scaffold induces excellent bone regeneration in periodontal bone defects of diabetic rats because of the synergistic effects of good conductive, ROS-scavenging, anti-inflammatory, and immunomodulatory abilities. This study provides fundamental insights into the synergistical effects of conductivity, osteoinductivity, and immunomodulatory abilities on bone regeneration and offers a novel strategy to design immunomodulatory biomaterials for treatment of immune-related diseases and tissue regeneration. The conductive PGO-PHA-AG scaffold can activate Ca2+ channels. •The PGO-PHA-AG scaffold had ROS-scavenging and anti-inflammatory activities. •The scaffold exhibited an immunomodulatory ability. •The scaffold induced excellent periodontal bone regeneration in diabetes.
Collapse
Affiliation(s)
- Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Yang
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yue Hou
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Maoxia Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jin Liu
- Lab for Aging Research and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Chaoming Xie
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| |
Collapse
|
23
|
Yazdanian M, Alam M, Abbasi K, Rahbar M, Farjood A, Tahmasebi E, Tebyaniyan H, Ranjbar R, Hesam Arefi A. Synthetic materials in craniofacial regenerative medicine: A comprehensive overview. Front Bioeng Biotechnol 2022; 10:987195. [PMID: 36440445 PMCID: PMC9681815 DOI: 10.3389/fbioe.2022.987195] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
The state-of-the-art approach to regenerating different tissues and organs is tissue engineering which includes the three parts of stem cells (SCs), scaffolds, and growth factors. Cellular behaviors such as propagation, differentiation, and assembling the extracellular matrix (ECM) are influenced by the cell's microenvironment. Imitating the cell's natural environment, such as scaffolds, is vital to create appropriate tissue. Craniofacial tissue engineering refers to regenerating tissues found in the brain and the face parts such as bone, muscle, and artery. More biocompatible and biodegradable scaffolds are more commensurate with tissue remodeling and more appropriate for cell culture, signaling, and adhesion. Synthetic materials play significant roles and have become more prevalent in medical applications. They have also been used in different forms for producing a microenvironment as ECM for cells. Synthetic scaffolds may be comprised of polymers, bioceramics, or hybrids of natural/synthetic materials. Synthetic scaffolds have produced ECM-like materials that can properly mimic and regulate the tissue microenvironment's physical, mechanical, chemical, and biological properties, manage adherence of biomolecules and adjust the material's degradability. The present review article is focused on synthetic materials used in craniofacial tissue engineering in recent decades.
Collapse
Affiliation(s)
- Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Rahbar
- Department of Restorative Dentistry, School of Dentistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amin Farjood
- Orthodontic Department, Dental School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Arian Hesam Arefi
- Dental Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
24
|
Zhang L, Oudeng G, Wen F, Liao G. Recent advances in near-infrared-II hollow nanoplatforms for photothermal-based cancer treatment. Biomater Res 2022; 26:61. [PMID: 36348441 PMCID: PMC9641873 DOI: 10.1186/s40824-022-00308-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/16/2022] [Indexed: 11/10/2022] Open
Abstract
Near-infrared-II (NIR-II, 1000–1700 nm) light-triggered photothermal therapy (PTT) has been regarded as a promising candidate for cancer treatment, but PTT alone often fails to achieve satisfactory curative outcomes. Hollow nanoplatforms prove to be attractive in the biomedical field owing to the merits including good biocompatibility, intrinsic physical-chemical nature and unique hollow structures, etc. On one hand, hollow nanoplatforms themselves can be NIR-II photothermal agents (PTAs), the cavities of which are able to carry diverse therapeutic units to realize multi-modal therapies. On the other hand, NIR-II PTAs are capable of decorating on the surface to combine with the functions of components encapsulated inside the hollow nanoplatforms for synergistic cancer treatment. Notably, PTAs generally can serve as good photoacoustic imaging (PAI) contrast agents (CAs), which means such kind of hollow nanoplatforms are also expected to be multifunctional all-in-one nanotheranostics. In this review, the recent advances of NIR-II hollow nanoplatforms for single-modal PTT, dual-modal PTT/photodynamic therapy (PDT), PTT/chemotherapy, PTT/catalytic therapy and PTT/gas therapy as well as multi-modal PTT/chemodynamic therapy (CDT)/chemotherapy, PTT/chemo/gene therapy and PTT/PDT/CDT/starvation therapy (ST)/immunotherapy are summarized for the first time. Before these, the typical synthetic strategies for hollow structures are presented, and lastly, potential challenges and perspectives related to these novel paradigms for future research and clinical translation are discussed.
Collapse
|
25
|
3D-Printing Graphene Scaffolds for Bone Tissue Engineering. Pharmaceutics 2022; 14:pharmaceutics14091834. [PMID: 36145582 PMCID: PMC9503344 DOI: 10.3390/pharmaceutics14091834] [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: 07/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Graphene-based materials have recently gained attention for regenerating various tissue defects including bone, nerve, cartilage, and muscle. Even though the potential of graphene-based biomaterials has been realized in tissue engineering, there are significantly many more studies reporting in vitro and in vivo data in bone tissue engineering. Graphene constructs have mainly been studied as two-dimensional (2D) substrates when biological organs are within a three-dimensional (3D) environment. Therefore, developing 3D graphene scaffolds is the next clinical standard, yet most have been fabricated as foams which limit control of consistent morphology and porosity. To overcome this issue, 3D-printing technology is revolutionizing tissue engineering, due to its speed, accuracy, reproducibility, and overall ability to personalize treatment whereby scaffolds are printed to the exact dimensions of a tissue defect. Even though various 3D-printing techniques are available, practical applications of 3D-printed graphene scaffolds are still limited. This can be attributed to variations associated with fabrication of graphene derivatives, leading to variations in cell response. This review summarizes selected works describing the different fabrication techniques for 3D scaffolds, the novelty of graphene materials, and the use of 3D-printed scaffolds of graphene-based nanoparticles for bone tissue engineering.
Collapse
|
26
|
Fe3O4/Graphene-Based Nanotheranostics for Bimodal Magnetic Resonance/Fluorescence Imaging and Cancer Therapy. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-022-02457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
27
|
Chae SY, Park R, Hong SW. Surface-mediated high antioxidant and anti-inflammatory effects of astaxanthin-loaded ultrathin graphene oxide film that inhibits the overproduction of intracellular reactive oxygen species. Biomater Res 2022; 26:30. [PMID: 35794645 PMCID: PMC9258176 DOI: 10.1186/s40824-022-00276-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/10/2022] [Indexed: 12/20/2022] Open
Abstract
Background Astaxanthin (AST) is known as a powerful antioxidant that affects the removal of active oxygen and inhibits the production of lipid peroxide caused by ultraviolet light. However, it is easily decomposed by heat or light during production and storage because of the unsaturated compound nature with a structural double bond. The activity of AST can be reduced and lose its antioxidant capability. Graphene oxide (GO) is an ultrathin nanomaterial produced by oxidizing layered graphite. The chemical combination of AST with GO can improve the dispersion properties to maintain structural stability and antioxidant activity because of the tightly bonded functionalized GO surface. Methods Layered GO films were used as nanocarriers for the AST molecule, which was produced via flow-enabled self-assembly and subsequent controlled solution deposition of RGD peptide and AST molecules. Synthesis of the GO-AST complex was also carried out for the optimized concentration. The characterization of prepared materials was analyzed through transmission electron microscopy (TEM), scanning electron microscope (SEM), Fourier-transform infrared spectroscopy (FT-IR), atomic force microscope (AFM), and Raman spectroscopy. Antioxidant activity was tested by 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2.2-diphenyl-1-picrylhydrazyl (DPPH) assays. The antibacterial effect and antioxidant effects were monitored for the ultrathin GO/RGD/AST Film. Further, reactive oxygen species (ROS) assay was used to evaluate the anti-inflammatory effects on L-929 fibroblasts. Results Cotreatment of GO-AST solution demonstrated a high antioxidant combined effect with a high ABTS and DPPH radicals scavenging activity. The GO/RGD/AST film was produced by the self-assembly process exhibited excellent antibacterial effects based on physicochemical damage against E. coli and S. aureus. In addition, the GO/RGD/AST film inhibited H2O2-induced intracellular ROS, suppressed the toxicity of lipopolysaccharide (LPS)-induced cells, and restored it, thereby exhibiting strong antioxidant and anti-inflammatory effects. Conclusion As GO nanocarrier-assisted AST exerted promising antioxidant and antibacterial reactions, presented a new concept to expand basic research into the field of tissue engineering. Supplementary Information The online version contains supplementary material available at 10.1186/s40824-022-00276-4.
Collapse
|
28
|
Jeong Y, Jin M, Kim KS, Na K. Biocompatible carbonized iodine-doped dots for contrast-enhanced CT imaging. Biomater Res 2022; 26:27. [PMID: 35752823 PMCID: PMC9233767 DOI: 10.1186/s40824-022-00277-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Computed tomography (CT) imaging has been widely used for the diagnosis and surveillance of diseases. Although CT is attracting attention due to its reasonable price, short scan time, and excellent diagnostic ability, there are severe drawbacks of conventional CT contrast agents, such as low sensitivity, serious toxicity, and complicated synthesis process. Herein, we describe iodine-doped carbon dots (IDC) for enhancing the abilities of CT contrast agents. Method IDC was synthesized by one-pot hydrothermal synthesis for 4 h at 180 ℃ and analysis of its structure and size distribution with UV–Vis, XPS, FT-IR, NMR, TEM, and DLS. Furthermore, the CT values of IDC were calculated and compared with those of conventional CT contrast agents (Iohexol), and the in vitro and in vivo toxicities of IDC were determined to prove their safety. Results IDC showed improved CT contrast enhancement compared to iohexol. The biocompatibility of the IDC was verified via cytotoxicity tests, hemolysis assays, chemical analysis, and histological analysis. The osmotic pressure of IDC was lower than that of iohexol, resulting in no dilution-induced contrast decrease in plasma. Conclusion Based on these results, the remarkable CT contrast enhancement and biocompatibility of IDC can be used as an effective CT contrast agent for the diagnosis of various diseases compared with conventional CT contrast agents. Supplementary Information The online version contains supplementary material available at 10.1186/s40824-022-00277-3.
Collapse
Affiliation(s)
- Yohan Jeong
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea.,Department of Research and Developmnet, SML Genetree, Seoul, 06741, Republic of Korea.,Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea
| | - Minyoung Jin
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea.,Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea. .,Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi do, 14662, Republic of Korea.
| |
Collapse
|
29
|
Sahebalzamani M, Ziminska M, McCarthy HO, Levingstone TJ, Dunne NJ, Hamilton AR. Advancing bone tissue engineering one layer at a time: a layer-by-layer assembly approach to 3D bone scaffold materials. Biomater Sci 2022; 10:2734-2758. [PMID: 35438692 DOI: 10.1039/d1bm01756j] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The layer-by-layer (LbL) assembly technique has shown excellent potential in tissue engineering applications. The technique is mainly based on electrostatic attraction and involves the sequential adsorption of oppositely charged electrolyte complexes onto a substrate, resulting in uniform single layers that can be rapidly deposited to form nanolayer films. LbL has attracted significant attention as a coating technique due to it being a convenient and affordable fabrication method capable of achieving a wide range of biomaterial coatings while keeping the main biofunctionality of the substrate materials. One promising application is the use of nanolayer films fabricated by LbL assembly in the development of 3-dimensional (3D) bone scaffolds for bone repair and regeneration. Due to their versatility, nanoscale films offer an exciting opportunity for tailoring surface and bulk property modification of implants for osseous defect therapies. This review article discusses the state of the art of the LbL assembly technique, and the properties and functions of LbL-assembled films for engineered bone scaffold application, combination of multilayers for multifunctional coatings and recent advancements in the application of LbL assembly in bone tissue engineering. The recent decade has seen tremendous advances in the promising developments of LbL film systems and their impact on cell interaction and tissue repair. A deep understanding of the cell behaviour and biomaterial interaction for the further development of new generations of LbL films for tissue engineering are the most important targets for biomaterial research in the field. While there is still much to learn about the biological and physicochemical interactions at the interface of nano-surface coated scaffolds and biological systems, we provide a conceptual review to further progress in the LbL approach to 3D bone scaffold materials and inform the future of LbL development in bone tissue engineering.
Collapse
Affiliation(s)
- MohammadAli Sahebalzamani
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland. .,Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland.
| | - Monika Ziminska
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK.
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK. .,School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland. .,Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland. .,Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland.,Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland.,Biodesign Europe, Dublin City University, Dublin 9, Ireland
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland. .,Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland. .,School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK. .,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, Ireland.,Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland.,Biodesign Europe, Dublin City University, Dublin 9, Ireland
| | - Andrew R Hamilton
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
30
|
Ricci A, Cataldi A, Zara S, Gallorini M. Graphene-Oxide-Enriched Biomaterials: A Focus on Osteo and Chondroinductive Properties and Immunomodulation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2229. [PMID: 35329679 PMCID: PMC8955105 DOI: 10.3390/ma15062229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Abstract
Due to its exceptional physical properties, such as high electronic conductivity, good thermal stability, excellent mechanical strength, and chemical versatility, graphene has sparked a lot of interest in the scientific community for various applications. It has therefore been employed as an antibacterial agent, in photothermal therapy (PTT) and biosensors, in gene delivery systems, and in tissue engineering for regenerative purposes. Since it was first discovered in 1947, different graphene derivatives have been synthetized from pristine graphene. The most adaptable derivate is graphene oxide (GO). Owing to different functional groups, the amphiphilic structure of GO can interact with cells and exogenous or endogenous growth/differentiation factors, allowing cell adhesion, growth, and differentiation. When GO is used as a coating for scaffolds and nanomaterials, it has been found to enhance bone, chondrogenic, cardiac, neuronal, and skin regeneration. This review focuses on the applications of graphene-based materials, in particular GO, as a coating for scaffolds in bone and chondrogenic tissue engineering and summarizes the most recent findings. Moreover, novel developments on the immunomodulatory properties of GO are reported.
Collapse
Affiliation(s)
| | | | | | - Marialucia Gallorini
- Department of Pharmacy, “G. d'Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (A.R.); (A.C.); (S.Z.)
| |
Collapse
|
31
|
Tural B, Ertaş E, Tural S. Investigation of the arsenic(V) retention performance of the nano-sorbent (M-TACA) synthesized by click chemistry. J DISPER SCI TECHNOL 2022. [DOI: 10.1080/01932691.2022.2040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Bilsen Tural
- Department of Chemistry, Faculty of Education, Dicle University, Diyarbakir, Turkey
| | - Erdal Ertaş
- Department of Chemistry, Faculty of Education, Dicle University, Diyarbakir, Turkey
| | - Servet Tural
- Department of Chemistry, Faculty of Education, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
32
|
Iron-doped calcium phytate nanoparticles as a bio-responsive contrast agent in 1H/ 31P magnetic resonance imaging. Sci Rep 2022; 12:2118. [PMID: 35136162 PMCID: PMC8826874 DOI: 10.1038/s41598-022-06125-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/19/2022] [Indexed: 12/29/2022] Open
Abstract
We present the MR properties of a novel bio-responsive phosphorus probe doped with iron for dual proton and phosphorus magnetic resonance imaging (1H/31P-MRI), which provide simultaneously complementary information. The probes consist of non-toxic biodegradable calcium phytate (CaIP6) nanoparticles doped with different amounts of cleavable paramagnetic Fe3+ ions. Phosphorus atoms in the phytate structure delivered an efficient 31P-MR signal, with iron ions altering MR contrast for both 1H and 31P-MR. The coordinated paramagnetic Fe3+ ions broadened the 31P-MR signal spectral line due to the short T2 relaxation time, resulting in more hypointense signal. However, when Fe3+ was decomplexed from the probe, relaxation times were prolonged. As a result of iron release, intensity of 1H-MR, as well as the 31P-MR signal increase. These 1H and 31P-MR dual signals triggered by iron decomplexation may have been attributable to biochemical changes in the environment with strong iron chelators, such as bacterial siderophore (deferoxamine). Analysing MR signal alternations as a proof-of-principle on a phantom at a 4.7 T magnetic field, we found that iron presence influenced 1H and 31P signals and signal recovery via iron chelation using deferoxamine.
Collapse
|
33
|
Um W, Gupta A, Song SH, Kim CH, Park JH. Biomaterials as Antigen Delivery Carrier for Cancer Immunotherapy. Macromol Res 2022. [DOI: 10.1007/s13233-021-9095-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Park R, Yoon JW, Lee JH, Hong SW, Kim JH. Phenotypic change of mesenchymal stem cells into smooth muscle cells regulated by dynamic cell-surface interactions on patterned arrays of ultrathin graphene oxide substrates. J Nanobiotechnology 2022; 20:17. [PMID: 34983551 PMCID: PMC8725258 DOI: 10.1186/s12951-021-01225-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
The topographical interface of the extracellular environment has been appreciated as a principal biophysical regulator for modulating cell functions, such as adhesion, migration, proliferation, and differentiation. Despite the existed approaches that use two-dimensional nanomaterials to provide beneficial effects, opportunities evaluating their impact on stem cells remain open to elicit unprecedented cellular responses. Herein, we report an ultrathin cell-culture platform with potential-responsive nanoscale biointerfaces for monitoring mesenchymal stem cells (MSCs). We designed an intriguing nanostructured array through self-assembly of graphene oxide sheets and subsequent lithographical patterning method to produce chemophysically defined regions. MSCs cultured on anisotropic micro/nanoscale patterned substrate were spontaneously organized in a highly ordered configuration mainly due to the cell-repellent interactions. Moreover, the spatially aligned MSCs were spontaneously differentiated into smooth muscle cells upon the specific crosstalk between cells. This work provides a robust strategy for directing stem cells and differentiation, which can be utilized as a potential cell culture platform to understand cell-substrate or cell-cell interactions, further developing tissue repair and stem cell-based therapies.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, 46241, Busan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea
| | - Jin-Ho Lee
- Department of Biomedical Convergence Engineering, Pusan National University, 50612, Yangsan, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, 46241, Busan, Republic of Korea.
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea.
| |
Collapse
|
35
|
Choi D, Choi M, Jeong H, Heo J, Kim T, Park S, Jin Y, Lee S, Hong J. Co-existing "spear-and-shield" air filter: Anchoring proteinaceous pathogen and self-sterilized nanocoating for combating viral pandemic. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 426:130763. [PMID: 34131388 PMCID: PMC8192840 DOI: 10.1016/j.cej.2021.130763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/23/2021] [Accepted: 06/05/2021] [Indexed: 06/12/2023]
Abstract
Infectious pollutants bioaerosols can threaten human public health. In particular, the indoor environment provides a unique exposure situation to induce infection through airborne transmission like SARS-CoV-2. To prevent the infection from spreading, personal protective equipment or indoor air purification is necessary. However, it has been discovered that the conventional filter can become contaminated by pathogen-containing aerosols, meaning that advanced filtering and self-sterilization systems are required. Here, we fabricate a multilayered nanocoating around the fabric using laponite (LAP) with Cu2+ ions (LAP-Cu2+ nanocoating) two contradictory functions in one system: trapping proteinaceous pathogens and antibacterial effect. Due to the strong LAP-protein interaction, albumin and spike protein (S-protein) are trapped into the fabric when proteins are sprayed using a nebulizer. The protein-blocking performance of the nanocoated fabric is 9.55-fold higher than bare fabric. These trapping capacities are retained after rinsing and repeated adsorption cycles, showing reproducibility for air filtration. Even though the protein-binding occurred, the LAP-Cu2+ fabric indicates antibacterial effect. LAP-Cu2+ fabric has an equivalent air and water transmittance rate to that of bare fabric with a stability under physiological environment. Therefore, given its excellent "Spear-and-shield" functions, the proposed LAP-Cu2+ fabric shows great potential for use in filter and masks during the viral pandemic.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Moonhyun Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyejoong Jeong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jiwoong Heo
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Taihyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sohyeon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Youngho Jin
- Agency for Defense Development, Daejeon 34186, Republic of Korea
| | - Sangmin Lee
- School of Mechanical Engineering, Chung-ang University, Seoul 06974, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
36
|
de Oliveira BM, Teodoro JBM, Ambrósio JAR, Gonçalves EP, Beltrame M, Cortez Marcolino LM, Pinto JG, Ferreira-Strixino J, Simioni AR. Zinc pthalocyanine loaded poly (lactic acid) nanoparticles by double emulsion methodology for photodynamic therapy against 9 L/LacZ gliosarcoma cells. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:93-109. [PMID: 34517784 DOI: 10.1080/09205063.2021.1980359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Development delivery systems, such as nanoparticles, represent a growing area in biomedical research. Nanoparticles (NP) were prepared using a double-emulsion method to load zinc(II) phthalocyanine (ZnPc). NP were obtained using poly (lactic acid) (PLA). ZnPc is a second generation of photosensitizer used in photodynamic therapy (PDT). ZnPc loaded PLA nanoparticles (NPLA-ZnPc) were prepared by double-emulsion method, characterized and available in cellular culture. The mean nanoparticle size presented particle size was 384.7 ± 84.2 nm with polydispersity index (PDI) of 0.150 ± 0.015, and the encapsulation efficiency was of 83%. The nanoparticle formulations presented negative zeta potential values (-27.5 ± 1.0 mV), explaining their colloidal stability. ZnPc loaded nanoparticles maintain its photophysical behavior after encapsulation. Photosensitizer release from nanoparticles was sustained over 168 h with a biphasic ZnPc release profile. An in vitro phototoxic effect in range of 80% was observed in 9 L/LacZ gliosarcoma cells at laser light doses (10 J cm-2) with 3.0 µg mL-1 of NPLA-ZnPc. All the physical-chemical, photophysical and photobiological measurements performed allow us to conclude that ZnPc loaded PLGA nanoparticles is a promising drug delivery system for PDT.
Collapse
Affiliation(s)
- Benedito Marcio de Oliveira
- Organic Synthesis Laboratory, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Jéssica Beatriz Miranda Teodoro
- Organic Synthesis Laboratory, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | | | - Erika Peterson Gonçalves
- Organic Synthesis Laboratory, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Milton Beltrame
- Organic Synthesis Laboratory, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Luciana Maria Cortez Marcolino
- Laboratory of Photobiology Applied to Health, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Juliana Guerra Pinto
- Laboratory of Photobiology Applied to Health, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Juliana Ferreira-Strixino
- Laboratory of Photobiology Applied to Health, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| | - Andreza Ribeiro Simioni
- Organic Synthesis Laboratory, Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, Brazil
| |
Collapse
|
37
|
Wang J, Harizaj A, Wu Y, Jiang X, Brans T, Fraire JC, Mejía Morales J, De Smedt SC, Tang Z, Xiong R, Braeckmans K. Black phosphorus mediated photoporation: a broad absorption nanoplatform for intracellular delivery of macromolecules. NANOSCALE 2021; 13:17049-17056. [PMID: 34622916 DOI: 10.1039/d1nr05461a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanoparticle-sensitized photoporation for intracellular delivery of external compounds usually relies on the use of spherical gold nanoparticles as sensitizing nanoparticles. As they need stimulation with visible laser light, they are less suited for transfection of cells in thick biological tissues. In this work, we have explored black phosphorus quantum dots (BPQDs) as alternative sensitizing nanoparticles for photoporation with a broad and uniform absorption spectrum from the visible to the near infra-red (NIR) range. We demonstrate that BPQD sensitized photoporation allows efficient intracellular delivery of both siRNA (>80%) and mRNA (>40%) in adherent cells as well as in suspension cells. Cell viability remained high (>80%) irrespective of whether irradiation was performed with visible (532 nm) or near infrared (800 nm) pulsed laser light. Finally, as a proof of concept, we used BPQD sensitized photoporation to deliver macromolecules in cells with thick phantom tissue in the optical path. NIR laser irradiation resulted in only 1.3× reduction in delivery efficiency as compared to photoporation without the phantom gel, while with visible laser light the delivery efficiency was reduced 2×.
Collapse
Affiliation(s)
- Jielin Wang
- School of Physics and Telecommunication Engineering, South China Normal University, Guangzhou, 510006, China
- Guangdong Research Center of Photoelectric Detection Instrument Engineering Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials, Guangzhou, 510006, China
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Aranit Harizaj
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Yongbo Wu
- School of Physics and Telecommunication Engineering, South China Normal University, Guangzhou, 510006, China
- Guangdong Research Center of Photoelectric Detection Instrument Engineering Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials, Guangzhou, 510006, China
| | - Xiaofang Jiang
- School of Physics and Telecommunication Engineering, South China Normal University, Guangzhou, 510006, China
- Guangdong Research Center of Photoelectric Detection Instrument Engineering Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials, Guangzhou, 510006, China
| | - Toon Brans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Julián Mejía Morales
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| | - Zhilie Tang
- School of Physics and Telecommunication Engineering, South China Normal University, Guangzhou, 510006, China
- Guangdong Research Center of Photoelectric Detection Instrument Engineering Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials, Guangzhou, 510006, China
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), International Innovation for Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing 210037, P. R. China
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
- Centre for Advanced Light Microscopy, Ghent University, Belgium
| |
Collapse
|
38
|
Shin S, Lee J, Han J, Li F, Ling D, Park W. Tumor Microenvironment Modulating Functional Nanoparticles for Effective Cancer Treatments. Tissue Eng Regen Med 2021; 19:205-219. [PMID: 34674182 DOI: 10.1007/s13770-021-00403-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the major diseases that threaten human life worldwide. Despite advances in cancer treatment techniques, such as radiation therapy, chemotherapy, targeted therapy, and immunotherapy, it is still difficult to cure cancer because of the resistance mechanism of cancer cells. Current understanding of tumor biology has revealed that resistance to these anticancer therapies is due to the tumor microenvironment (TME) represented by hypoxia, acidity, dense extracellular matrix, and immunosuppression. This review demonstrates the latest strategies for effective cancer treatment using functional nanoparticles that can modulate the TME. Indeed, preclinical studies have shown that functional nanoparticles can effectively modulate the TME to treat refractory cancer. This strategy of using TMEs with controllable functional nanoparticles is expected to maximize cancer treatment efficiency in the future by combining it with various modern cancer therapeutics.
Collapse
Affiliation(s)
- Seungyong Shin
- Department of Biomedical-Chemical Engineering and Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea.,Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea
| | - Jiyoung Lee
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Jieun Han
- Department of Biomedical-Chemical Engineering and Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea.,Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, People's Republic of China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310027, Zhejiang, People's Republic of China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, People's Republic of China.,National Center for Translational Medicine, Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Wooram Park
- Department of Biomedical-Chemical Engineering and Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea. .,Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi, 14662, Republic of Korea.
| |
Collapse
|
39
|
Genetic profiling of human bone marrow and adipose tissue-derived mesenchymal stem cells reveals differences in osteogenic signaling mediated by graphene. J Nanobiotechnology 2021; 19:285. [PMID: 34551771 PMCID: PMC8459567 DOI: 10.1186/s12951-021-01024-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In the last decade, graphene surfaces have consistently supported osteoblast development of stem cells, holding promise as a therapeutic implant for degenerative bone diseases. However, until now no study has specifically examined the genetic changes when stem cells undergo osteogenic differentiation on graphene. RESULTS In this study, we provide a detailed overview of gene expressions when human mesenchymal stem cells (MSCs) derived from either adipose tissue (AD-MSCs) or bone marrow (BM-MSCs), are cultured on graphene. Genetic expressions were measured using osteogenic RT2 profiler PCR arrays and compared either over time (7 or 21 days) or between each cell source at each time point. Genes were categorized as either transcriptional regulation, osteoblast-related, extracellular matrix, cellular adhesion, BMP and SMAD signaling, growth factors, or angiogenic factors. Results showed that both MSC sources cultured on low oxygen graphene surfaces achieved osteogenesis by 21 days and expressed specific osteoblast markers. However, each MSC source cultured on graphene did have genetically different responses. When compared between each other, we found that genes of BM-MSCs were robustly expressed, and more noticeable after 7 days of culturing, suggesting BM-MSCs initiate osteogenesis at an earlier time point than AD-MSCs on graphene. Additionally, we found upregulated angiogenic markers in both MSCs sources, suggesting graphene could simultaneously attract the ingrowth of blood vessels in vivo. Finally, we identified several novel targets, including distal-less homeobox 5 (DLX5) and phosphate-regulating endopeptidase homolog, X-linked (PHEX). CONCLUSIONS Overall, this study shows that graphene genetically supports differentiation of both AD-MSCs and BM-MSCs but may involve different signaling mechanisms to achieve osteogenesis. Data further demonstrates the lack of aberrant signaling due to cell-graphene interaction, strengthening the application of specific form and concentration of graphene nanoparticles in bone tissue engineering.
Collapse
|
40
|
Wu M, Zou L, Jiang L, Zhao Z, Liu J. Osteoinductive and antimicrobial mechanisms of graphene-based materials for enhancing bone tissue engineering. J Tissue Eng Regen Med 2021; 15:915-935. [PMID: 34469046 DOI: 10.1002/term.3239] [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: 03/01/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023]
Abstract
Graphene-based materials (GMs) have great application prospects in bone tissue engineering due to their osteoinductive ability and antimicrobial activity. GMs induce osteogenic differentiation through several mechanisms and pathways in bone tissue engineering. First of all, the surface and high hardness of the porous folds of graphene or graphene oxide (GO) can generate mechanical stimulation to initiate a cascade of reactions that promote osteogenic differentiation without any chemical inducers. In addition, change of the extracellular matrix (ECM), regulation of macrophage polarization, the oncostatin M (OSM) signaling pathway, the MAPK signaling pathway, the BMP signaling pathway, the Wnt/β-catenin signaling pathway, and other pathways are involved in GMs' regulation of osteogenesis. In bone tissue engineering, GMs prevent the formation of microbial biofilms mainly through preventing microbial adhesion and killing them. The former is mainly achieved by reducing surface free energy (SFE) and increasing hydrophobicity. The latter mainly includes oxidative stress and photothermal/photodynamic effects. Graphene and its derivatives (GDs) are mainly combined with bioactive ceramic materials, metal materials and macromolecular polymers to play an antimicrobial effect in bone tissue engineering. Concentration, number of layers, and type of GDs often affect the antimicrobial activity of GMs. In this paper, we reviewed relevant osteoinductive and antimicrobial mechanisms of GMs and their applications in bone tissue engineering.
Collapse
Affiliation(s)
- Mengsong Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Review on material parameters to enhance bone cell function in vitro and in vivo. Biochem Soc Trans 2021; 48:2039-2050. [PMID: 32940685 DOI: 10.1042/bst20200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
Bone plays critical roles in support, protection, movement, and metabolism. Although bone has an innate capacity for regeneration, this capacity is limited, and many bone injuries and diseases require intervention. Biomaterials are a critical component of many treatments to restore bone function and include non-resorbable implants to augment bone and resorbable materials to guide regeneration. Biomaterials can vary considerably in their biocompatibility and bioactivity, which are functions of specific material parameters. The success of biomaterials in bone augmentation and regeneration is based on their effects on the function of bone cells. Such functions include adhesion, migration, inflammation, proliferation, communication, differentiation, resorption, and vascularization. This review will focus on how different material parameters can enhance bone cell function both in vitro and in vivo.
Collapse
|
42
|
Kwon N, Kim H, Li X, Yoon J. Supramolecular agents for combination of photodynamic therapy and other treatments. Chem Sci 2021; 12:7248-7268. [PMID: 34163818 PMCID: PMC8171357 DOI: 10.1039/d1sc01125a] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/04/2021] [Indexed: 12/17/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising treatment for cancers such as superficial skin cancers, esophageal cancer, and cervical cancer. Unfortunately, PDT often does not have sufficient therapeutic benefits due to its intrinsic oxygen dependence and the limited permeability of irradiating light. Side effects from "always on" photosensitizers (PSs) can be problematic, and PDT cannot treat tumor metastases or recurrences. In recent years, supramolecular approaches using non-covalent interactions have attracted attention due to their potential in PS development. A supramolecular PS assembly could be built to maximize photodynamic effects and minimize side effects. A combination of two or more therapies can effectively address shortcomings while maximizing the benefits of each treatment regimen. Using the supramolecular assembly, it is possible to design a multifunctional supramolecular PS to exert synergistic effects by combining PDT with other treatment methods. This review provides a summary of important research progress on supramolecular systems that can be used to combine PDT with photothermal therapy, chemotherapy, and immunotherapy to compensate for the shortcomings of PDT, and it provides an overview of the prospects for future cancer treatment advances and clinical applications.
Collapse
Affiliation(s)
- Nahyun Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| | - Heejeong Kim
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| | - Xingshu Li
- College of Chemistry, State Key Laboratory of Photocatalysis on Energy and Environment, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University Fuzhou 350116 China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| |
Collapse
|
43
|
Bagherifar R, Kiaie SH, Hatami Z, Ahmadi A, Sadeghnejad A, Baradaran B, Jafari R, Javadzadeh Y. Nanoparticle-mediated synergistic chemoimmunotherapy for tailoring cancer therapy: recent advances and perspectives. J Nanobiotechnology 2021; 19:110. [PMID: 33865432 PMCID: PMC8052859 DOI: 10.1186/s12951-021-00861-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Nowadays, a potent challenge in cancer treatment is considered the lack of efficacious strategy, which has not been able to significantly reduce mortality. Chemoimmunotherapy (CIT) as a promising approach in both for the first-line and relapsed therapy demonstrated particular benefit from two key gating strategies, including chemotherapy and immunotherapy to cancer therapy; therefore, the discernment of their participation and role of potential synergies in CIT approach is determinant. In this study, in addition to balancing the pros and cons of CIT with the challenges of each of two main strategies, the recent advances in the cancer CIT have been discussed. Additionally, immunotherapeutic strategies and the immunomodulation effect induced by chemotherapy, which boosts CIT have been brought up. Finally, harnessing and development of the nanoparticles, which mediated CIT have expatiated in detail.
Collapse
Affiliation(s)
- Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Kiaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Armin Ahmadi
- Department of Chemical & Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd., P.O. BoX: 1138, 57147, Urmia, Iran.
- Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Yousef Javadzadeh
- Biotechnology Research Center, and Faculty of Pharmacy, Tabriz University of Medical Science, 5166-15731, Tabriz, Iran.
| |
Collapse
|
44
|
Llewellyn SH, Faroni A, Iliut M, Bartlam C, Vijayaraghavan A, Reid AJ. Graphene Oxide Substrate Promotes Neurotrophic Factor Secretion and Survival of Human Schwann-Like Adipose Mesenchymal Stromal Cells. Adv Biol (Weinh) 2021; 5:e2000271. [PMID: 33852181 DOI: 10.1002/adbi.202000271] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/04/2021] [Indexed: 11/11/2022]
Abstract
Mesenchymal stromal cells from adipose tissue (AD-MSCs) exhibit favorable clinical traits for autologous transplantation and can develop 'Schwann-like' phenotypes (sAD-MSCs) to improve peripheral nerve regeneration, where severe injuries yield insufficient recovery. However, sAD-MSCs regress without biochemical stimulation and detach from conduits under unfavorable transplant conditions, negating their paracrine effects. Graphene-derived materials support AD-MSC attachment, regulating cell adhesion and function through physiochemistry and topography. Graphene oxide (GO) is a suitable substrate for human sAD-MSCs incubation toward severe peripheral nerve injuries by evaluating transcriptome changes, neurotrophic factor expression over a 7-days period, and cell viability in apoptotic conditions is reported. Transcriptome changes from GO incubation across four patients are minor compared to biological variance. Nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial-derived neurotrophic factor (GDNF) gene expression is unchanged from sAD-MSCs on GO substrates, but NGF and GDNF protein secretion increase at day 3 and 7. Secretome changes do not improve dorsal root ganglia neuron axon regeneration in conditioned media culture models. Fewer sAD-MSCs detach from GO substrates compared to glass following phosphate buffer saline exposure, which simulates apoptotic conditions. Overall, GO substrates are compatible with sAD-MSC primed for peripheral nerve regeneration strategies and protect the cell population in harsh environments.
Collapse
Affiliation(s)
- Steffan H Llewellyn
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK.,Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK
| | - Maria Iliut
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Cian Bartlam
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK.,Institute of Physics, EIT 2, Bundeswehr University Munich, Neubiberg, 85577, Germany
| | - Aravind Vijayaraghavan
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK.,Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M23 9LT, UK
| |
Collapse
|
45
|
Go EJ, Yang D, Ryu W, Chon HJ, Kim C, Park KS, Kim DH, Han DK, Park W. Optimal Voltage and Electrical Pulse Conditions for Electrical Ablation to Induce Immunogenic Cell Death (ICD). J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2020.10.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
46
|
Daneshmandi L, Barajaa M, Tahmasbi Rad A, Sydlik SA, Laurencin CT. Graphene-Based Biomaterials for Bone Regenerative Engineering: A Comprehensive Review of the Field and Considerations Regarding Biocompatibility and Biodegradation. Adv Healthc Mater 2021; 10:e2001414. [PMID: 33103370 PMCID: PMC8218309 DOI: 10.1002/adhm.202001414] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Graphene and its derivatives have continued to garner worldwide interest due to their unique characteristics. Having expanded into biomedical applications, there have been efforts to employ their exceptional properties for the regeneration of different tissues, particularly bone. This article presents a comprehensive review on the usage of graphene-based materials for bone regenerative engineering. The graphene family of materials (GFMs) are used either alone or in combination with other biomaterials in the form of fillers in composites, coatings for both scaffolds and implants, or vehicles for the delivery of various signaling and therapeutic agents. The applications of the GFMs in each of these diverse areas are discussed and emphasis is placed on the characteristics of the GFMs that have implications in this regard. In tandem and of importance, this article evaluates the safety and biocompatibility of the GFMs and carefully elucidates how various factors influence the biocompatibility and biodegradability of this new class of nanomaterials. In conclusion, the challenges and opportunities regarding the use of the GFMs in regenerative engineering applications are discussed, and future perspectives for the developments in this field are proposed.
Collapse
Affiliation(s)
- Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Mohammed Barajaa
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Armin Tahmasbi Rad
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Stefanie A Sydlik
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| |
Collapse
|
47
|
Salapa J, Bushman A, Lowe K, Irudayaraj J. Nano drug delivery systems in upper gastrointestinal cancer therapy. NANO CONVERGENCE 2020; 7:38. [PMID: 33301056 PMCID: PMC7728832 DOI: 10.1186/s40580-020-00247-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/23/2020] [Indexed: 05/02/2023]
Abstract
Upper gastrointestinal (GI) carcinomas are characterized as one of the deadliest cancer types with the highest recurrence rates. Their treatment is challenging due to late diagnosis, early metastasis formation, resistance to systemic therapy and complicated surgeries performed in poorly accessible locations. Current cancer medication face deficiencies such as high toxicity and systemic side-effects due to the non-specific distribution of the drug agent. Nanomedicine has the potential to offer sophisticated therapeutic possibilities through adjusted delivery systems. This review aims to provide an overview of novel approaches and perspectives on nanoparticle (NP) drug delivery systems for gastrointestinal carcinomas. Present regimen for the treatment of upper GI carcinomas are described prior to detailing various NP drug delivery formulations and their current and potential role in GI cancer theranostics with a specific emphasis on targeted nanodelivery systems. To date, only a handful of NP systems have met the standard of care requirements for GI carcinoma patients. However, an increasing number of studies provide evidence supporting NP-based diagnostic and therapeutic tools. Future development and strategic use of NP-based drug formulations will be a hallmark in the treatment of various cancers. This article seeks to highlight the exciting potential of novel NPs for targeted cancer therapy in GI carcinomas and thus provide motivation for further research in this field.
Collapse
Affiliation(s)
- Julia Salapa
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Department of Physics, Technical University of Vienna, Karlsplatz 13, 1040 Vienna, Austria
| | - Allison Bushman
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Kevin Lowe
- Carle Foundation Hospital South, Urbana, IL 61801 USA
- Carle-Illinois College of Medicine, Urbana, IL 61801 USA
| | - Joseph Irudayaraj
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
- Carle-Illinois College of Medicine, Urbana, IL 61801 USA
- Cancer Center at Illinois, Urbana, IL 61801 USA
- Biomedical Research Facility, 3rd Floor Mills Breast Cancer Institute, Carle Foundation Hospital South, Urbana, IL 61801 USA
| |
Collapse
|
48
|
Eckhart KE, Schmidt SJ, Starvaggi FA, Wolf ME, Vickery WM, Sydlik SA. Peptide- and Protein-Graphene Oxide Conjugate Materials for Controlling Mesenchymal Stem Cell Fate. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00182-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Sorroza-Martínez K, González-Méndez I, Martínez-Serrano RD, Solano JD, Ruiu A, Illescas J, Zhu XX, Rivera E. Efficient modification of PAMAM G1 dendrimer surface with β-cyclodextrin units by CuAAC: impact on the water solubility and cytotoxicity. RSC Adv 2020; 10:25557-25566. [PMID: 35518581 PMCID: PMC9055266 DOI: 10.1039/d0ra02574g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
The toxicity of the poly(amidoamine) dendrimers (PAMAM) caused by the peripheral amino groups has been a limitation for their use as drug carriers in clinical applications. In this work, we completely modified the periphery of PAMAM dendrimer generation 1 (PAMAM G1) with β-cyclodextrin (β-CD) units through the Cu(i)-catalyzed azide–alkyne cycloaddition (CuAAC) to obtain the PAMAM G1-β-CD dendrimer with high yield. The PAMAM G1-β-CD was characterized by 1H- and 13C-NMR and mass spectrometry studies. Moreover, the PAMAM G1-β-CD dendrimer showed remarkably higher water solubility than native β-CD. Finally, we studied the toxicity of PAMAM G1-β-CD dendrimer in four different cell lines, human breast cancer cells (MCF-7 and MDA-MB-231), human cervical adenocarcinoma cancer cells (HeLa) and pig kidney epithelial cells (LLC-PK1). The PAMAM G1-β-CD dendrimer did not present any cytotoxicity in cell lines tested which shows the potentiality of this new class of dendrimers. The toxicity of the poly(amidoamine) dendrimers (PAMAM) caused by the peripheral amino groups has been a limitation for their use as drug carriers in clinical applications.![]()
Collapse
Affiliation(s)
- Kendra Sorroza-Martínez
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| | - Israel González-Méndez
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| | - Ricardo D Martínez-Serrano
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| | - José D Solano
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| | - Andrea Ruiu
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| | - Javier Illescas
- Tecnológico Nacional de México/Instituto Tecnológico de Toluca Av. Tecnológico S/N, Col. Agrícola Bellavista CP 52149 Metepec México
| | - Xiao Xia Zhu
- Département de Chimie, Université de Montréal C.P. 6128, Succursale Centre-ville Montreal QC H3C 3J7 Canada
| | - Ernesto Rivera
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México Circuito Exterior, Ciudad Universitaria CP 04510 México City México
| |
Collapse
|
50
|
Silanization improves biocompatibility of graphene oxide. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110647. [DOI: 10.1016/j.msec.2020.110647] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/19/2019] [Accepted: 01/03/2020] [Indexed: 11/21/2022]
|