1
|
Liu J, Xi Z, Fan C, Mei Y, Zhao J, Jiang Y, Zhao M, Xu L. Hydrogels for Nucleic Acid Drugs Delivery. Adv Healthc Mater 2024; 13:e2401895. [PMID: 39152918 DOI: 10.1002/adhm.202401895] [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: 05/22/2024] [Revised: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Nucleic acid drugs are one of the hot spots in the field of biomedicine in recent years, and play a crucial role in the treatment of many diseases. However, its low stability and difficulty in target drug delivery are the bottlenecks restricting its application. Hydrogels are proven to be promising for improving the stability of nucleic acid drugs, reducing the adverse effects of rapid degradation, sudden release, and unnecessary diffusion of nucleic acid drugs. In this review, the strategies of loading nucleic acid drugs in hydrogels are summarized for various biomedical research, and classify the mechanism principles of these strategies, including electrostatic binding, hydrogen bond based binding, hydrophobic binding, covalent bond based binding and indirect binding using various carriers. In addition, this review also describes the release strategies of nucleic acid drugs, including photostimulation-based release, enzyme-responsive release, pH-responsive release, and temperature-responsive release. Finally, the applications and future research directions of hydrogels for delivering nucleic acid drugs in the field of medicine are discussed.
Collapse
Affiliation(s)
- Jiaping Liu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ziyue Xi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Chuanyong Fan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yihua Mei
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Jiale Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yingying Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ming Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| |
Collapse
|
2
|
Xiao L, Yang Y, Yu J, Li Y, Chen S, Gu Y, Tang C, Yang H, Wang Z, Geng D. Urolithin B inhibits the differentiation of M1 macrophages and relieves the inflammation around the implants under osteoporosis via down-regulating the phosphorylation of VEGFR2. Int Immunopharmacol 2024; 140:112854. [PMID: 39116494 DOI: 10.1016/j.intimp.2024.112854] [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: 05/10/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
The inflammation causes the destroyed osseointegration at the implant-bone interface, significantly increasing the probability of implant loosening in osteoporotic patients. Currently, inhibiting the differentiation of M1 macrophages and the inflammatory response could be a solution to stabilize the microenvironment of implants. Interestingly, some natural products have anti-inflammatory and anti-polarization effects, which could be a promising candidate for stabilizing the implants' microenvironment in osteoporotic patients. This research aims to explore the inhibitory effect of Urolithin B(UB) on macrophage M1 polarization, which ameliorates inflammation, thus alleviating implant instability. We established an osteoporosis mouse model of implant loosening. The mouse tissues were taken out for morphological analysis, staining analysis, and bone metabolic index analysis. In in vitro experiments, RAW264.7 cells were polarized to M1 macrophages using lipopolysaccharide (LPS) and analyzed by immunofluorescence (IF) staining, Western blot (WB), and flow cytometry. The CSP100 plus chip experiments were used to explore the potential mechanisms behind the inhibiting effects of UB. Through observation of these experiments, UB can improve the osseointegration between the implants and femurs in osteoporotic mice and enhance the stability of implants. The UB can inhibit the differentiation of M1 macrophages and local inflammation via inhibiting the phosphorylation of VEGFR2, which can be further proved by the weakened inhibited effects of UB in macrophages with lentivirus-induced overexpression of VEGFR2. Overall, UB can specifically inhibit the activation of VEGFR2, alleviate local inflammation, and improve the stability of implants in osteoporotic mice.
Collapse
Affiliation(s)
- Long Xiao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China; Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Yunshang Yang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Jingxian Yu
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Yajun Li
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Shuangshuang Chen
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Yong Gu
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Cheng Tang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
3
|
Liu HC, Huang CH, Chiang MR, Hsu RS, Chou TC, Lu TT, Lee IC, Liao LD, Chiou SH, Lin ZH, Hu SH. Sustained Release of Nitric Oxide-Mediated Angiogenesis and Nerve Repair by Mussel-Inspired Adaptable Microreservoirs for Brain Traumatic Injury Therapy. Adv Healthc Mater 2024; 13:e2302315. [PMID: 37713592 DOI: 10.1002/adhm.202302315] [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: 07/20/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Traumatic brain injury (TBI) triggers inflammatory response and glial scarring, thus substantially hindering brain tissue repair. This process is exacerbated by the accumulation of activated immunocytes at the injury site, which contributes to scar formation and impedes tissue repair. In this study, a mussel-inspired nitric oxide-release microreservoir (MINOR) that combines the features of reactive oxygen species (ROS) scavengers and sustained NO release to promote angiogenesis and neurogenesis is developed for TBI therapy. The injectable MINOR fabricated using a microfluidic device exhibits excellent monodispersity and gel-like self-healing properties, thus allowing the maintenance of its structural integrity and functionality upon injection. Furthermore, polydopamine in the MINOR enhances cell adhesion, significantly reduces ROS levels, and suppresses inflammation. Moreover, a nitric oxide (NO) donor embedded into the MINOR enables the sustained release of NO, thus facilitating angiogenesis and mitigating inflammatory responses. By harnessing these synergistic effects, the biocompatible MINOR demonstrates remarkable efficacy in enhancing recovery in mice. These findings benefit future therapeutic interventions for patients with TBI.
Collapse
Affiliation(s)
- Hsiu-Ching Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Chu-Han Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Min-Ren Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Ru-Siou Hsu
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Tsu-Chin Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, 300044, Hsinchu, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, 320314, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35053, Miaoli County, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, National Yang Ming Chiao Tung University, Taipei Veterans General Hospital, 112304, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Zhong-Hong Lin
- Department of Biomedical Engineering, National Taiwan University, 10617, Taipei, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| |
Collapse
|
4
|
Pantović Pavlović MR, Ignjatović NL, Gudić S, Vrsalović L, Božić KĐ, Popović ME, Pavlović MM. Modified Titanium Surface with Nano Amorphous Calcium Phosphate@Chitosan Oligolactate as Ion Loading Platform with Multifunctional Properties for Potential Biomedical Application. Ann Biomed Eng 2024; 52:2221-2233. [PMID: 38662122 DOI: 10.1007/s10439-024-03521-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Titanium (Ti) is widely used in medical and dental implants. Calcium phosphate (CPs) coatings enhance Ti implants' osteoinductive properties, and additives further improve these coatings. Recently, a nano amorphous calcium phosphate (nACP) coating decorated with chitosan oligolactate (ChOL) and selenium (Se) showed immunomodulatory effects. This study investigates the surface morphology, composition, bioactivity, mechanical properties, and Se-release mechanism of the nACP@ChOL-Se hybrid coating on Ti substrates. Amorphous calcium phosphate (ACP) was synthesized, and the nACP@ChOL-Se hybrid coating was deposited on Ti substrates using in situ anaphoretic deposition. Physico-chemical characterization was used to analyze the surface of the coating (scanning electron microscopy (SEM), X-ray diffraction (XRD), and Fourier Transform Infrared Spectroscopy). The distribution of Se within the coating was examined with energy-dispersive X-ray spectroscopy (EDS). Bioactivity was evaluated in simulated body fluid (SBF), and adhesion was tested using a scratch test method. In vitro testing determined the release mechanism of Se. SEM images illustrated the surface morphology, while AFM provided a detailed analysis of surface roughness. XRD analysis revealed structural and phase composition, and EDS confirmed Se distribution within the coating. The coating exhibited bioactivity in SBF and showed good adhesion according to the scratch test. In vitro testing uncovered the release mechanism of Se from the coating. This study successfully characterized the surface morphology, composition, bioactivity, and Se-release mechanism of the nACP@ChOL-Se hybrid coating on Ti substrates, offering insights for developing immunomodulatory coatings for medical and dental applications.
Collapse
Affiliation(s)
- Marijana R Pantović Pavlović
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, 11000, Serbia
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, Belgrade, 11000, Serbia
| | - Nenad L Ignjatović
- Institute of Technical Sciences of the Serbian Academy of Sciences and Arts, Belgrade, 11000, Serbia
| | - Senka Gudić
- Faculty of Chemistry and Technology, University of Split, 21000, Split, Croatia
| | - Ladislav Vrsalović
- Faculty of Chemistry and Technology, University of Split, 21000, Split, Croatia
| | - Katarina Đ Božić
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, 11000, Serbia
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, Belgrade, 11000, Serbia
| | - Marko E Popović
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, 11000, Serbia
| | - Miroslav M Pavlović
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, 11000, Serbia.
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, Belgrade, 11000, Serbia.
| |
Collapse
|
5
|
Li Z, Jin L, Yang X, Liu H, Qian S, Wang Z, Liu J, Wang J, Chen J, Su B, Peng C, Wang J, Shi Z. A multifunctional ionic liquid coating on 3D-Printed prostheses: Combating infection, promoting osseointegration. Mater Today Bio 2024; 26:101076. [PMID: 38711938 PMCID: PMC11070339 DOI: 10.1016/j.mtbio.2024.101076] [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: 01/18/2024] [Revised: 03/27/2024] [Accepted: 04/27/2024] [Indexed: 05/08/2024] Open
Abstract
Periprosthetic infection and mechanical loosening are two leading causes of implant failure in orthopedic surgery that have devastating consequences for patients both physically and financially. Hence, advanced prostheses to simultaneously prevent periprosthetic infection and promote osseointegration are highly desired to achieve long-term success in orthopedics. In this study, we proposed a multifunctional three-dimensional printed porous titanium alloy prosthesis coated with imidazolium ionic liquid. The imidazolium ionic liquid coating exhibited excellent bacterial recruitment property and near-infrared (NIR) triggered photothermal bactericidal activity, enabling the prosthesis to effectively trap bacteria in its vicinity and kill them remotely via tissue-penetrating NIR irradiation. In vivo anti-infection and osseointegration investigations in infected animal models confirmed that our antibacterial prosthesis could provide long-term and sustainable prevention against periprosthetic infection, while promoting osseointegration simultaneously. It is expected to accelerate the development of next-generation prostheses and improve patient outcomes after prosthesis implantation.
Collapse
Affiliation(s)
- Zuhao Li
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Lunqiang Jin
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xijing Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Shengxu Qian
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Zhonghan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jiaqi Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jingxia Wang
- Radiation Chemistry Department, Sichuan Institute of Atomic Energy, Chengdu 610101, China
| | - Junjun Chen
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Baihai Su
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Chaorong Peng
- Radiation Chemistry Department, Sichuan Institute of Atomic Energy, Chengdu 610101, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhenqiang Shi
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
6
|
Boys AJ. There and Back Again: Building Systems That Integrate, Interface, and Interact with the Human Body. Adv Biol (Weinh) 2024; 8:e2300366. [PMID: 38400703 DOI: 10.1002/adbi.202300366] [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: 07/23/2023] [Revised: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Since Dr. Theodor Schwann posed the extension of Cell Theory to mammals in 1839, scientists have dreamt up ways to interface with and influence the cells. Recently, considerable ground in this area is gained, particularly in the scope of bioelectronics. New advances in this area have provided with a means to record electrical activity from cells, examining neural firing or epithelial barrier integrity, and stimulate cells through applied electrical fields. Many of these applications utilize invasive implantation systems to perform this interaction in close proximity to the cells in question. Traditionally, the body's immune system fights back against these systems through the foreign body response, limiting the efficacy of long-term interactions. New technologies in tissue engineering, biomaterials science, and bioelectronics offer the potential to circumvent the foreign body response and create stable long-term biological interfaces. Looking ahead, the next advancements in the biomedical sciences can truly integrate, interface, and interact with the human body.
Collapse
Affiliation(s)
- Alexander J Boys
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| |
Collapse
|
7
|
Tuftee C, Alsberg E, Ozbolat IT, Rizwan M. Emerging granular hydrogel bioinks to improve biological function in bioprinted constructs. Trends Biotechnol 2024; 42:339-352. [PMID: 37852853 PMCID: PMC10939978 DOI: 10.1016/j.tibtech.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
Advancements in 3D bioprinting have been hindered by the trade-off between printability and biological functionality. Existing bioinks struggle to meet both requirements simultaneously. However, new types of bioinks composed of densely packed microgels promise to address this challenge. These bioinks possess intrinsic porosity, allowing for cell growth, oxygen and nutrient transport, and better immunomodulatory properties, leading to superior biological functions. In this review, we highlight key trends in the development of these granular bioinks. Using examples, we demonstrate how granular bioinks overcome the trade-off between printability and cell function. Granular bioinks show promise in 3D bioprinting, yet understanding their unique structure-property-function relationships is crucial to fully leverage the transformative capabilities of these new types of bioinks in bioprinting.
Collapse
Affiliation(s)
- Cody Tuftee
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Eben Alsberg
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Mechanical & Industrial Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, Chicago, IL 60612, USA
| | - Ibrahim Tarik Ozbolat
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Engineering Science and Mechanics, Penn State University, University Park, PA 16802, USA; Neurosurgery Department, Penn State University; Hershey, PA 17033, USA; Medical Oncology Department, Cukurova University, Adana 01330, Turkey
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA.
| |
Collapse
|
8
|
You C, Cao J, Du Y, Peng C, Cheng L, Ren J, Zhang W, Zheng H, Guo K, Gao X, Zhang F, Wang J, Li H, Liu T. ε-Poly-l-lysine-hydroxyphenyl propionic acid/IL-4 composite hydrogels with inflammation regulation and antibacterial activity for improving integration stability of soft tissues and orthopedic implants. Int J Biol Macromol 2024; 254:127937. [PMID: 37939753 DOI: 10.1016/j.ijbiomac.2023.127937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
The failure of orthopedic implants is usually caused by inflammation, poor tissue integration, and infection, which can lead to pain, limited mobility, dysfunction of patients. This may require additional surgical interventions, such as removal, replacement, or repair of implants, as well as related treatment measures such as antibiotic therapy, physical therapy. Here, an injectable hydrogel carrier was developed for the steady release of inflammatory regulators to reduce the surface tissue inflammatory response of orthopedic implants and induce soft tissue regeneration, ultimately achieving the promotion of implants stability. The hydrogels carrier was prepared by hydroxyphenyl propionic acid-modified ε-Poly-l-lysine (EPA), hydrogen peroxide and horseradish peroxidase, which showed antibacterial bioactive and stable factor release ability. Due to the introduction of IL-4, EPA@IL-4 hydrogels showed good inflammatory regulation. EPA@IL-4 hydrogels regulated the differentiation of macrophages into M2 in inflammatory environment in vitro, and promoted endothelial cells to show a more obvious trend of tube formation. The composite hydrogels reduced the inflammation on the surface of the implants in vivo, induced local endothelial cell angiogenesis, and had more collagen deposition and new granulation tissue. Therefore, EPA hydrogels based on IL-4 release are promising candidates for promoting of implants surface anti-inflammatory, soft tissue regeneration, and anti-infection.
Collapse
Affiliation(s)
- Chaoqun You
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China; Department of Orthopedics, Clinical Medical College, Weifang Medical University, No. 7166 Baotong West Street, Weifang, Shandong 261000, China
| | - Jiashi Cao
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China; Department of Orthopedics, No. 455 Hospital of the Chinese People's Liberation Army, The Navy Medical University, No. 338 Huaihai West Road, Shanghai 200052, China
| | - Yan Du
- College of Physical Science and Technology, Sichuan University, Chengdu 610065, China
| | - Cheng Peng
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Linfei Cheng
- School of Medicine, Anhui University of Science and Technology, No.168 Taifeng Road, Huainan 232001, China
| | - Jiaji Ren
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China; Department of Orthopedics, Clinical Medical College, Weifang Medical University, No. 7166 Baotong West Street, Weifang, Shandong 261000, China
| | - Wanli Zhang
- College of Physical Science and Technology, Sichuan University, Chengdu 610065, China
| | - Heng Zheng
- College of Physical Science and Technology, Sichuan University, Chengdu 610065, China
| | - Kai Guo
- Department of Orthopedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No. 164 Lanxi Road, Shanghai 200062, China
| | - Xin Gao
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Fei Zhang
- Ningbo Beilun Changzheng Orthopaedic Hospital, 458 Fengyang 1st Road, Beilun District, Ningbo, Zhejiang 315800, China
| | - Jing Wang
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China.
| | - Hong Li
- College of Physical Science and Technology, Sichuan University, Chengdu 610065, China.
| | - Tielong Liu
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China; Department of Orthopedics, Clinical Medical College, Weifang Medical University, No. 7166 Baotong West Street, Weifang, Shandong 261000, China.
| |
Collapse
|
9
|
Frisch E, Clavier L, Belhamdi A, Vrana NE, Lavalle P, Frisch B, Heurtault B, Gribova V. Preclinical in vitro evaluation of implantable materials: conventional approaches, new models and future directions. Front Bioeng Biotechnol 2023; 11:1193204. [PMID: 37576997 PMCID: PMC10416115 DOI: 10.3389/fbioe.2023.1193204] [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: 03/24/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Nowadays, implants and prostheses are widely used to repair damaged tissues or to treat different diseases, but their use is associated with the risk of infection, inflammation and finally rejection. To address these issues, new antimicrobial and anti-inflammatory materials are being developed. Aforementioned materials require their thorough preclinical testing before clinical applications can be envisaged. Although many researchers are currently working on new in vitro tissues for drug screening and tissue replacement, in vitro models for evaluation of new biomaterials are just emerging and are extremely rare. In this context, there is an increased need for advanced in vitro models, which would best recapitulate the in vivo environment, limiting animal experimentation and adapted to the multitude of these materials. Here, we overview currently available preclinical methods and models for biological in vitro evaluation of new biomaterials. We describe several biological tests used in biocompatibility assessment, which is a primordial step in new material's development, and discuss existing challenges in this field. In the second part, the emphasis is made on the development of new 3D models and approaches for preclinical evaluation of biomaterials. The third part focuses on the main parameters to consider to achieve the optimal conditions for evaluating biocompatibility; we also overview differences in regulations across different geographical regions and regulatory systems. Finally, we discuss future directions for the development of innovative biomaterial-related assays: in silico models, dynamic testing models, complex multicellular and multiple organ systems, as well as patient-specific personalized testing approaches.
Collapse
Affiliation(s)
- Emilie Frisch
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Lisa Clavier
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | | | | | - Philippe Lavalle
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- SPARTHA Medical, Strasbourg, France
| | - Benoît Frisch
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Béatrice Heurtault
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Varvara Gribova
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| |
Collapse
|
10
|
Zhou L, Xing Y, Ou Y, Ding J, Han Y, Lin D, Chen J. Prolonged release of an antimicrobial peptide GL13K-loaded thermosensitive hydrogel on a titanium surface improves its antibacterial and anti-inflammatory properties. RSC Adv 2023; 13:23308-23319. [PMID: 37538512 PMCID: PMC10395452 DOI: 10.1039/d3ra03414c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/17/2023] [Indexed: 08/05/2023] Open
Abstract
The application of titanium in the orthopedic and dental fields is associated with bacterial infection and chronic inflammation, especially in the early stages after its implantation. In the present study, we investigated the antibacterial and anti-inflammatory activities of a titanium surface that was immobilized in a thermosensitive PLGA-PEG-PLGA hydrogel containing the antimicrobial peptide GL13K. The FTIR results confirmed the successful loading of GL13K. The degradation of the hydrogel and release of GL13K persisted for two weeks. The modified titanium surface exhibited a significant inhibitory effect on Porphyromonas gingivalis in contact with its surface, as well as an inhibitory effect on P.g in the surrounding environment by releasing GL13K antimicrobial peptides. The modified titanium surfaces were biocompatible with RAW264.7. Furthermore, the expression of pro-inflammatory cytokines IL-1β, TNF-α and iNOS was down-regulated, whereas anti-inflammatory cytokines Arg-1, IL-10 and VEGF-A were up-regulated on the modified titanium surfaces on days 3 and 5. This effect was attributed to the polarization of macrophages from the M1 to M2 phenotype, which was confirmed by the detection of macrophage M1/M2 biomarkers via immunofluorescence staining and flow cytometry. Thus, the thermosensitive PLGA-PEG-PLGA hydrogel release system carrying the antimicrobial peptide GL13K on a titanium surface exhibited antibacterial and anti-inflammatory properties and promoted macrophage polarization from the M1 to M2 phenotype, which may help create a favourable niche for bone formation under infective condition.
Collapse
Affiliation(s)
- Lin Zhou
- Affiliated Stomatological Hospital of Fujian Medical University, Fujian Medical University Fujian China
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University Fuzhou China
| | - Yifeng Xing
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University Fuzhou China
- Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University Fuzhou China
| | - Yanjin Ou
- Affiliated Stomatological Hospital of Fujian Medical University, Fujian Medical University Fujian China
- Fujian Biological Materials Engineering and Technology Center of Stomatology, Fujian Medical University Fuzhou China
| | - Jiamin Ding
- Department of Oral Mucosa, Affiliated Stomatological Hospital of Fujian Medical University Fuzhou China
| | - Yu Han
- Division of Craniofacial Development and Tissue Biology, Graduate School of Dentistry, Tohoku University Sendai City Japan
| | - Dong Lin
- Affiliated Stomatological Hospital of Fujian Medical University, Fujian Medical University Fujian China
| | - Jiang Chen
- Affiliated Stomatological Hospital of Fujian Medical University, Fujian Medical University Fujian China
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University Fuzhou China
| |
Collapse
|
11
|
Pantović Pavlović MR, Ignjatović NL, Panić VV, Mirkov II, Kulaš JB, Malešević AL, Pavlović MM. Immunomodulatory Effects Mediated by Nano Amorphous Calcium Phosphate/Chitosan Oligosaccharide Lactate Coatings Decorated with Selenium on Titanium Implants. J Funct Biomater 2023; 14:jfb14040227. [PMID: 37103318 PMCID: PMC10143504 DOI: 10.3390/jfb14040227] [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: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 04/28/2023] Open
Abstract
The aim of this work is in situ anodization/anaphoretic deposition of a nano amorphous calcium phosphate (ACP)/chitosan oligosaccharide lactate (ChOL) multifunctional hybrid coating decorated with selenium (Se) on a titanium substrate and in vivo investigation of its immunomodulatory and anti-inflammatory effect. Investigating phenomena at the implant-tissue interface of interest for controlled inflammation and immunomodulation was also the aim of the research. In our earlier research, we designed coatings based on ACP and ChOL on titanium with anticorrosive, antibacterial and biocompatible properties, while in the presented results we show that selenium addition makes this coating an immunomodulator. The immunomodulatory effect of the novel hybrid coating is characterized by the examination of the functional aspects in the tissue around the implant (in vivo): proinflammatory cytokines' gene expression, M1 (iNOS) and M2 (Arg1) macrophages, fibrous capsule formation (TGF-β) and vascularization (VEGF). The EDS, FTIR and XRD analyses prove the formation of a ACP/ChOL/Se multifunctional hybrid coating on Ti and the presence of Se. A higher M2/M1 macrophage ratio in the ACP/ChOL/Se-coated implants compared to pure titanium implants (a higher level of Arg1 expression) is noted at all time points examined (after 7, 14 and 28 days). Lower inflammation measured by gene expression of proinflammatory cytokines IL-1β and TNF, lower expression of TGF-β in the surrounding tissue and higher IL-6 expression (solely at day 7 post-implantation) is noted in presence of the ACP/ChOL/Se-coated implants.
Collapse
Affiliation(s)
- Marijana R Pantović Pavlović
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, 11000 Belgrade, Serbia
| | - Nenad L Ignjatović
- Institute of Technical Science of the Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Vladimir V Panić
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, 11000 Belgrade, Serbia
- Department of Chemical-Technological Sciences, State University of Novi Pazar, 36300 Novi Pazar, Serbia
| | - Ivana I Mirkov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic"-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena B Kulaš
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic"-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Anastasija Lj Malešević
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic"-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Miroslav M Pavlović
- Department of Electrochemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Center of Excellence in Chemistry and Environmental Engineering-ICTM, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
12
|
Pu Y, Lin X, Zhi Q, Qiao S, Yu C. Microporous Implants Modified by Bifunctional Hydrogel with Antibacterial and Osteogenic Properties Promote Bone Integration in Infected Bone Defects. J Funct Biomater 2023; 14:jfb14040226. [PMID: 37103316 PMCID: PMC10143991 DOI: 10.3390/jfb14040226] [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: 03/13/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
Prosthesis implantation and bone integration under bacterial infection are arduous challenges in clinical practice. It is well known that the reactive oxygen species (ROS) produced by bacterial infection around the bone defects will further hinder bone healing. To solve this problem, we prepared a ROS-scavenging hydrogel by cross-linking polyvinyl alcohol and a ROS-responsive linker, N1-(4-boronobenzyl)-N3-(4-boronophenyl)-N1, N1, N3, N3-tetramethylpropane-1, 3-diaminium, to modify the microporous titanium alloy implant. The prepared hydrogel was used as an advanced ROS-scavenging tool to promote bone healing by inhibiting the ROS levels around the implant. Bifunctional hydrogel serving as a drug delivery system can release therapeutic molecules, including vancomycin, to kill bacteria and bone morphogenetic protein-2 to induce bone regeneration and integration. This multifunctional implant system that combines mechanical support and disease microenvironment targeting provides a novel strategy for bone regeneration and integration of implants in infected bone defects.
Collapse
Affiliation(s)
- Yiping Pu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200001, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200001, China
| | - Xuecai Lin
- Hongqiao Community Health Service Center, Minhang District, Shanghai 201103, China
| | - Qiang Zhi
- National Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, Shanghai 200011, China
| | - Shichong Qiao
- National Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, Shanghai 200011, China
| | - Chuangqi Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200001, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200001, China
| |
Collapse
|
13
|
Gribova V, Dominguez JMA, Morin A, Sepulveda Diaz J, Lavalle P, Vrana NE. A miniaturized genotoxicity evaluation system for fast biomaterial-related risk assessment. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:1584-1593. [PMID: 36883977 DOI: 10.1039/d2ay01873j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Implants and prostheses are widely used to either repair damaged tissues or treat different diseases. Before an implant reaches the market, multiple preclinical and clinical tests must be performed. Along with cytotoxicity or hemocompatibility preclinical tests, genotoxicity is an essential feature to investigate. Indeed, the materials used for implantation should be non-genotoxic, i.e. they should not promote mutations that can potentially lead to tumour formation. However, given the complexity level of genotoxicity tests, such tests are not readily available to biomaterials researchers, which is the reason why this aspect is severely underreported in the literature. To solve this problem, we developed a simplified genotoxicity test that can be further adapted by standard biomaterials laboratories. We started by simplifying the classic Ames test in Petri dishes, after which we developed a miniaturized test in a microfluidic chip, which takes only 24 hours, requiring significantly less material and space. An automatization option with a customized testing chamber architecture and microfluidics-based control system has been designed as well. This optimized microfluidic chip system can significantly improve the availability of genotoxicity tests for biomaterials developers, with the additional benefit of more in-depth observation and quantitative comparison due to the availability of processable image components.
Collapse
Affiliation(s)
- Varvara Gribova
- INSERM UMR 1121, Biomaterials and Bioengineering, 1 rue Eugène Boeckel, FR-67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 1 place de l'Hôpital, FR-67000 Strasbourg, France
| | | | - Alan Morin
- Elvesys, 172 Rue de Charonne, FR-75011 Paris, France
| | | | - Philippe Lavalle
- INSERM UMR 1121, Biomaterials and Bioengineering, 1 rue Eugène Boeckel, FR-67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 1 place de l'Hôpital, FR-67000 Strasbourg, France
- SPARTHA Medical, 1 Rue Eugène Boeckel, FR-67000 Strasbourg, France.
| | | |
Collapse
|
14
|
Rafikova G, Piatnitskaia S, Shapovalova E, Chugunov S, Kireev V, Ialiukhova D, Bilyalov A, Pavlov V, Kzhyshkowska J. Interaction of Ceramic Implant Materials with Immune System. Int J Mol Sci 2023; 24:4200. [PMID: 36835610 PMCID: PMC9959507 DOI: 10.3390/ijms24044200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
The immuno-compatibility of implant materials is a key issue for both initial and long-term implant integration. Ceramic implants have several advantages that make them highly promising for long-term medical solutions. These beneficial characteristics include such things as the material availability, possibility to manufacture various shapes and surface structures, osteo-inductivity and osteo-conductivity, low level of corrosion and general biocompatibility. The immuno-compatibility of an implant essentially depends on the interaction with local resident immune cells and, first of all, macrophages. However, in the case of ceramics, these interactions are insufficiently understood and require intensive experimental examinations. Our review summarizes the state of the art in variants of ceramic implants: mechanical properties, different chemical modifications of the basic material, surface structures and modifications, implant shapes and porosity. We collected the available information about the interaction of ceramics with the immune system and highlighted the studies that reported ceramic-specific local or systemic effects on the immune system. We disclosed the gaps in knowledge and outlined the perspectives for the identification to ceramic-specific interactions with the immune system using advanced quantitative technologies. We discussed the approaches for ceramic implant modification and pointed out the need for data integration using mathematic modelling of the multiple ceramic implant characteristics and their contribution for long-term implant bio- and immuno-compatibility.
Collapse
Affiliation(s)
- Guzel Rafikova
- Laboratory of Immunology, Institute of Urology and Clinical Oncology, Bashkir State Medical University, 450008 Ufa, Russia
| | - Svetlana Piatnitskaia
- Institute of Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia
| | - Elena Shapovalova
- Department of Chemistry, Tomsk State University, 634050 Tomsk, Russia
| | | | - Victor Kireev
- Institute of Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia
- Department of Applied Physics, Ufa University of Science and Technology, 450076 Ufa, Russia
| | - Daria Ialiukhova
- Institute of Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia
| | - Azat Bilyalov
- Institute of Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia
| | | | - Julia Kzhyshkowska
- Institute of Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia
- Department of Chemistry, Tomsk State University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciecnes (MI3), Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg, 68167 Mannheim, Germany
| |
Collapse
|
15
|
Šušteršič T, Gribova V, Nikolic M, Lavalle P, Filipovic N, Vrana NE. The Effect of Machine Learning Algorithms on the Prediction of Layer-by-Layer Coating Properties. ACS OMEGA 2023; 8:4677-4686. [PMID: 36777619 PMCID: PMC9909801 DOI: 10.1021/acsomega.2c06471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/30/2022] [Indexed: 06/18/2023]
Abstract
Layer-by-layer film (LbL) coatings made of polyelectrolytes are a powerful tool for surface modification, including the applications in the biomedical field, for food packaging, and in many electrochemical systems. However, despite the number of publications related to LbL assembly, predicting LbL coating properties represents quite a challenge, can take a long time, and be very costly. Machine learning (ML) methodologies that are now emerging can accelerate and improve new coating development and potentially revolutionize the field. Recently, we have demonstrated a preliminary ML-based model for coating thickness prediction. In this paper, we compared several ML algorithms for optimizing a methodology for coating thickness prediction, namely, linear regression, Support Vector Regressor, Random Forest Regressor, and Extra Tree Regressor. The current research has shown that learning algorithms are effective in predicting the coating output value, with the Extra Tree Regressor algorithm demonstrating superior predictive performance, when used in combination with optimized hyperparameters and with missing data imputation. The best predictors of the coating thickness were determined, and they can be later used to accurately predict coating thickness, avoiding measurement of multiple parameters. The development of optimized methodologies will ensure different reliable predictive models for coating property/function relations. As a continuation, the methodology can be adapted and used for predicting the outputs connected to antimicrobial, anti-inflammatory, and antiviral properties in order to be able to respond to actual biomedical problems such as antibiotic resistance, implant rejection, or COVID-19 outbreak.
Collapse
Affiliation(s)
- Tijana Šušteršič
- Faculty
of Engineering, University of Kragujevac (FINK), Kragujevac34000, Serbia
- Steinbeis
Advanced Risk Technologies Institute doo Kragujevac (SARTIK), Kragujevac34000, Serbia
- Bioengineering
Research and Development Center (BioIRC), Kragujevac34000, Serbia
| | - Varvara Gribova
- Biomaterials
and Bioengineering laboratory, INSERM UMR
1121, Strasbourg67100, France
- Université
de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg67000, France
| | - Milica Nikolic
- Steinbeis
Advanced Risk Technologies Institute doo Kragujevac (SARTIK), Kragujevac34000, Serbia
- Institute
of Information Technologies, University of Kragujevac, Kragujevac34000, Serbia
- Eindhoven
University of Technology, Eindhoven5611 CB, The Netherlands
| | - Philippe Lavalle
- Biomaterials
and Bioengineering laboratory, INSERM UMR
1121, Strasbourg67100, France
- Université
de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg67000, France
- SPARTHA
Medical, Strasbourg67100, France
| | - Nenad Filipovic
- Faculty
of Engineering, University of Kragujevac (FINK), Kragujevac34000, Serbia
- Steinbeis
Advanced Risk Technologies Institute doo Kragujevac (SARTIK), Kragujevac34000, Serbia
- Bioengineering
Research and Development Center (BioIRC), Kragujevac34000, Serbia
| | | |
Collapse
|
16
|
Chen C, Chen Y, Lan YJ, Tian MN, Zhang YM, Lei ZY, Fan DL. Effects of substrate topography on the regulation of human fibroblasts and capsule formation via modulating macrophage polarization. Colloids Surf B Biointerfaces 2023; 222:113086. [PMID: 36542953 DOI: 10.1016/j.colsurfb.2022.113086] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/25/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
The host-material interface is critical in determining the successful integration of medical devices into human tissue. The surface topography can regulate the fibrous capsule formation around implants through macrophage polarization, but the exact mechanism remains unclear. In this study, four types of microgrooves (10 or 50 µm in groove depths and 50 or 200 µm in groove widths) were fabricated on polydimethylsiloxane (PDMS) using lithography. The microgroove surfaces were characterized using the laser scanning confocal microscopy and fourier transform infrared spectroscopy. The effect of surface topography on macrophage phenotypes and conditioned medium (CM) collected from macrophages on human foreskin fibroblast 1 (HFF-1) were investigated. The result revealed that a deeper and narrower microgroove structure means a rougher surface. Macrophages tended to adhere and aggregate on group 50-50 surface (groove depths and widths of 50 µm). THP-1 cell polarized toward both inflammatory M1 and anti-inflammatory M2 macrophages on the surface of each group. Meanwhile, CM from macrophages culture on PDMS differentially up-regulated the proliferation, migration and fibrosis of HFF-1. Among them, the group 50-50 had the strongest promoting effect. In vivo, the inflammatory response and fibrotic capsule around the implants were observed at 1 week and 4 weeks. As time passed, the inflammatory response decreased, while the capsule thickness continued to increase. The rough material surface was more inclined to develop a severe fibrotic encapsulation. In conclusion, this finding further suggested a potential immunomodulatory effect of macrophages in mediating the fibrotic response to implants and facilitated the design of biomaterial interfaces for improving tissue integration.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Yao Chen
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Yu-Jie Lan
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Meng-Nan Tian
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Yi-Ming Zhang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Ze-Yuan Lei
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China
| | - Dong-Li Fan
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing 400037, China.
| |
Collapse
|
17
|
Avery D, Morandini L, Sheakley LS, Shah AH, Bui L, Abaricia JO, Olivares-Navarrete R. Canonical Wnt signaling enhances pro-inflammatory response to titanium by macrophages. Biomaterials 2022; 289:121797. [PMID: 36156410 PMCID: PMC10262842 DOI: 10.1016/j.biomaterials.2022.121797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
Biomaterial characteristics like surface roughness and wettability can determine the phenotype of macrophages following implantation. We have demonstrated that inhibiting Wnt ligand secretion abolishes macrophage polarization in vitro and in vivo; however, the role of canonical Wnt signaling in macrophage activation in response to physical and chemical biomaterial cues is unknown. The aim of this study was to understand whether canonical Wnt signaling affects the response of macrophages to titanium (Ti) surface roughness or wettability in vitro and in vivo. Activating canonical Wnt signaling increased expression of toll-like receptors and interleukin receptors and secreted pro-inflammatory cytokines and reduced anti-inflammatory cytokines on Ti, regardless of surface properties. Inhibiting canonical Wnt signaling reduced pro-inflammatory cytokines on all Ti surfaces and increased anti-inflammatory cytokines on rough or rough-hydrophilic Ti. In vivo, activating canonical Wnt signaling increased total macrophages, pro-inflammatory macrophages, and T cells and decreased anti-inflammatory macrophages on both smooth and rough-hydrophilic implants. Functionally, canonical Wnt activation increases pro-inflammatory macrophage response to cell and cell-extracellular matrix lysates. These results demonstrate that activating canonical Wnt signaling primes macrophages to a pro-inflammatory phenotype that affects their response to Ti implants in vitro and in vivo.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke S Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Loc Bui
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
18
|
Wan D, Liu Y, Guo X, Zhang J, Pan J. Intelligent Drug Delivery by Peptide-Based Dual-Function Micelles. Int J Mol Sci 2022; 23:ijms23179698. [PMID: 36077102 PMCID: PMC9456463 DOI: 10.3390/ijms23179698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
To endow the polymeric prodrug with smart properties through a safe and simple method, matrix metalloproteinase (MMPs) responsive peptide GPLGVRGDG was introduced into the block copolymer to prepare TPGS3350-GPLGVRGDG-DOX&DOX micelles, where TPGS3350 is D-α-tocopheryl polyethylene glycol 3350 succinate. During the doxorubicin delivery, the cleavage of the peptide chain triggers de-PEGylation, and the remaining VRGDG sequence was retained on the surface of the micelles, which can act as a ligand to facilitate cell uptake. Moreover, the cytotoxicity of TPGS3350-GPLGVRGDG-DOX&DOX micelles against 4T1 cells was significantly improved, compared with TPGS3350-GPLGVRG-DOX&DOX micelles and TPGS3350-DOX&DOX micelles. During in vivo studies, TPGS3350-GPLGVRGDG-DOX&DOX micelles exhibited good anticancer efficacy with long circulation in the body and more efficient accumulation at the tumor site. Therefore, TPGS3350-GPLGVRGDG-DOX&DOX micelles have improved antitumor activity and reduced toxic side effects. This work opens new potential for exploring the strategy of drug delivery in clinical applications.
Collapse
Affiliation(s)
- Dong Wan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Yujun Liu
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Xinhao Guo
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jianxin Zhang
- School of Chemistry, Tiangong University, Tianjin 300387, China
- Correspondence: (J.Z.); (J.P.)
| | - Jie Pan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
- Correspondence: (J.Z.); (J.P.)
| |
Collapse
|
19
|
Xing D, Zuo W, Chen J, Ma B, Cheng X, Zhou X, Qian Y. Spatial Delivery of Triple Functional Nanoparticles via an Extracellular Matrix-Mimicking Coaxial Scaffold Synergistically Enhancing Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37380-37395. [PMID: 35946874 DOI: 10.1021/acsami.2c08784] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
It remains a major challenge to simultaneously achieve bone regeneration and prevent infection in the complex microenvironment of repairing bone defects. Here, we developed a novel ECM-mimicking scaffold by coaxial electrospinning to be endowed with multibiological functions. Lysophosphatidic acid (LPA) and zinc oxide (ZnO) nanoparticles were loaded into the poly-lactic-co-glycolic acid/polycaprolactone (PLGA/PCL, PP) sheath layer of coaxial nanofibers, and deferoxamine (DFO) nanoparticles were loaded into its core layer. The novel scaffold PP-LPA-ZnO/DFO maintained a porous nanofibrous architecture after incorporating three active nanoparticles, showing better physicochemical properties and eximious biocompatibility. In vitro studies showed that the bio-scaffold loaded with LPA nanoparticles had excellent cell adhesion, proliferation, and differentiation for MC3T3-E1 cells and synergistic osteogenesis with the addition of ZnO and DFO nanoparticles. Further, the PP-LPA-ZnO/DFO scaffold promoted tube formation and facilitated the expression of vascular endothelial markers in HUVECs. In vitro antibacterial studies against Escherichia Coli and Staphylococcus aureus demonstrated effective antibacterial activity of the PP-LPA-ZnO/DFO scaffold. In vivo studies showed that the PP-LPA-ZnO/DFO scaffold exhibited excellent biocompatibility after subcutaneous implantation and remarkable osteogenesis at 4 weeks post-implantation in the mouse alveolar bone defects. Importantly, the PP-LPA-ZnO/DFO scaffold showed significant antibacterial activity, prominent neovascularization, and new bone formation in the rat fenestration defect model. Overall, the spatially sustained release of LPA, ZnO, and DFO nanoparticles through the coaxial scaffold synergistically enhanced biocompatibility, osteogenesis, angiogenesis, and effective antibacterial properties, which is ultimately beneficial for bone regeneration. This project provides the optimized design of bone regenerative biomaterials and a new strategy for bone regeneration, especially in the potentially infected microenvironment.
Collapse
Affiliation(s)
- Danlei Xing
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
- Department of Plastic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| | - Wei Zuo
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| | - Jiahong Chen
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| | - Buyun Ma
- Nano Science and Technology Institute, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Xi Cheng
- Department of Plastic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| | - Xuefeng Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Yunzhu Qian
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| |
Collapse
|
20
|
Nikovics K, Durand M, Castellarin C, Burger J, Sicherre E, Collombet JM, Oger M, Holy X, Favier AL. Macrophages Characterization in an Injured Bone Tissue. Biomedicines 2022; 10:biomedicines10061385. [PMID: 35740407 PMCID: PMC9219779 DOI: 10.3390/biomedicines10061385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
Biomaterial use is a promising approach to facilitate wound healing of the bone tissue. Biomaterials induce the formation of membrane capsules and the recruitment of different types of macrophages. Macrophages are immune cells that produce diverse combinations of cytokines playing an important role in bone healing and regeneration, but the exact mechanism remains to be studied. Our work aimed to identify in vivo macrophages in the Masquelet induced membrane in a rat model. Most of the macrophages in the damaged area were M2-like, with smaller numbers of M1-like macrophages. In addition, high expression of IL-1β and IL-6 cytokines were detected in the membrane region by RT-qPCR. Using an innovative combination of two hybridization techniques (in situ hybridization and in situ hybridization chain reaction (in situ HCR)), M2b-like macrophages were identified for the first time in cryosections of non-decalcified bone. Our work has also demonstrated that microspectroscopical analysis is essential for macrophage characterization, as it allows the discrimination of fluorescence and autofluorescence. Finally, this work has revealed the limitations of immunolabelling and the potential of in situ HCR to provide valuable information for in vivo characterization of macrophages.
Collapse
Affiliation(s)
- Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (C.C.); (E.S.); (M.O.); (A.-L.F.)
- Correspondence: or ; Tel.: +33-(0)-1-78-65-13-331
| | - Marjorie Durand
- Osteo-Articulary Biotherapy Unit, Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (M.D.); (J.-M.C.)
| | - Cédric Castellarin
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (C.C.); (E.S.); (M.O.); (A.-L.F.)
| | - Julien Burger
- Microbiology and Infectious Diseases Department, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | - Emma Sicherre
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (C.C.); (E.S.); (M.O.); (A.-L.F.)
| | - Jean-Marc Collombet
- Osteo-Articulary Biotherapy Unit, Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (M.D.); (J.-M.C.)
| | - Myriam Oger
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (C.C.); (E.S.); (M.O.); (A.-L.F.)
| | - Xavier Holy
- Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | - Anne-Laure Favier
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France; (C.C.); (E.S.); (M.O.); (A.-L.F.)
| |
Collapse
|
21
|
A 3D in vitro co-culture model for evaluating biomaterial-mediated modulation of foreign-body responses. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
22
|
Gribova V, Petit L, Seguin C, Fournel S, Kichler A, Vrana NE, Lavalle P. Polyarginine as a simultaneous antimicrobial, immunomodulatory and miRNA delivery agent within polyanionic hydrogels. Macromol Biosci 2022; 22:e2200043. [PMID: 35332672 DOI: 10.1002/mabi.202200043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/07/2022]
Abstract
Implantation of biomedical devices is followed by immune response to the implant, as well as occasionally bacterial, yeast and/or fungal infections. In this context, new implant materials and coatings that deal with medical device-associated complications are required. Antibacterial and anti-inflammatory materials are also required for wound healing applications, especially in diabetic patients with chronic wounds. In this work, we present hyaluronic acid (HA) hydrogels with triple activity: antimicrobial, immunomodulatory and miRNA delivery agent. We demonstrate that polyarginine with a degree of polymerization of 30 (PAR30), which was previously shown to have a prolonged antibacterial activity, decreases inflammatory response of LPS-stimulated macrophages. In addition, PAR30 accelerated fibroblast migration in macrophage/fibroblast co-culture system, suggesting a positive effect on wound healing. Furthermore, PAR30 allowed to load miRNA into HA hydrogels, and then to deliver them into the cells. To our knowledge, this study is the first describing miRNA-loaded hydrogels with antibacterial effect and anti-inflammatory features. Such system can become a tool for the treatment of infected wounds, e.g. diabetic ulcers, as well as for foreign body response modulation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Varvara Gribova
- Institut National de la Santé et de la Recherche Médicale, INSERM U1121 Biomaterials and Bioengineering, 1 rue Eugène Boeckel, Strasbourg, 67000, France.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, Strasbourg, 67000, France
| | - Lauriane Petit
- Institut National de la Santé et de la Recherche Médicale, INSERM U1121 Biomaterials and Bioengineering, 1 rue Eugène Boeckel, Strasbourg, 67000, France.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, Strasbourg, 67000, France
| | - Cendrine Seguin
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, Illkirch Cedex, 67401, France
| | - Sylvie Fournel
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, Illkirch Cedex, 67401, France
| | - Antoine Kichler
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, Illkirch Cedex, 67401, France
| | - Nihal Engin Vrana
- SPARTHA Medical, 14B rue de la Canardière, Strasbourg, 67100, France
| | - Philippe Lavalle
- Institut National de la Santé et de la Recherche Médicale, INSERM U1121 Biomaterials and Bioengineering, 1 rue Eugène Boeckel, Strasbourg, 67000, France.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, Strasbourg, 67000, France.,SPARTHA Medical, 14B rue de la Canardière, Strasbourg, 67100, France
| |
Collapse
|
23
|
In Situ Gene Expression in Native Cryofixed Bone Tissue. Biomedicines 2022; 10:biomedicines10020484. [PMID: 35203694 PMCID: PMC8962289 DOI: 10.3390/biomedicines10020484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 01/21/2023] Open
Abstract
Bone is a very complex tissue that is constantly changing throughout the lifespan. The precise mechanism of bone regeneration remains poorly understood. Large bone defects can be caused by gunshot injury, trauma, accidents, congenital anomalies and tissue resection due to cancer. Therefore, understanding bone homeostasis and regeneration has considerable clinical and scientific importance in the development of bone therapy. Macrophages are well known innate immune cells secreting different combinations of cytokines and their role in bone regeneration during bone healing is essential. Here, we present a method to identify mRNA transcripts in cryosections of non-decalcified rat bone using in situ hybridization and hybridization chain reaction to explore gene expression in situ for better understanding the gene expression of the bone tissues.
Collapse
|
24
|
Li D, Shi S, Zhao D, Rong Y, Zhou Y, Ding J, He C, Chen X. Effect of Polymer Topology and Residue Chirality on Biodegradability of Polypeptide Hydrogels. ACS Biomater Sci Eng 2022; 8:626-637. [PMID: 35090109 DOI: 10.1021/acsbiomaterials.1c01127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Polypeptide-based injectable hydrogels have attracted the attention of biomedical researchers due to their unique biocompatibility and biodegradability, tunable residue chirality, and secondary conformation of polypeptide chains. In the present study, four types of poly(ethylene glycol)-block-poly(glutamic acid)s with different topological structures and residue chirality of polypeptide segments were developed, which were grafted with tyramine side groups for further cross-linking. The results demonstrated that the covalent conjugation between the tyramine groups in the presence of horseradish peroxidase and hydrogen peroxide could form porous hydrogels rapidly. Additionally, the gelation time and mechanical strength of the hydrogels were measured. All the polymer precursors and hydrogels exhibited good cytocompatibility in vitro. Further assessment of the enzymatic degradability of the hydrogels and copolymers in vitro revealed that the degradation rate was influenced by the adjustment of polymer topology or residue chirality of polypeptide copolymers. Subsequently, the effect of copolymer topology and polypeptide chirality on in vivo biodegradability and biocompatibility was assessed. This study will provide insights into the relationship between copolymer structures and hydrogel properties and benefit future polypeptide-based hydrogel studies in biomedical applications.
Collapse
Affiliation(s)
- Dong Li
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| | - Shun Shi
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 Sichuan, P. R. China
| | - Dan Zhao
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| | - Yan Rong
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China
| | - Yuhao Zhou
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| | - Junfeng Ding
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 Jilin, P. R. China.,University of Science and Technology of China, Hefei, 230026 Anhui, P. R. China
| |
Collapse
|
25
|
Agarwal T, Tan SA, Onesto V, Law JX, Agrawal G, Pal S, Lim WL, Sharifi E, Moghaddam FD, Maiti TK. Engineered herbal scaffolds for tissue repair and regeneration: Recent trends and technologies. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
26
|
Perera K, Ivone R, Natekin E, Wilga CA, Shen J, Menon JU. 3D Bioprinted Implants for Cartilage Repair in Intervertebral Discs and Knee Menisci. Front Bioeng Biotechnol 2021; 9:754113. [PMID: 34746106 PMCID: PMC8570130 DOI: 10.3389/fbioe.2021.754113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023] Open
Abstract
Cartilage defects pose a significant clinical challenge as they can lead to joint pain, swelling and stiffness, which reduces mobility and function thereby significantly affecting the quality of life of patients. More than 250,000 cartilage repair surgeries are performed in the United States every year. The current gold standard is the treatment of focal cartilage defects and bone damage with nonflexible metal or plastic prosthetics. However, these prosthetics are often made from hard and stiff materials that limits mobility and flexibility, and results in leaching of metal particles into the body, degeneration of adjacent soft bone tissues and possible failure of the implant with time. As a result, the patients may require revision surgeries to replace the worn implants or adjacent vertebrae. More recently, autograft - and allograft-based repair strategies have been studied, however these too are limited by donor site morbidity and the limited availability of tissues for surgery. There has been increasing interest in the past two decades in the area of cartilage tissue engineering where methods like 3D bioprinting may be implemented to generate functional constructs using a combination of cells, growth factors (GF) and biocompatible materials. 3D bioprinting allows for the modulation of mechanical properties of the developed constructs to maintain the required flexibility following implantation while also providing the stiffness needed to support body weight. In this review, we will provide a comprehensive overview of current advances in 3D bioprinting for cartilage tissue engineering for knee menisci and intervertebral disc repair. We will also discuss promising medical-grade materials and techniques that can be used for printing, and the future outlook of this emerging field.
Collapse
Affiliation(s)
- Kalindu Perera
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Ryan Ivone
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Evelina Natekin
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK, United States
| | - Cheryl. A. Wilga
- Department of Biological Sciences, University of Rhode Island, Kingston, RI, United States
- Department of Electrical, Computer and Biomedical Engineering, University of Rhode Island, Kingston, RI, United States
| | - Jie Shen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI, United States
| | - Jyothi U. Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI, United States
| |
Collapse
|