1
|
Pho T, Janecka MA, Pustulka SM, Champion JA. Nanoetched Stainless Steel Architecture Enhances Cell Uptake of Biomacromolecules and Alters Protein Corona Abundancy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58427-58438. [PMID: 39417567 PMCID: PMC11533172 DOI: 10.1021/acsami.4c14492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024]
Abstract
Nanotexture on biocompatible surfaces promotes cell adhesion and proliferation. High aspect ratio nanoachitecture serves as an ideal interface between implant materials and host cells that is well-suited for localized therapeutic delivery. Despite this potential, nanotextured surfaces have not been widely applied for biomacromolecule delivery. Here, we employed a low-cost, industrially relevant nanoetching process to modify the surface of biocompatible stainless steel 316 (SS316L), creating nanotextured SS316L (NT-SS316L) as a material for intracellular biomacromolecule delivery. As biomacromolecule cargoes are adsorbed to the steel and ultimately would be used in protein-rich environments, we performed serum protein corona analysis on unmodified SS316L and NT-SS316L using tandem mass spectrometry. We observed an increase in proteins associated with cell adhesion on the surface of NT-SS316L compared to that of SS316L, supporting literature reports of enhanced adhesion on nanotextured materials. For delivery to adherent cells, a "hard corona" of model biomacromolecule cargoes including superfolder green fluorescent protein (sfGFP) charge variants, cytochrome c, and siRNA was adsorbed on NT-SS316L to assess delivery. Nanotextured surfaces enhanced cellular biomacromolecule uptake and delivered cytosolic-functional proteins and nucleic acids through energy-dependent endocytosis. Collectively, these findings indicate that NT-SS316L holds potential as a surface modification for implants to achieve localized drug delivery for a variety of biomedical applications.
Collapse
Affiliation(s)
- Thomas Pho
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
- BioEngineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Maeve A. Janecka
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Samantha M. Pustulka
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
- BioEngineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
2
|
Bright R, Visalakshan RM, Simon J, Rokstad AM, Ghazaryan A, Morsbach S, Hayles A, Mailänder V, Landfester K, Vasilev K. Manipulation of Serum Protein Adsorption by Nanoengineered Biomaterials Influences Subsequent Immune Responses. ACS Biomater Sci Eng 2024; 10:6230-6240. [PMID: 39213601 DOI: 10.1021/acsbiomaterials.4c01103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The adsorption of serum proteins on biomaterial surfaces is a critical determinant for the outcome of medical procedures and therapies, which involve inserting materials and devices into the body. In this study, we aimed to understand how surface topography at the nanoscale influences the composition of the protein corona that forms on the (bio)material surface when placed in contact with serum proteins. To achieve that, we developed nanoengineered model surfaces with finely tuned topography of 16, 40, and 70 nm, overcoated with methyl oxazoline to ensure uniform outermost chemistry across all surfaces. Our findings revealed that within the studied height range, surface nanotopography had no major influence on the overall quantity of adsorbed proteins. However, significant alterations were observed in the composition of the adsorbed protein corona. For instance, clusterin adsorption decreased on all the nanotopography-modified surfaces. Conversely, there was a notable increase in the adsorption of ApoB and IgG gamma on the 70 nm nanotopography. In comparison, the adsorption of albumin was greater on surfaces that had a topography scale of 40 nm. Analysis of the gene enrichment data revealed a reduction in protein adsorption across all immune response-related biological pathways on nanotopography-modified surfaces. This reduction became more pronounced for larger surface nanoprotrusions. Macrophages were used as representative immune cells to assess the influence of the protein corona composition on inflammatory outcomes. Gene expression analysis demonstrated reduced inflammatory responses on the nanotopographically modified surface, a trend further corroborated by cytokine analysis. These findings underscore the potential of precisely engineered nanotopography-coated surfaces for augmenting biomaterial functionality.
Collapse
Affiliation(s)
- Richard Bright
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Rahul M Visalakshan
- Academic Unit of STEM, University of South Australia, Mawson Lakes, Adelaide, South Australia 5095, Australia
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Anne Mari Rokstad
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Arthur Ghazaryan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Andrew Hayles
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Dermatology Department, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| |
Collapse
|
3
|
Amani H, Alipour M, Shahriari E, Taboas JM. Immunomodulatory Biomaterials: Tailoring Surface Properties to Mitigate Foreign Body Reaction and Enhance Tissue Regeneration. Adv Healthc Mater 2024:e2401253. [PMID: 39370571 DOI: 10.1002/adhm.202401253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/28/2024] [Indexed: 10/08/2024]
Abstract
The immune cells have demonstrated the ability to promote tissue repair by removing debris, breaking down the extracellular matrix, and regulating cytokine secretion profile. If the behavior of immune cells is not well directed, chronic inflammation and foreign body reaction (FBR) will lead to scar formation and loss of biomaterial functionality. The immunologic response toward tissue repair or chronic inflammation after injury and implantation can be modulated by manipulating the surface properties of biomaterials. Tailoring surface properties of biomaterials enables the regulation of immune cell fate such as adhesion, proliferation, recruitment, polarization, and cytokine secretion profile. This review begins with an overview of the role of immune cells in tissue healing and their interactions with biomaterials. It then discusses how the surface properties of biomaterials influence immune cell behavior. The core focus is reviewing surface modification methods to create innovative materials that reduce foreign body reactions and enhance tissue repair and regeneration by modulating immune cell activities. The review concludes with insights into future advancements in surface modification techniques and the associated challenges.
Collapse
Affiliation(s)
- Hamed Amani
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mahdieh Alipour
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Elahe Shahriari
- Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Juan M Taboas
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| |
Collapse
|
4
|
Hou Y, Conklin B, Choi HK, Yang L, Lee KB. Probing Nanotopography-Mediated Macrophage Polarization via Integrated Machine Learning and Combinatorial Biophysical Cue Mapping. ACS NANO 2024; 18:25465-25477. [PMID: 39226301 DOI: 10.1021/acsnano.4c04406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Inflammatory responses, leading to fibrosis and potential host rejection, significantly hinder the long-term success and widespread adoption of biomedical implants. The ability to control and investigated macrophage inflammatory responses at the implant-macrophage interface would be critical for reducing chronic inflammation and improving tissue integration. Nonetheless, the systematic investigation of how surface topography affects macrophage polarization is typically complicated by the restricted complexity of accessible nanostructures, difficulties in achieving exact control, and biased preselection of experimental parameters. In response to these problems, we developed a large-scale, high-content combinatorial biophysical cue (CBC) array for enabling high-throughput screening (HTS) of the effects of nanotopography on macrophage polarization and subsequent inflammatory processes. Our CBC array, created utilizing the dynamic laser interference lithography (DLIL) technology, contains over 1 million nanotopographies, ranging from nanolines and nanogrids to intricate hierarchical structures with dimensions ranging from 100 nm to several microns. Using machine learning (ML) based on the Gaussian process regression algorithm, we successfully identified certain topographical signals that either repress (pro-M2) or stimulate (pro-M1) macrophage polarization. The upscaling of these nanotopographies for further examination has shown mechanisms such as cytoskeletal remodeling and ROCK-dependent epigenetic activation to be critical to the mechanotransduction pathways regulating macrophage fate. Thus, we have also developed a platform combining advanced DLIL nanofabrication techniques, HTS, ML-driven prediction of nanobio interactions, and mechanotransduction pathway evaluation. In short, our developed platform technology not only improves our ability to investigate and understand nanotopography-regulated macrophage inflammatory responses but also holds great potential for guiding the design of nanostructured coatings for therapeutic biomaterials and biomedical implants.
Collapse
Affiliation(s)
- Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
5
|
He Y, Cen Y, Tian M. Immunomodulatory hydrogels for skin wound healing: cellular targets and design strategy. J Mater Chem B 2024; 12:2435-2458. [PMID: 38284157 DOI: 10.1039/d3tb02626d] [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: 01/30/2024]
Abstract
Skin wounds significantly impact the global health care system and represent a significant burden on the economy and society due to their complicated dynamic healing processes, wherein a series of immune events are required to coordinate normal and sequential healing phases, involving multiple immunoregulatory cells such as neutrophils, macrophages, keratinocytes, and fibroblasts, since dysfunction of these cells may impede skin wound healing presenting persisting inflammation, impaired vascularization, and excessive collagen deposition. Therefore, cellular target-based immunomodulation is promising to promote wound healing as cells are the smallest unit of life in immune response. Recently, immunomodulatory hydrogels have become an attractive avenue to promote skin wound healing. However, a detailed and comprehensive review of cellular targets and related hydrogel design strategies remains lacking. In this review, the roles of the main immunoregulatory cells participating in skin wound healing are first discussed, and then we highlight the cellular targets and state-of-the-art design strategies for immunomodulatory hydrogels based on immunoregulatory cells that cover defect, infected, diabetic, burn and tumor wounds and related scar healing. Finally, we discuss the barriers that need to be addressed and future prospects to boost the development and prosperity of immunomodulatory hydrogels.
Collapse
Affiliation(s)
- Yinhai He
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Baheti W, Chen X, La M, He H. Biomimetic HA-GO implant coating for enhanced osseointegration via macrophage M2 polarization-induced osteo-immunomodulation. J Appl Biomater Funct Mater 2024; 22:22808000241266665. [PMID: 39129373 DOI: 10.1177/22808000241266665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Abstract
The pro-inflammatory/anti-inflammatory polarized phenotypes of macrophages (M1/M2) can be used to predict the success of implant integration. Hence, activating and inducing the transformation of immunocytes that promote tissue repair appears to be a highly promising strategy for facilitating osteo-anagenesis. In a previous study, titanium implants were coated with a graphene oxide-hydroxyapatite (GO-HA) nanocomposite via electrophoretic deposition, and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) was found to be significantly enhanced when the GO content was 2wt%. However, the effectiveness of the GO-HA nanocomposite coating in modifying the in vivo immune microenvironment still remains unclear. In this study, the effects of GO-HA coatings on osteogenesis were investigated based on the GO-HA-mediated immune regulation of macrophages. The HA-2wt%GO nanocomposite coatings exhibited good biocompatibility and favored M2 macrophage polarization. Meanwhile, they could also significantly upregulate IL-10 (anti-inflammatory factor) expression and downregulate TNF-α (pro-inflammatory factor) expression. Additionally, the microenvironment, which was established by M2 macrophages, favored the osteogenesis of BMSCs both in vivo and in vitro. These findings show that the GO-HA nanocomposite coating is a promising surface-modification material. Hence, this study provides a reference for the development of next-generation osteoimmunomodulatory biomaterials.
Collapse
Affiliation(s)
- Wufanbieke Baheti
- Department of Stomatology, People's Hospital of Xinjiang Autonomous Region, Urumqi, China
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaotao Chen
- Department of Stomatology, People's Hospital of Xinjiang Autonomous Region, Urumqi, China
| | - Mi La
- Department of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huiyu He
- Department of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
7
|
Mikhail AS, Morhard R, Mauda-Havakuk M, Kassin M, Arrichiello A, Wood BJ. Hydrogel drug delivery systems for minimally invasive local immunotherapy of cancer. Adv Drug Deliv Rev 2023; 202:115083. [PMID: 37673217 DOI: 10.1016/j.addr.2023.115083] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/27/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
Although systemic immunotherapy has achieved durable responses and improved survival for certain patients and cancer types, low response rates and immune system-related systemic toxicities limit its overall impact. Intratumoral (intralesional) delivery of immunotherapy is a promising technique to combat mechanisms of tumor immune suppression within the tumor microenvironment and reduce systemic drug exposure and associated side effects. However, intratumoral injections are prone to variable tumor drug distribution and leakage into surrounding tissues, which can compromise efficacy and contribute to toxicity. Controlled release drug delivery systems such as in situ-forming hydrogels are promising vehicles for addressing these challenges by providing improved spatio-temporal control of locally administered immunotherapies with the goal of promoting systemic tumor-specific immune responses and abscopal effects. In this review we will discuss concepts, applications, and challenges in local delivery of immunotherapy using controlled release drug delivery systems with a focus on intratumorally injected hydrogel-based drug carriers.
Collapse
Affiliation(s)
- Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Robert Morhard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michal Mauda-Havakuk
- Interventional Oncology service, Interventional Radiology, Tel Aviv Sourasky Medical Center, Tel Aviv District, Israel
| | - Michael Kassin
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Tripathi AS, Zaki MEA, Al-Hussain SA, Dubey BK, Singh P, Rind L, Yadav RK. Material matters: exploring the interplay between natural biomaterials and host immune system. Front Immunol 2023; 14:1269960. [PMID: 37936689 PMCID: PMC10627157 DOI: 10.3389/fimmu.2023.1269960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023] Open
Abstract
Biomaterials are widely used for various medical purposes, for instance, implants, tissue engineering, medical devices, and drug delivery systems. Natural biomaterials can be obtained from proteins, carbohydrates, and cell-specific sources. However, when these biomaterials are introduced into the body, they trigger an immune response which may lead to rejection and failure of the implanted device or tissue. The immune system recognizes natural biomaterials as foreign substances and triggers the activation of several immune cells, for instance, macrophages, dendritic cells, and T cells. These cells release pro-inflammatory cytokines and chemokines, which recruit other immune cells to the implantation site. The activation of the immune system can lead to an inflammatory response, which can be beneficial or detrimental, depending on the type of natural biomaterial and the extent of the immune response. These biomaterials can also influence the immune response by modulating the behavior of immune cells. For example, biomaterials with specific surface properties, such as charge and hydrophobicity, can affect the activation and differentiation of immune cells. Additionally, biomaterials can be engineered to release immunomodulatory factors, such as anti-inflammatory cytokines, to promote a tolerogenic immune response. In conclusion, the interaction between biomaterials and the body's immune system is an intricate procedure with potential consequences for the effectiveness of therapeutics and medical devices. A better understanding of this interplay can help to design biomaterials that promote favorable immune responses and minimize adverse reactions.
Collapse
Affiliation(s)
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Sami A Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Bidhyut Kumar Dubey
- Department of Pharmaceutical Chemistry, Era College of Pharmacy, Era University, Lucknow, India
| | - Prabhjot Singh
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Laiba Rind
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Rajnish Kumar Yadav
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| |
Collapse
|
9
|
Zhang L, Zhang L, Wang Y, Jiang K, Gao C, Zhang P, Xie Y, Wang B, Zhao Y, Xiao H, Song J. Regulating the surface topography of CpG nanoadjuvants via coordination-driven self-assembly for enhanced tumor immunotherapy. NANOSCALE ADVANCES 2023; 5:4758-4769. [PMID: 37705793 PMCID: PMC10496906 DOI: 10.1039/d3na00322a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/09/2023] [Indexed: 09/15/2023]
Abstract
Immunoadjuvants play a key role in enhancing the efficacy of therapeutic tumor vaccines for treating malignant and recurrent cancers. However, due to the bottleneck in the rational design and mechanistic understanding of novel adjuvants, currently available immunoadjuvants in clinical practice are very limited. To boost adjuvant design and development, herein we propose a surface topography regulatory strategy for constructing novel adjuvants with improved adjuvant properties. One of the licensed adjuvants with a well-defined molecular mechanism of immune activation, cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODNs), was used as the material framework. We constructed immunostimulatory CpG nanoparticles (CpG NPs) with different surface topographies by coordination-driven self-assembly between CpG ODNs and ferrous ions. These self-assembled CpG NPs combine the biological and physical activation abilities of innate immunity and can be used as adjuvants of tumor antigens for malignant tumor immunotherapy. The experimental results showed that these CpG NPs could rapidly enter innate immune cells and remold the tumor microenvironment (TME) to enhance anti-tumor immunotherapy via (i) inducing proinflammatory cytokine production; (ii) promoting the transformation of macrophages from immunosuppressed M2 types into immunoactivated M1 types; (iii) amplifying the antigen presentation of mature dendritic cells (DCs), and (iv) activating T cells in tumor sites. Among the prepared nanostructures, pompon-shaped nanoparticles (NPpo) showed the strongest adjuvant properties and anti-tumor immunotherapeutic effect as the adjuvant of ovalbumin in melanoma-bearing mice. Overall, this work provides an effective strategy for designing novel adjuvants for activating the immunosuppressed TME to enable better cancer immunotherapy.
Collapse
Affiliation(s)
- Li Zhang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou Zhejiang 310024 China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences Hangzhou Zhejiang 310022 China
- School of Pharmacy, Changzhou University Changzhou Jiangsu 213164 China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Yuqi Wang
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University Shanghai 200240 China
| | - Kai Jiang
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University Shanghai 200240 China
| | - Chao Gao
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University Shanghai 200240 China
| | - Pengfei Zhang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences Hangzhou Zhejiang 310022 China
| | - Yujie Xie
- School of Chemistry, University of Birmingham Edgbaston Birmingham B15 2TT UK
| | - Bin Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Yun Zhao
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou Zhejiang 310024 China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Jie Song
- Hangzhou Institute of Medicine, Chinese Academy of Sciences Hangzhou Zhejiang 310022 China
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University Shanghai 200240 China
| |
Collapse
|
10
|
Fu Y, Jing Z, Chen T, Xu X, Wang X, Ren M, Wu Y, Wu T, Li Y, Zhang H, Ji P, Yang S. Nanotube patterning reduces macrophage inflammatory response via nuclear mechanotransduction. J Nanobiotechnology 2023; 21:229. [PMID: 37468894 DOI: 10.1186/s12951-023-01912-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/26/2023] [Indexed: 07/21/2023] Open
Abstract
The inflammatory immune environment surrounding titanium bone implants determines the formation of osseointegration, and nanopatterning on implant surfaces modulates the immune microenvironment in the implant region. Among many related mechanisms, the mechanism by which nanopatterning controls macrophage inflammatory response still needs to be elucidated. In this paper, we found that inhibition of the nuclear envelope protein lamin A/C by titania nanotubes (TNTs) reduced the macrophage inflammatory response. Knockdown of lamin A/C reduced macrophage inflammatory marker expression, while overexpression of lamin A/C significantly elevated inflammatory marker expression. We further found that suppression of lamin A/C by TNTs limited actin polymerization, thereby reducing the nuclear translocation of the actin-dependent transcriptional cofactor MRTF-A, which subsequently reduced the inflammatory response. In addition, emerin, which is a key link between lamin A/C and actin, was delocalized from the nucleus in response to mechanical stimulation by TNTs, resulting in reduced actin organization. Under inflammatory conditions, TNTs exerted favourable osteoimmunomodulatory effects on the osteogenic differentiation of mouse bone marrow-derived stem cells (mBMSCs) in vitro and osseointegration in vivo. This study shows and confirms for the first time that lamin A/C-mediated nuclear mechanotransduction controls macrophage inflammatory response, and this study provides a theoretical basis for the future design of immunomodulatory nanomorphologies on the surface of metallic bone implants.
Collapse
Affiliation(s)
- Yiru Fu
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Zheng Jing
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tao Chen
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Xinxin Xu
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Xu Wang
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Mingxing Ren
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Yanqiu Wu
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Tianli Wu
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
| | - Yuzhou Li
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - He Zhang
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ping Ji
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Sheng Yang
- College of Stomatology, Chongqing Medical University, 426# Songshi-bei Road, Yubei District, Chongqing, 401147, China.
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
11
|
Li L, Liu K, Chen J, Wen W, Li H, Li L, Ding S, Liu M, Zhou C, Luo B. Bone ECM-inspired biomineralization chitin whisker liquid crystal hydrogels for bone regeneration. Int J Biol Macromol 2023; 231:123335. [PMID: 36690237 DOI: 10.1016/j.ijbiomac.2023.123335] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/04/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
As a particular cell niche, natural bone extracellular matrix (ECM) is an organic-inorganic composite material formed by mineralization of liquid crystal (LC) collagen fiber network. However, designing bone repair materials that highly imitate the LC characteristic and composite components of natural bone ECM is a great challenge. Here, we report a novel kind of bone ECM-inspired biomineralization chitin whisker LC hydrogels. First, photocurable chitin whisker LC hydrogels with bone ECM-like chiral nematic LC state and viscoelasticity are created. Next, biomineralization, guided by LC hydrogels, is carried out to truly mimic the mineralization process of natural bone, so as to obtain the organic-inorganic composite materials with bone ECM-like microenvironment. The chitin whisker LC hydrogels exhibit superior biomineralization, protein adsorption and osteogenesis ability, more importantly, LC hydrogel with negatively charged -COOH groups is more conducive to biomineralization and shows more desirable osteogenic activity than that with positively charged -NH2 groups. Notably, compared with the pristine LC hydrogels, the biomineralization LC hydrogels display more favorable osteogenesis ability due to their bone ECM-like LC texture and bone-like hydroxyapatite. This study opens an avenue toward the design of bone ECM-inspired biomineralization chitin whisker LC hydrogels for bone regeneration.
Collapse
Affiliation(s)
- Lin Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Jingsheng Chen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Hong Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Lihua Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Shan Ding
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China.
| |
Collapse
|
12
|
Luo Z, Sun L, Bian F, Wang Y, Yu Y, Gu Z, Zhao Y. Erythrocyte-Inspired Functional Materials for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206150. [PMID: 36581585 PMCID: PMC9951328 DOI: 10.1002/advs.202206150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/03/2022] [Indexed: 05/30/2023]
Abstract
Erythrocytes are the most abundant cells in the blood. As the results of long-term natural selection, their specific biconcave discoid morphology and cellular composition are responsible for gaining excellent biological performance. Inspired by the intrinsic features of erythrocytes, various artificial biomaterials emerge and find broad prospects in biomedical applications such as therapeutic delivery, bioimaging, and tissue engineering. Here, a comprehensive review from the fabrication to the applications of erythrocyte-inspired functional materials is given. After summarizing the biomaterials mimicking the biological functions of erythrocytes, the synthesis strategies of particles with erythrocyte-inspired morphologies are presented. The emphasis is on practical biomedical applications of these bioinspired functional materials. The perspectives for the future possibilities of the advanced erythrocyte-inspired biomaterials are also discussed. It is hoped that the summary of existing studies can inspire researchers to develop novel biomaterials; thus, accelerating the progress of these biomaterials toward clinical biomedical applications.
Collapse
Affiliation(s)
- Zhiqiang Luo
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Lingyu Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Feika Bian
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yu Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yunru Yu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhou325001China
| | - Zhuxiao Gu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhou325001China
| |
Collapse
|
13
|
Wesdorp MA, Schwab A, Bektas EI, Narcisi R, Eglin D, Stoddart MJ, Van Osch GJ, D'Este M. A culture model to analyze the acute biomaterial-dependent reaction of human primary neutrophils in vitro. Bioact Mater 2023; 20:627-637. [PMID: 35846845 PMCID: PMC9256821 DOI: 10.1016/j.bioactmat.2022.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/08/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Marinus A. Wesdorp
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Andrea Schwab
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ezgi Irem Bektas
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - David Eglin
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U1059 Sainbiose, Saint-Étienne, France
- Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands
| | | | - Gerjo J.V.M. Van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - Matteo D'Este
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
- Corresponding author. AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland.
| |
Collapse
|
14
|
Angeloni L, Popa B, Nouri-Goushki M, Minneboo M, Zadpoor AA, Ghatkesar MK, Fratila-Apachitei LE. Fluidic Force Microscopy and Atomic Force Microscopy Unveil New Insights into the Interactions of Preosteoblasts with 3D-Printed Submicron Patterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204662. [PMID: 36373704 DOI: 10.1002/smll.202204662] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Physical patterns represent potential surface cues for promoting osteogenic differentiation of stem cells and improving osseointegration of orthopedic implants. Understanding the early cell-surface interactions and their effects on late cellular functions is essential for a rational design of such topographies, yet still elusive. In this work, fluidic force microscopy (FluidFM) and atomic force microscopy (AFM) combined with optical and electron microscopy are used to quantitatively investigate the interaction of preosteoblasts with 3D-printed patterns after 4 and 24 h of culture. The patterns consist of pillars with the same diameter (200 nm) and interspace (700 nm) but distinct heights (500 and 1000 nm) and osteogenic properties. FluidFM reveals a higher cell adhesion strength after 24 h of culture on the taller pillars (32 ± 7 kPa versus 21.5 ± 12.5 kPa). This is associated with attachment of cells partly on the sidewalls of these pillars, thus requiring larger normal forces for detachment. Furthermore, the higher resistance to shear forces observed for these cells indicates an enhanced anchorage and can be related to the persistence and stability of lamellipodia. The study explains the differential cell adhesion behavior induced by different pillar heights, enabling advancements in the rational design of osteogenic patterns.
Collapse
Affiliation(s)
- Livia Angeloni
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Bogdan Popa
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Mahdiyeh Nouri-Goushki
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Michelle Minneboo
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Murali K Ghatkesar
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| |
Collapse
|
15
|
He Y, Gao Y, Ma Q, Zhang X, Zhang Y, Song W. Nanotopographical cues for regulation of macrophages and osteoclasts: emerging opportunities for osseointegration. J Nanobiotechnology 2022; 20:510. [PMID: 36463225 PMCID: PMC9719660 DOI: 10.1186/s12951-022-01721-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Nanotopographical cues of bone implant surface has direct influences on various cell types during the establishment of osseointegration, a prerequisite of implant bear-loading. Given the important roles of monocyte/macrophage lineage cells in bone regeneration and remodeling, the regulation of nanotopographies on macrophages and osteoclasts has arisen considerable attentions recently. However, compared to osteoblastic cells, how nanotopographies regulate macrophages and osteoclasts has not been properly summarized. In this review, the roles and interactions of macrophages, osteoclasts and osteoblasts at different stages of bone healing is firstly presented. Then, the diversity and preparation methods of nanotopographies are summarized. Special attentions are paid to the regulation characterizations of nanotopographies on macrophages polarization and osteoclast differentiation, as well as the focal adhesion-cytoskeleton mediated mechanism. Finally, an outlook is indicated of coordinating nanotopographies, macrophages and osteoclasts to achieve better osseointegration. These comprehensive discussions may not only help to guide the optimization of bone implant surface nanostructures, but also provide an enlightenment to the osteoimmune response to external implant.
Collapse
Affiliation(s)
- Yide He
- grid.233520.50000 0004 1761 4404State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
| | - Yuanxue Gao
- grid.233520.50000 0004 1761 4404State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
| | - Qianli Ma
- grid.5510.10000 0004 1936 8921Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway
| | - Xige Zhang
- grid.233520.50000 0004 1761 4404State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Shaanxi Xi’an, 710032 China
| | - Yumei Zhang
- grid.233520.50000 0004 1761 4404State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
| | - Wen Song
- grid.233520.50000 0004 1761 4404State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
| |
Collapse
|
16
|
Hosseinalizadeh H, Mahmoodpour M, Razaghi Bahabadi Z, Hamblin MR, Mirzaei H. Neutrophil mediated drug delivery for targeted glioblastoma therapy: A comprehensive review. Biomed Pharmacother 2022; 156:113841. [DOI: 10.1016/j.biopha.2022.113841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
|
17
|
Xu B, He Y, Zhang Y, Ma Z, Zhang Y, Song W. In Situ Growth of Tunable Gold Nanoparticles by Titania Nanotubes Templated Electrodeposition for Improving Osteogenesis through Modulating Macrophages Polarization. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50520-50533. [PMID: 36330544 DOI: 10.1021/acsami.2c13976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Driving macrophages M2 polarization has attracted growing attention for improving osteogenesis. Here, the in situ growth of tunable gold nanoparticles (AuNPs) on titania nanotubes (TiNTs) array was realized by electrodeposition, with the guidance of TiNTs. The fabricated Au layer showed excellent biocompatibility with different osteoimmune effects. Briefly, the Au deposition on 5 and 10 V anodized TiNTs surface could induce RAW264.7 cells to M2 polarization, whereas the Au deposition on 20 V anodized TiNTs surface showed M1 polarization, as indicated by various markers determination through immunofluorescence staining, qPCR, Western blot, and ELISA. Furthermore, the osteogenic differentiation of MC3T3-E1 was significantly enhanced by the macrophages conditioned medium from the Au@10VNTs surface. The in vivo tests also confirmed denser and thicker new trabecula bone formation and more M2 macrophages infiltration both on and adjacent to the Au@10VNTs implant surface. In mechanism, the cytokine array analysis of macrophages conditioned medium from the Au@10VNTs surface revealed the upregulation of pro-healing cytokines such as IL-10 and VEGF and downregulation of pro-inflammatory cytokines such as IL-1β and MCSF. In addition, the NF-κB pathway was significantly inhibited. In conclusion, the electrodeposition of a Au layer guided by TiNTs is a promising strategy for reducing postoperative inflammatory reactions and improving osseointegration through modulating macrophages polarization.
Collapse
Affiliation(s)
- Boya Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yide He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yan Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Zhiwei Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yumei Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Wen Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
18
|
Wang F, Qiu T, Ling Y, Yang Y, Zhou Y. Physical and Chemical Cues at the Nano–Bio Interface for Immunomodulation. Angew Chem Int Ed Engl 2022; 61:e202209499. [DOI: 10.1002/anie.202209499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Feng‐Yuan Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Department of Chemistry Fudan University Shanghai 200433 China
| | - Tianze Qiu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Department of Chemistry Fudan University Shanghai 200433 China
| | - Yun Ling
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Department of Chemistry Fudan University Shanghai 200433 China
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia Brisbane 4072 Australia
| | - Yaming Zhou
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Department of Chemistry Fudan University Shanghai 200433 China
| |
Collapse
|
19
|
Li J, Luo X, Lv ZY, Qiang HF, Hou CY, Liu K, Meng CX, Zhang YJ, Liu FZ, Zhang B. Microporous structures on mineralized collagen mediate osteogenesis by modulating the osteo-immune response of macrophages. Front Bioeng Biotechnol 2022; 10:917655. [PMID: 36105601 PMCID: PMC9464819 DOI: 10.3389/fbioe.2022.917655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
It is a new hot pot in tissue engineering and regenerative medicine to study the effects of physicochemical properties of implanted biomaterials on regulating macrophage polarization to promote bone regeneration. In this study, we designed and fabricated mineralized collagen (MC) with different microporous structures via in vitro biomimetic mineralization method. The microporous structures, mechanical properties, shore hardness and water contact angle measurements were tested. Live/dead cell staining, CCK-8 assay, phalloidine staining, staining of focal adhesions were used to detect cell behavior. ELISA, qRT-PCR, ALP, and alizarin red staining (ARS) were performed to appraise osteogenic differentiation and investigated macrophage response and their subsequent effects on the osteogenic differentiation. The results showed that RAW264.7 and MC3T3-E1 cells were able to survive on the MC. MC with the microporous structure of approximately 84 μm and 70%–80% porosity could promote M2 macrophage polarization and increase the expression level of TGF-β and VEGF. Moreover, the gene expression of the osteogenic markers ALP, COL-1, and OCN increased. Therefore, MC with different microporous structures mediated osteoimmunomodulation in bone regeneration. These data will provide a new idea of biomaterials inducing bone repair and direct the optimal design of novel immune biomaterials, development, and rational usage.
Collapse
Affiliation(s)
- Jun Li
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Xin Luo
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Zhao-Yong Lv
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Hui-Fen Qiang
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Cai-Yao Hou
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Kun Liu
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Chun-Xiu Meng
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Yu-Jue Zhang
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| | - Feng-Zhen Liu
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| | - Bin Zhang
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| |
Collapse
|
20
|
Wang FY, Qiu T, Ling Y, Yang Y, Zhou Y. Physical and Chemical Cues at Nano‐bio Interface for Immunomodulation. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Tianze Qiu
- Fudan University Department of Chemistry CHINA
| | - Yun Ling
- Fudan University Department of Chemistry CHINA
| | - Yannan Yang
- The Univeristy of Queensland AIBN The Univeristy of Queensland 4072 St lucia AUSTRALIA
| | - Yaming Zhou
- Fudan University Department of Chemistry AUSTRALIA
| |
Collapse
|
21
|
Zhao T, Chu Z, Ma J, Ouyang L. Immunomodulation Effect of Biomaterials on Bone Formation. J Funct Biomater 2022; 13:jfb13030103. [PMID: 35893471 PMCID: PMC9394331 DOI: 10.3390/jfb13030103] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Traditional bone replacement materials have been developed with the goal of directing the osteogenesis of osteoblastic cell lines toward differentiation and therefore achieving biomaterial-mediated osteogenesis, but the osteogenic effect has been disappointing. With advances in bone biology, it has been revealed that the local immune microenvironment has an important role in regulating the bone formation process. According to the bone immunology hypothesis, the immune system and the skeletal system are inextricably linked, with many cytokines and regulatory factors in common, and immune cells play an essential role in bone-related physiopathological processes. This review combines advances in bone immunology with biomaterial immunomodulatory properties to provide an overview of biomaterials-mediated immune responses to regulate bone regeneration, as well as methods to assess the bone immunomodulatory properties of bone biomaterials and how these strategies can be used for future bone tissue engineering applications.
Collapse
Affiliation(s)
- Tong Zhao
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Zhuangzhuang Chu
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Jun Ma
- Department of General Practitioners, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Correspondence: (L.O.); (J.M.); Tel.: +86-21-52039999 (L.O.); +86-21-52039999 (J.M.)
| | - Liping Ouyang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Correspondence: (L.O.); (J.M.); Tel.: +86-21-52039999 (L.O.); +86-21-52039999 (J.M.)
| |
Collapse
|
22
|
Qi D, Su J, Li S, Zhu H, Cheng L, Hua S, Yuan X, Jiang J, Shu Z, Shi Y, Xiao J. 3D printed magnesium-doped β-TCP gyroid scaffold with osteogenesis, angiogenesis, immunomodulation properties and bone regeneration capability in vivo. BIOMATERIALS ADVANCES 2022; 136:212759. [PMID: 35929304 DOI: 10.1016/j.bioadv.2022.212759] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/25/2022] [Accepted: 03/10/2022] [Indexed: 06/15/2023]
Abstract
Bioceramics have been used in orthopedic surgery for several years. Magnesium (Mg) is an essential element in bone tissue and plays an important role in bone metabolism. Mg-doped bioceramics has attracted the attention of researchers recently. However, the optimal doping amount of Mg in β-TCP and the immunomodulatory property of Mg-doped β-TCP (Mg-TCP) have not been determined yet. In this study, β-TCP scaffolds doped with different contents of magnesium oxide (0 wt%, 1 wt%, 3 wt%, and 5 wt%) with gyroid structure were printed by digital light processing (DLP) method, and the physicochemical and biological functions were then investigated. Mg-doping improved the physicochemical properties of the β-TCP scaffolds. In vitro experiments confirmed that the doping of Mg in β-TCP scaffolds promoted the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and angiogenic differentiation of endothelial progenitor cells (EPCs), where the 5Mg-TCP has the optimal properties when using the "one cell type" method. It was also found that all Mg-TCP facilitated the polarization of RAW264.7 cells to the M2 phenotype, especially the 3Mg-TCP. However, 3Mg-TCP displayed the optimal osteogenic and angiogenic potential when using a "multiple cell type" method, which referred to culturing the BMSCs or EPCs in the macrophage-conditioned medium. Finally, the in vivo experiments were conducted and the results confirmed that the 3Mg-TCP scaffolds possessed the satisfying bone defect repair capability both after 6 and 12 weeks of implantation. This study suggests that 3Mg-TCP scaffolds provide the optimal biological performance and thus have the potential for clinical translation.
Collapse
Affiliation(s)
- Dahu Qi
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Song Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Hao Zhu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Lijin Cheng
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuaibin Hua
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xi Yuan
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jiawei Jiang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Zixing Shu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Jun Xiao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
23
|
Buck E, Lee S, Gao Q, Tran SD, Tamimi F, Stone LS, Cerruti M. The Role of Surface Chemistry in the Osseointegration of PEEK Implants. ACS Biomater Sci Eng 2022; 8:1506-1521. [PMID: 35290033 DOI: 10.1021/acsbiomaterials.1c01434] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Poly(etheretherketone) (PEEK) implants suffer from poor osseointegration because of chronic inflammation. In this study, we hypothesized that adding NH2 and COOH groups to the surface of PEEK could modulate macrophage responses by altering protein adsorption and improve its osseointegration. NH2 and COOH-functionalized PEEK surfaces induced pro- and anti-inflammatory macrophage responses, respectively, and differences in protein adsorption patterns on these surfaces were related to the varied inflammatory responses. The macrophage responses to NH2 surfaces significantly reduced the osteogenic differentiation of mesenchymal stem cells (MSCs). MSCs cultured on NH2 surfaces differentiated less than those on COOH surfaces even though NH2 surfaces promoted the most mineralization in simulated body fluid solutions. After 14 days in rat tibia unicortical defects, the bone around NH2 surfaces had thinner trabeculae and higher specific bone surface than the bone around unmodified implants; surprisingly, the NH2 implants significantly increased bone-binding over the unmodified implants, while COOH implants only showed a trend for increasing bone-binding. Taken together, these results suggest that both mineral-binding and immune responses play a role in osseointegration, and PEEK implant integration may be improved with mixtures of these two functional groups to harness the ability to reduce inflammation and bind bone strongly.
Collapse
Affiliation(s)
- Emily Buck
- Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, Quebec H3A 0C5, Canada
| | - Seunghwan Lee
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada.,Alan Edwards Center for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec H3A 0G1, Canada
| | - Qiman Gao
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Simon D Tran
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Faleh Tamimi
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Laura S Stone
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada.,Alan Edwards Center for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec H3A 0G1, Canada
| | - Marta Cerruti
- Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, Quebec H3A 0C5, Canada
| |
Collapse
|
24
|
Necula MG, Mazare A, Negrescu AM, Mitran V, Ozkan S, Trusca R, Park J, Schmuki P, Cimpean A. Macrophage-like Cells Are Responsive to Titania Nanotube Intertube Spacing-An In Vitro Study. Int J Mol Sci 2022; 23:3558. [PMID: 35408918 PMCID: PMC8998567 DOI: 10.3390/ijms23073558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/23/2022] Open
Abstract
With the introduction of a new interdisciplinary field, osteoimmunology, today, it is well acknowledged that biomaterial-induced inflammation is modulated by immune cells, primarily macrophages, and can be controlled by nanotopographical cues. Recent studies have investigated the effect of surface properties in modulating the immune reaction, and literature data indicate that various surface cues can dictate both the immune response and bone tissue repair. In this context, the purpose of the present study was to investigate the effects of titanium dioxide nanotube (TNT) interspacing on the response of the macrophage-like cell line RAW 264.7. The cells were maintained in contact with the surfaces of flat titanium (Ti) and anodic TNTs with an intertube spacing of 20 nm (TNT20) and 80 nm (TNT80), under standard or pro-inflammatory conditions. The results revealed that nanotube interspacing can influence macrophage response in terms of cell survival and proliferation, cellular morphology and polarization, cytokine/chemokine expression, and foreign body reaction. While the nanostructured topography did not tune the macrophages' differentiation into osteoclasts, this behavior was significantly reduced as compared to flat Ti surface. Overall, this study provides a new insight into how nanotubes' morphological features, particularly intertube spacing, could affect macrophage behavior.
Collapse
Affiliation(s)
- Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Anca Mazare
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Advanced Institute for Materials Research (AIMR), National University Corporation Tohoku University (TU), Sendai 980-8577, Japan
| | - Andreea Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Selda Ozkan
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
| | - Roxana Trusca
- Faculty of Engineering in Foreign Languages, University Politehnica of Bucharest, 313 Splaiul Indendentei, 060042 Bucharest, Romania;
| | - Jung Park
- Department of Pediatrics, Division of Molecular Pediatrics, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Patrik Schmuki
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Czech Republic
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21569, Saudi Arabia
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| |
Collapse
|
25
|
Kyriakides TR, Kim HJ, Zheng C, Harkins L, Tao W, Deschenes E. Foreign body response to synthetic polymer biomaterials and the role of adaptive immunity. Biomed Mater 2022; 17:10.1088/1748-605X/ac5574. [PMID: 35168213 PMCID: PMC9159526 DOI: 10.1088/1748-605x/ac5574] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 02/15/2022] [Indexed: 02/06/2023]
Abstract
Implanted biomaterials elicit a series of distinct immune and repair-like responses that are collectively known as the foreign body reaction (FBR). These include processes involving innate immune inflammatory cells and wound repair cells that contribute to the encapsulation of biomaterials with a dense collagenous and largely avascular capsule. Numerous studies have shown that the early phase is dominated by macrophages that fuse to form foreign body giant cells that are considered a hallmark of the FBR. With the advent of more precise cell characterization techniques, specific macrophage subsets have been identified and linked to more or less favorable outcomes. Moreover, studies comparing synthetic- and natural-based polymer biomaterials have allowed the identification of macrophage subtypes that distinguish between fibrotic and regenerative responses. More recently, cells associated with adaptive immunity have been shown to participate in the FBR to synthetic polymers. This suggests the existence of cross-talk between innate and adaptive immune cells that depends on the nature of the implants. However, the exact participation of adaptive immune cells, such as T and B cells, remains unclear. In fact, contradictory studies suggest either the independence or dependence of the FBR on these cells. Here, we review the evidence for the involvement of adaptive immunity in the FBR to synthetic polymers with a focus on cellular and molecular components. In addition, we examine the possibility that such biomaterials induce specific antibody responses resulting in the engagement of adaptive immune cells.
Collapse
Affiliation(s)
- Themis R. Kyriakides
- Department of Biomedical Engineering, Yale University. New Haven CT 06405,Department of Pathology, Yale University. New Haven CT 06405,Vascular Biology and Therapeutics Program. Yale University. New Haven CT 06405
| | - Hyun-Je Kim
- Department of Biomedical Engineering, Yale University. New Haven CT 06405
| | - Christy Zheng
- Department of Biomedical Engineering, Yale University. New Haven CT 06405
| | - Lauren Harkins
- Department of Biomedical Engineering, Yale University. New Haven CT 06405
| | - Wanyun Tao
- Department of Biomedical Engineering, Yale University. New Haven CT 06405
| | - Emily Deschenes
- Department of Biomedical Engineering, Yale University. New Haven CT 06405
| |
Collapse
|
26
|
Yang Y, Chu C, Xiao W, Liu L, Man Y, Lin J, Qu Y. Strategies for advanced particulate bone substitutes regulating the osteo-immune microenvironment. Biomed Mater 2022; 17. [PMID: 35168224 DOI: 10.1088/1748-605x/ac5572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/15/2022] [Indexed: 02/05/2023]
Abstract
The usage of bone substitute granule materials has improved the clinical results of alveolar bone deficiencies treatment and thus broadened applications in implant dentistry. However, because of the complicated mechanisms controlling the foreign body response, no perfect solution can avoid the fibrotic encapsulation of materials till now, which may impair the results of bone regeneration, even cause the implant materials rejection. Recently, the concept of 'osteoimmunology' has been stressed. The outcomes of bone regeneration are proved to be related to the bio-physicochemical properties of biomaterials, which allow them to regulate the biological behaviours of both innate and adaptive immune cells. With the development of single cell transcriptome, the truly heterogeneity of osteo-immune cells has been clarifying, which is helpful to overcome the limitations of traditional M1/M2 macrophage nomenclature and drive the advancements of particulate biomaterials applications. This review aims at introducing the mechanisms of optimal osseointegration regulated by immune systems and provides feasible strategies for the design of next generation 'osteoimmune-smart' particulate bone substitute materials in dental clinic.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chenyu Chu
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Wenlan Xiao
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Li Liu
- State Key Laboratory of Biotherapy and Laboratory, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yi Man
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jie Lin
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yili Qu
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
27
|
Fu D, Xie D, Wang F, Chen B, Wang Z, Peng F. Mechanically Optimize T Cells Activation by Spiky Nanomotors. Front Bioeng Biotechnol 2022; 10:844091. [PMID: 35273958 PMCID: PMC8902353 DOI: 10.3389/fbioe.2022.844091] [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: 12/27/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
T cell activation is vital for immune response initiation and modulation. Except for the strength of the interaction between T cell receptors (TCR) and peptides on major histocompatibility complex molecules (MHC), mechanical force, mediated by professional mechanosensitive ion channels, contributes to activating T cells. The intrinsic characteristic of synthetic micro/nanomotors that convert diverse energy sources into physical movement and force, opening up new possibilities for T cell regulation. In this work, Pd/Au nanomotors with spiky morphology were fabricated, and in the presence of low concentrations of hydrogen peroxide fuel, the motors exhibited continuous locomotion in the cellular biological environment. Physical cues (force and pressure) generated by the dynamic performance are sensed by mechanosensitive ion channels of T cells and trigger Ca2+ influx and subsequent activation. The successful demonstration that mechanical signals generated in the bio microenvironment can potentiate T cells activation, represents a potential approach for cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Dongmei Fu
- School of Materials Science and Engineering, Sun-Yat-sen University, Guangzhou, China
| | - Dazhi Xie
- School of Materials Science and Engineering, Sun-Yat-sen University, Guangzhou, China
| | - Fei Wang
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, China
| | - Bin Chen
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, China
| | - Zhen Wang
- School of Materials Science and Engineering, Sun-Yat-sen University, Guangzhou, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun-Yat-sen University, Guangzhou, China
- *Correspondence: Fei Peng,
| |
Collapse
|
28
|
Nanoparticles Surface Chemistry Influence on Protein Corona Composition and Inflammatory Responses. NANOMATERIALS 2022; 12:nano12040682. [PMID: 35215013 PMCID: PMC8879273 DOI: 10.3390/nano12040682] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/30/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022]
Abstract
Nanoparticles are widely used for biomedical applications such as vaccine, drug delivery, diagnostics, and therapeutics. This study aims to reveal the influence of nanoparticle surface functionalization on protein corona formation from blood serum and plasma and the subsequent effects on the innate immune cellular responses. To achieve this goal, the surface chemistry of silica nanoparticles of 20 nm diameter was tailored via plasma polymerization with amine, carboxylic acid, oxazolines, and alkane functionalities. The results of this study show significant surface chemistry-induced differences in protein corona composition, which reflect in the subsequent inflammatory consequences. Nanoparticles rich with carboxylic acid surface functionalities increased the production of pro-inflammatory cytokines in response to higher level of complement proteins and decreased the number of lipoproteins found in their protein coronas. On another hand, amine rich coatings led to increased expressions of anti-inflammatory markers such as arginase. The findings demonstrate the potential to direct physiological responses to nanomaterials via tailoring their surface chemical composition.
Collapse
|
29
|
Coburn PT, Li X, Li JY, Kishimoto Y, Li-Jessen NY. Progress in Vocal Fold Regenerative Biomaterials: An Immunological Perspective. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100119. [PMID: 35434718 PMCID: PMC9007544 DOI: 10.1002/anbr.202100119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vocal folds, housed in the upper respiratory tract, are important to daily breathing, speech and swallowing functions. Irreversible changes to the vocal fold mucosae, such as scarring and atrophy, require a regenerative medicine approach to promote a controlled regrowth of the extracellular matrix (ECM)-rich mucosa. Various biomaterial systems have been engineered with an emphasis on stimulating local vocal fold fibroblasts to produce new ECM. At the same time, it is imperative to limit the foreign body reaction and associated immune components that can hinder the integration of the biomaterial into the host tissue. Modern biomaterial designs have become increasingly focused on actively harnessing the immune system to accelerate and optimize the process of tissue regeneration. An array of physical and chemical biomaterial parameters have been reported to effectively modulate local immune cells, such as macrophages, to initiate tissue repair, stimulate ECM production, promote biomaterial-tissue integration, and restore the function of the vocal folds. In this perspective paper, the unique immunological profile of the vocal folds will first be reviewed. Key physical and chemical biomaterial properties relevant to immunomodulation will then be highlighted and discussed. A further examination of the physicochemical properties of recent vocal fold biomaterials will follow to generate deeper insights into corresponding immune-related outcomes. Lastly, a perspective will be offered on the opportunity of integrating material-led immunomodulatory strategies into future vocal fold tissue engineering therapies.
Collapse
Affiliation(s)
- Patrick T. Coburn
- School of Communication Sciences and Disorders, McGill University, Canada
| | - Xuan Li
- Department of Mechanical Engineering, McGill University, Canada
| | - Jianyu. Y. Li
- Department of Mechanical Engineering, McGill University, Canada
- Department of Biomedical Engineering, McGill University, Canada
| | - Yo Kishimoto
- Department of Otolaryngology – Head & Neck Surgery, Kyoto University, Kyoto, Japan
| | - Nicole Y.K. Li-Jessen
- School of Communication Sciences and Disorders, McGill University, Canada
- Department of Biomedical Engineering, McGill University, Canada
- Department of Otolaryngology – Head & Neck Surgery, McGill University, Canada
| |
Collapse
|
30
|
Ishmukhametov I, Batasheva S, Rozhina E, Akhatova F, Mingaleeva R, Rozhin A, Fakhrullin R. DNA/Magnetic Nanoparticles Composite to Attenuate Glass Surface Nanotopography for Enhanced Mesenchymal Stem Cell Differentiation. Polymers (Basel) 2022; 14:344. [PMID: 35054750 PMCID: PMC8779295 DOI: 10.3390/polym14020344] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have extensive pluripotent potential to differentiate into various cell types, and thus they are an important tool for regenerative medicine and biomedical research. In this work, the differentiation of hTERT-transduced adipose-derived MSCs (hMSCs) into chondrocytes, adipocytes and osteoblasts on substrates with nanotopography generated by magnetic iron oxide nanoparticles (MNPs) and DNA was investigated. Citrate-stabilized MNPs were synthesized by the chemical co-precipitation method and sized around 10 nm according to microscopy studies. It was shown that MNPs@DNA coatings induced chondrogenesis and osteogenesis in hTERT-transduced MSCs. The cells had normal morphology and distribution of actin filaments. An increase in the concentration of magnetic nanoparticles resulted in a higher surface roughness and reduced the adhesion of cells to the substrate. A glass substrate modified with magnetic nanoparticles and DNA induced active chondrogenesis of hTERT-transduced MSC in a twice-diluted differentiation-inducing growth medium, suggesting the possible use of nanostructured MNPs@DNA coatings to obtain differentiated cells at a reduced level of growth factors.
Collapse
Affiliation(s)
| | | | - Elvira Rozhina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kreml uramı 18, 420008 Kazan, Republic of Tatarstan, Russian Federation; (I.I.); (S.B.); (F.A.); (R.M.); (A.R.)
| | | | | | | | - Rawil Fakhrullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kreml uramı 18, 420008 Kazan, Republic of Tatarstan, Russian Federation; (I.I.); (S.B.); (F.A.); (R.M.); (A.R.)
| |
Collapse
|
31
|
González García LE, Ninan N, Simon J, Madathiparambil Visalakshan R, Bright R, Wahono SK, Ostrikov K, Mailänder V, Landfester K, Goswami N, Vasilev K. Ultra-small gold nanoclusters assembled on plasma polymer-modified zeolites: a multifunctional nanohybrid with anti-haemorrhagic and anti-inflammatory properties. NANOSCALE 2021; 13:19936-19945. [PMID: 34820678 DOI: 10.1039/d1nr06591b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hemostatic agents are pivotal for managing clinical and traumatic bleeding during emergency and domestic circumstances. Herein, a novel functional hybrid nanocomposite material consisting of plasma polymer-modified zeolite 13X and ultra-small gold nanoclusters (AuNCs) was fabricated as an efficient hemostatic agent. The surface of zeolite 13X was functionalised with amine groups which served as binding sites for carboxylate terminated AuNCs. Protein corona studies revealed the enhanced adsorption of two proteins, namely, coagulation factors and plasminogen as a result of AuNCs immobilization on the zeolite surface. The immune response studies showed that the hybrid nanocomposites are effective in reducing inflammation, which combined with a greater attachment of vitronectin, may promote wound healing. The hemostatic potential of the nanocomposite could be directly correlated with their immunomodulatory and anti-haemorrhagic properties. Together, the hybrid nanoengineered material developed in this work could provide a new avenue to tackle life-threatening injuries in civilian and other emergencies.
Collapse
Affiliation(s)
- Laura E González García
- Academic Unit of STEM, The University of South Australia, Mawson Lakes, SA 5095, Australia.
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Neethu Ninan
- Academic Unit of STEM, The University of South Australia, Mawson Lakes, SA 5095, Australia.
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Johanna Simon
- Max-Planck-Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | | | - Richard Bright
- Academic Unit of STEM, The University of South Australia, Mawson Lakes, SA 5095, Australia.
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Satriyo K Wahono
- Research Division for Natural Product Technology, Indonesian Institutes of Sciences, Jl. Jogja-Wonosari km 32, Gading, Playen, Gunungkidul, Yogyakarta 55861, Indonesia
| | - Kostya Ostrikov
- School of Chemistry and Physics, Centre for Materials Science, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Volker Mailänder
- Max-Planck-Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max-Planck-Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Nirmal Goswami
- Academic Unit of STEM, The University of South Australia, Mawson Lakes, SA 5095, Australia.
- Materials Chemistry Department, CSIR-Institute of Minerals and Materials Technology, Acharya Vihar, Bhubaneswar-751013, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201 002, India
| | - Krasimir Vasilev
- Academic Unit of STEM, The University of South Australia, Mawson Lakes, SA 5095, Australia.
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| |
Collapse
|
32
|
Mijanović O, Pylaev T, Nikitkina A, Artyukhova M, Branković A, Peshkova M, Bikmulina P, Turk B, Bolevich S, Avetisov S, Timashev P. Tissue Engineering Meets Nanotechnology: Molecular Mechanism Modulations in Cornea Regeneration. MICROMACHINES 2021; 12:mi12111336. [PMID: 34832752 PMCID: PMC8618371 DOI: 10.3390/mi12111336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/23/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Nowadays, tissue engineering is one of the most promising approaches for the regeneration of various tissues and organs, including the cornea. However, the inability of biomaterial scaffolds to successfully integrate into the environment of surrounding tissues is one of the main challenges that sufficiently limits the restoration of damaged corneal tissues. Thus, the modulation of molecular and cellular mechanisms is important and necessary for successful graft integration and long-term survival. The dynamics of molecular interactions affecting the site of injury will determine the corneal transplantation efficacy and the post-surgery clinical outcome. The interactions between biomaterial surfaces, cells and their microenvironment can regulate cell behavior and alter their physiology and signaling pathways. Nanotechnology is an advantageous tool for the current understanding, coordination, and directed regulation of molecular cell-transplant interactions on behalf of the healing of corneal wounds. Therefore, the use of various nanotechnological strategies will provide new solutions to the problem of corneal allograft rejection, by modulating and regulating host-graft interaction dynamics towards proper integration and long-term functionality of the transplant.
Collapse
Affiliation(s)
- Olja Mijanović
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- Correspondence:
| | - Timofey Pylaev
- Saratov Medical State University N.A. V.I. Razumovsky, 112 Bolshaya Kazachya St., 410012 Saratov, Russia;
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, 410049 Saratov, Russia
| | - Angelina Nikitkina
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
| | - Margarita Artyukhova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
| | - Ana Branković
- Department of Forensic Engineering, University of Criminal Investigation and Police Studies, 196 Cara Dušana St., Belgrade 11000, Serbia;
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Boris Turk
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sergey Bolevich
- Department of Human Pathology, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia;
| | - Sergei Avetisov
- Department of Eye Diseases, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia;
- Research Institute of Eye Diseases, 11 Rossolimo St., 119021 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, 119991 Moscow, Russia
| |
Collapse
|
33
|
Muhammad W, Zhai Z, Wang S, Gao C. Inflammation-modulating nanoparticles for pneumonia therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1763. [PMID: 34713969 DOI: 10.1002/wnan.1763] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
Pneumonia is a common but serious infectious disease, and is the sixth leading cause for death. The foreign pathogens such as viruses, fungi, and bacteria establish an inflammation response after interaction with lung, leading to the filling of bronchioles and alveoli with fluids. Although the pharmacotherapies have shown their great effectiveness to combat pathogens, advanced methods are under developing to treat complicated cases such as virus-infection and lung inflammation or acute lung injury (ALI). The inflammation modulation nanoparticles (NPs) can effectively suppress immune cells and inhibit inflammatory molecules in the lung site, and thereby alleviate pneumonia and ALI. In this review, the pathological inflammatory microenvironments in pneumonia, which are instructive for the design of biomaterials therapy, are summarized. The focus is then paid to the inflammation-modulating NPs that modulate the inflammatory cells, cytokines and chemokines, and microenvironments of pneumonia for better therapeutic effects. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease.
Collapse
Affiliation(s)
- Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Strohbach A, Busch R. Predicting the In Vivo Performance of Cardiovascular Biomaterials: Current Approaches In Vitro Evaluation of Blood-Biomaterial Interactions. Int J Mol Sci 2021; 22:ijms222111390. [PMID: 34768821 PMCID: PMC8583792 DOI: 10.3390/ijms222111390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 12/29/2022] Open
Abstract
The therapeutic efficacy of a cardiovascular device after implantation is highly dependent on the host-initiated complement and coagulation cascade. Both can eventually trigger thrombosis and inflammation. Therefore, understanding these initial responses of the body is of great importance for newly developed biomaterials. Subtle modulation of the associated biological processes could optimize clinical outcomes. However, our failure to produce truly blood compatible materials may reflect our inability to properly understand the mechanisms of thrombosis and inflammation associated with biomaterials. In vitro models mimicking these processes provide valuable insights into the mechanisms of biomaterial-induced complement activation and coagulation. Here, we review (i) the influence of biomaterials on complement and coagulation cascades, (ii) the significance of complement-coagulation interactions for the clinical success of cardiovascular implants, (iii) the modulation of complement activation by surface modifications, and (iv) in vitro testing strategies.
Collapse
Affiliation(s)
- Anne Strohbach
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany;
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Fleischmannstr. 42-44, 17489 Greifswald, Germany
- Correspondence:
| | - Raila Busch
- Department of Internal Medicine B Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany;
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Fleischmannstr. 42-44, 17489 Greifswald, Germany
| |
Collapse
|
35
|
Dabare PRL, Bachhuka A, Visalakshan RM, Shirazi HS, Ostriko K, Smith LE, Vasilev K. Mechanistic Insight in Surface Nanotopography Driven Cellular Migration. ACS Biomater Sci Eng 2021; 7:4921-4932. [PMID: 34477378 DOI: 10.1021/acsbiomaterials.1c00853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cellular migration plays a vital role in many physiological processes. To elucidate the role of surface nanotopography on the downstream signaling pathways underlying cell migration, model surfaces having well-defined hill-like surface nanotopography and uniform surface chemistry were designed and implemented using plasma polymerization and covalent attachment of nanoparticles of predetermined size. A scratch wound assay, immunostaining, and gene expression of focal adhesion (FA) proteins were performed to determine the influence of surface nanotopography on cell migration. The results of this study demonstrate that the gap closure between cell monolayers is faster on surfaces having greater nanoscale topography. The phenomenon is predominantly driven by cell migration and was independent from cell proliferation. Qualitative and quantitative assessment of proteins involved in the signaling pathways underlying cell migration showed significant modulation by surface nanotopography. Specifically, focal adhesion sites decreased with the increase in surface nanotopography scale while the expression of FA proteins increased. This implies that nanotopography mediated modulation of cell migration is directly governed by the recruitment of receptor and adapter proteins responsible for cell-surface interaction. The results of this study indicate that biomaterial devices and constructs having rationally designed surface nanotopography and chemistry could be utilized to regulate wound healing and tissue regeneration.
Collapse
Affiliation(s)
| | - Akash Bachhuka
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Rahul M Visalakshan
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Hanieh S Shirazi
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Kostya Ostriko
- School of Chemistry and Physics, Centre for Materials Science and Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Louise E Smith
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Krasimir Vasilev
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia.,Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| |
Collapse
|
36
|
Richard AS, Verma RS. Bioactive nano yarns as surgical sutures for wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112334. [PMID: 34474885 DOI: 10.1016/j.msec.2021.112334] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 11/19/2022]
Abstract
Surgical sutures are the most widely used medical device in any surgical procedure worldwide. In this study, modified electrospinning technique has been used as manufacturing technique to produce nanofiber bundles twisted simultaneously to obtain nanofiber yarns. Taking the advantage of nanofiber yarns in terms of biomimetic structure, mechanical strength and handling properties, the material is chosen. Curcumin, a natural compound is incorporated to the nanofiber yarns by blend electrospinning technique for its anti-inflammatory, antibiotic and wound healing properties. The synthesized nanofiber yarns were characterized by various characterization techniques such as XRD, FTIR, SEM, Tensile testing, stem cell interaction, hemocompatibility, bacterial response, drug release profiling and in vivo studies. Curcumin loaded nanofiber yarns demonstrated sustained release with improved antibacterial, antiplatelet, cell migration and stem cell interaction in vitro. The results from skin inflammation animal model revealed that curcumin laden nanofiber yarn suture manifested reduced inflammation and cellularity. The three dimensional structure, adequate mechanical strength and biological properties of the nanofiber yarn provide naive environment for wound healing with the balanced degradation of suture material in rat model.
Collapse
Affiliation(s)
- Arthi Sunil Richard
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Rama Shankar Verma
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
37
|
Harawaza K, Cousins B, Roach P, Fernandez A. Modification of the surface nanotopography of implant devices: A translational perspective. Mater Today Bio 2021; 12:100152. [PMID: 34746736 PMCID: PMC8554633 DOI: 10.1016/j.mtbio.2021.100152] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/24/2023] Open
Abstract
There is an increasing need for the development of superior, safe, and more sophisticated implants, especially as our society historically has been moving towards an increasingly aging population. Currently, most research is being focused on the next generation of advanced medical implants, that are not only biocompatible but have modified surfaces that direct specific immunomodulation at cellular level. While there is a plethora of information on cell-surface interaction and how surfaces can be nanofabricated at research level, less is known about how the academic knowledge has been translated into clinical trials and commercial technologies. In this review, we provide a clinical translational perspective on the use of controlled physical surface modification of medical implants, presenting an analysis of data acquired from clinical trials and commercial products. We also evaluate the state-of-the-art of nanofabrication techniques that are being applied for implant surface modification at a clinical level. Finally, we identify some current challenges in the field, including the need of more advanced nanopatterning techniques, the comparatively small number of clinical trials and comment on future avenues to be explored for a successful clinical translation.
Collapse
Affiliation(s)
- K. Harawaza
- Chemistry Department, School of Science, Loughborough University, Loughborough, LE11 3TU, UK
| | - B. Cousins
- Chemistry Department, School of Science, Loughborough University, Loughborough, LE11 3TU, UK
| | - P. Roach
- Chemistry Department, School of Science, Loughborough University, Loughborough, LE11 3TU, UK
| | - A. Fernandez
- Chemistry Department, School of Science, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
38
|
Wu S, Xia Y, Hu Y, Ma G. Bio-mimic particles for the enhanced vaccinations: Lessons learnt from the natural traits and pathogenic invasion. Adv Drug Deliv Rev 2021; 176:113871. [PMID: 34311014 DOI: 10.1016/j.addr.2021.113871] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
In the combat against pathogens, the immune systems were evolved with the immune recognitions against the various danger signals, which responded vigorously upon the pathogen invasions and elicited potent antibodies or T cell engagement against the re-infections. Envisage with the prevailing pandemics and increasing demands for cancer vaccines, bio-mimic particles were developed to imitate the natural traits of the pathogens, which conferred the optimal strategies to stimulate the immune engagement and let to the increased vaccine efficacy. Here, the recent development in bio-mimic particles, as well as the natural cues from the pathogens were discussed. As such, the designing principles that adapted from the physiochemical properties of the pathogens were unfolded as the surface characteristics (hydrophobic, nano-pattern, antigen display, charge), properties (size, shape, softness) and the delivered components (peptide, protein, nuclear acids, toll-like receptor (TLR) agonist, antibody). Additionally, the strategies for the efficient delivery, regarding the biodistribution, internalization and presentation of the antigens were also illustrated. Through reviewing the state-of-art in biomimetic particles, the lesson learnt from the natural traits and pathogenic invasion may shed light on the rational design for the enhanced vaccinations.
Collapse
|
39
|
Mackay BS, Marshall K, Grant-Jacob JA, Kanczler J, Eason RW, Oreffo ROC, Mills B. The future of bone regeneration: integrating AI into tissue engineering. Biomed Phys Eng Express 2021; 7. [PMID: 34271556 DOI: 10.1088/2057-1976/ac154f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/16/2021] [Indexed: 01/16/2023]
Abstract
Tissue engineering is a branch of regenerative medicine that harnesses biomaterial and stem cell research to utilise the body's natural healing responses to regenerate tissue and organs. There remain many unanswered questions in tissue engineering, with optimal biomaterial designs still to be developed and a lack of adequate stem cell knowledge limiting successful application. Advances in artificial intelligence (AI), and deep learning specifically, offer the potential to improve both scientific understanding and clinical outcomes in regenerative medicine. With enhanced perception of how to integrate artificial intelligence into current research and clinical practice, AI offers an invaluable tool to improve patient outcome.
Collapse
Affiliation(s)
- Benita S Mackay
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Karen Marshall
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - James A Grant-Jacob
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - Robert W Eason
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Ben Mills
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| |
Collapse
|
40
|
Heydari P, Kharaziha M, Varshosaz J, Javanmard SH. Current knowledge of immunomodulation strategies for chronic skin wound repair. J Biomed Mater Res B Appl Biomater 2021; 110:265-288. [PMID: 34318595 DOI: 10.1002/jbm.b.34921] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022]
Abstract
In orchestrating the wound healing process, the immune system plays a critical role. Hence, controlling the immune system to repair skin defects is an attractive approach. The highly complex immune system includes the coordinated actions of several immune cells, which can produce various inflammatory and antiinflammatory cytokines and affect the healing of skin wounds. This process can be optimized using biomaterials, bioactive molecules, and cell delivery. The present review discusses various immunomodulation strategies for supporting the healing of chronic wounds. In this regard, following the evolution of the immune system and its role in the wound healing mechanism, the interaction between the extracellular mechanism and immune cells for acceleration wound healing will be firstly investigated. Consequently, the immune-based chronic wounds will be briefly examined and the mechanism of progression, and conventional methods of their treatment are evaluated. In the following, various biomaterials-based immunomodulation strategies are introduced to stimulate and control the immune system to treat and regenerate skin defects. Other effective methods of controlling the immune system in wound healing which is the release of bioactive agents (such as antiinflammatory, antigens, and immunomodulators) and stem cell therapy at the site of injury are reviewed.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Jaleh Varshosaz
- School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
41
|
Jin S, Yang R, Chu C, Hu C, Zou Q, Li Y, Zuo Y, Man Y, Li J. Topological structure of electrospun membrane regulates immune response, angiogenesis and bone regeneration. Acta Biomater 2021; 129:148-158. [PMID: 34082097 DOI: 10.1016/j.actbio.2021.05.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023]
Abstract
The fate of biomaterials is orchestrated by biocompatibility and bioregulation characteristics, reported to be closely related to topographical structures. For the purpose to investigate the topography of fibrous membranes on the guided bone regeneration performance, we successfully fabricated poly (lactate-co-glycolate)/fish collagen/nano-hydroxyapatite (PFCH) fibrous membranes with random, aligned and latticed topography by electrospinning. The physical, chemical and biological properties of the three topographical PFCH membranes were systematically investigated by in vitro and in vivo experiments. The subcutaneous implantation of C57BL6 mice showed an acceptable mild foreign body reaction of all three topological membranes. Interestingly, the latticed PFCH membrane exhibited superior abilities to recruit macrophage/monocyte and induce angiogenesis. We further investigated the osteogenesis of the three topographical PFCH membranes via the critical-size calvarial bone defect model of rats and mice and the results suggested that latticed PFCH membrane manifested promising performance to promote angiogenesis through upregulation of the HIF-1α signaling pathway; thereby enhancing bone regeneration. Our research illustrated that the topological structure of fibrous membranes, as one of the characteristics of biomaterials, could regulate its biological functions, and the fibrous structure of latticed topography could serve as a favorable surface design of biomaterials for bone regeneration. STATEMENT OF SIGNIFICANCE: In material-mediated regeneration medicine, the interaction between the biomaterial and the host is key to successful tissue regeneration. The micro-and nano-structure becomes one of the most critical physical clues for designing biomaterials. In this study, we fabricated three topological electrospun membranes (Random, Aligned and Latticed) to understand how topological structural clues mediate bone tissue regeneration. Interestingly, we found that the Latticed topographical PFCH membrane promotes macrophage recruitment, angiogenesis, and osteogenesis in vivo, indicating the fibrous structure of latticed topography could serve as a favorable surface design of biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Shue Jin
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, Sichuan 610065, China
| | - Renli Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Oral Implantology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Chu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Oral Implantology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chen Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Oral Implantology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qin Zou
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yubao Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yi Zuo
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Oral Implantology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jidong Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, Sichuan 610065, China.
| |
Collapse
|
42
|
Liu X, Wang Y, He Y, Wang X, Zhang R, Bachhuka A, Madathiparambil Visalakshan R, Feng Q, Vasilev K. Synergistic Effect of Surface Chemistry and Surface Topography Gradient on Osteogenic/Adipogenic Differentiation of hMSCs. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30306-30316. [PMID: 34156811 DOI: 10.1021/acsami.1c03915] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Much attention has been paid to understanding the individual effects of surface chemistry or topography on cell behavior. However, the synergistic influence of both surface chemistry and surface topography on differentiation of human mesenchymal stem cells (hMSCs) should also be addressed. Here, gold nanoparticles were immobilized in an increasing number density manner to achieve a surface topography gradient; a thin film rich in amine (-NH2) or methyl (-CH3) chemical groups was plasma-polymerized to adjust the surface chemistry of the outermost layer (ppAA and ppOD, respectively). hMSCs were cultured on these model substrates with defined surface chemistry and surface topography gradient. The morphology and focal adhesion (FA) formation of hMSCs were first examined. hMSC differentiation was then co-induced in osteogenic and adipogenic medium, as well as in the presence of extracellular-signal-regulated kinase1/2 (ERK1/2) and RhoA/Rho-associated protein kinase (ROCK) inhibitors. The results show that the introduction of nanotopography could enhance FA formation and osteogenesis but inhibited adipogenesis on both ppAA and ppOD surfaces, indicating that the surface chemistry could regulate hMSC differentiation, in a surface topography-dependent manner. RhoA/ROCK and ERK1/2 signaling pathways may participate in this process. This study demonstrated that surface chemistry and surface topography can jointly affect cell morphology, FA formation, and thus osteogenic/adipogenic differentiation of hMSCs. These findings highlight the importance of the synergistic effect of different material properties on regulation of cell response, which has important implications in designing functional biomaterials.
Collapse
Affiliation(s)
- Xujie Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yakun Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaofeng Wang
- Department of Hand Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315040, China
| | - Ranran Zhang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Akash Bachhuka
- Unit of STEM, University of South Australia, Mawson Lakes 5095, Australia
| | | | - Qingling Feng
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Krasimir Vasilev
- Unit of STEM, University of South Australia, Mawson Lakes 5095, Australia
| |
Collapse
|
43
|
Tao C, Wang D. Tissue Engineering for Mimics and Modulations of Immune Functions. Adv Healthc Mater 2021; 10:e2100146. [PMID: 33871178 DOI: 10.1002/adhm.202100146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/21/2021] [Indexed: 11/12/2022]
Abstract
In the field of regenerative medicine, advances in tissue engineering have surpassed the reconstruction of individual tissues or organs and begun to work towards engineering systemic factors such as immune objects and functions. The immune system plays a crucial role in protecting and regulating systemic functions in the human body. Engineered immune tissues and organs have shown potential in recovering dysfunctions and aplasia of the immune system and the evasion from immune-mediated inflammatory responses and rejection elicited by engineered implants from allogeneic or xenogeneic sources are also being pursued to facilitate clinical transplantation of tissue engineered grafts. Here, current progress in tissue engineering to mimic or modulate immune functions is reviewed and elaborated from two perspectives: 1) engineering of immune tissues and organs per se and 2) immune evasion of host immunoinflammatory rejection by tissue-engineered implants.
Collapse
Affiliation(s)
- Chao Tao
- Department of Biomedical Engineering City University of Hong Kong 83 Tat Chee Avenue Kowloon Hong Kong SAR China
| | - Dong‐An Wang
- Department of Biomedical Engineering City University of Hong Kong 83 Tat Chee Avenue Kowloon Hong Kong SAR China
- Karolinska Institute Ming Wai Lau Centre for Reparative Medicine HKSTP Sha Tin Hong Kong SAR China
- Shenzhen Research Institute City University of Hong Kong Shenzhen 518057 P. R. China
| |
Collapse
|
44
|
Buck E, Lee S, Stone LS, Cerruti M. Protein Adsorption on Surfaces Functionalized with COOH Groups Promotes Anti-inflammatory Macrophage Responses. ACS APPLIED MATERIALS & INTERFACES 2021; 13:7021-7036. [PMID: 33539069 DOI: 10.1021/acsami.0c16509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Implants can induce a foreign body reaction that leads to chronic inflammation and fibrosis in the surrounding tissue. Macrophages help detect the foreign material, play a role in the inflammatory response, and may promote fibrosis instead of the desired tissue regeneration around implants. Implant surface properties impact macrophage responses by changing the nature of the adsorbed protein layer, but conflicting studies highlight the complexity of this relationship. In this study, the effect of surface chemistry on macrophage behavior was investigated with poly(styrene) surfaces containing common functional groups at similar surface densities. The protein layer was characterized to identify the proteins that adsorbed on the surfaces from the medium and the proteins secreted onto the surfaces by adherent macrophages. Of the surface chemistries studied, carboxylic acid (COOH) groups promoted anti-inflammatory responses from unstimulated macrophages and did not exacerbate inflammation upon stimulation. These surfaces also enhanced the adsorption of proteins involved in integrin signaling and promoted the secretion of proteins related to angiogenesis, integrin signaling, and cytokine signaling, which have been previously associated with improved biomaterial integration. Therefore, this study suggests that surface modification with COOH groups may help improve the integration of implants in the body by enhancing anti-inflammatory macrophage responses through altered protein adsorption.
Collapse
Affiliation(s)
- Emily Buck
- Department of Mining and Materials Engineering, McGill University, Montreal, QC H3A 0C5, Canada
| | - Seunghwan Lee
- Faculty of Dentistry, McGill University, Montreal, QC H3A 1G1, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QC H3A 0G1, Canada
| | - Laura S Stone
- Faculty of Dentistry, McGill University, Montreal, QC H3A 1G1, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QC H3A 0G1, Canada
| | - Marta Cerruti
- Department of Mining and Materials Engineering, McGill University, Montreal, QC H3A 0C5, Canada
| |
Collapse
|
45
|
Welch NG, Winkler DA, Thissen H. Antifibrotic strategies for medical devices. Adv Drug Deliv Rev 2020; 167:109-120. [PMID: 32553685 DOI: 10.1016/j.addr.2020.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
A broad range of medical devices initiate an immune reaction known as the foreign body response (FBR) upon implantation. Here, collagen deposition at the surface of the implant occurs as a result of the FBR, ultimately leading to fibrous encapsulation and, in many cases, reduced function or failure of the device. Despite significant efforts, the prevention of fibrotic encapsulation has not been realized at this point in time. However, many next-generation medical technologies including cellular therapies, sensors and devices depend on the ability to modulate and control the FBR. For these technologies to become viable, significant advances must be made in understanding the underlying mechanism of this response as well as in the methods modulating this response. In this review, we highlight recent advances in the development of materials and coatings providing a reduced FBR and emphasize key characteristics of high-performing approaches. We also provide a detailed overview of the state-of-the-art in strategies relying on controlled drug release, the surface display of bioactive signals, materials-based approaches, and combinations of these approaches. Finally, we offer perspectives on future directions in this field.
Collapse
|
46
|
Chen Y, Wang J, Li X, Hu N, Voelcker NH, Xie X, Elnathan R. Emerging Roles of 1D Vertical Nanostructures in Orchestrating Immune Cell Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001668. [PMID: 32844502 PMCID: PMC7461044 DOI: 10.1002/adma.202001668] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/16/2020] [Indexed: 05/07/2023]
Abstract
Engineered nano-bio cellular interfaces driven by 1D vertical nanostructures (1D-VNS) are set to prompt radical progress in modulating cellular processes at the nanoscale. Here, tuneable cell-VNS interfacial interactions are probed and assessed, highlighting the use of 1D-VNS in immunomodulation, and intracellular delivery into immune cells-both crucial in fundamental and translational biomedical research. With programmable topography and adaptable surface functionalization, 1D-VNS provide unique biophysical and biochemical cues to orchestrate innate and adaptive immunity, both ex vivo and in vivo. The intimate nanoscale cell-VNS interface leads to membrane penetration and cellular deformation, facilitating efficient intracellular delivery of diverse bioactive cargoes into hard-to-transfect immune cells. The unsettled interfacial mechanisms reported to be involved in VNS-mediated intracellular delivery are discussed. By identifying up-to-date progress and fundamental challenges of current 1D-VNS technology in immune-cell manipulation, it is hoped that this report gives timely insights for further advances in developing 1D-VNS as a safe, universal, and highly scalable platform for cell engineering and enrichment in advanced cancer immunotherapy such as chimeric antigen receptor-T therapy.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVIC3168Australia
- INM‐Leibniz Institute for New MaterialsCampus D2 2Saarbrücken66123Germany
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Roey Elnathan
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
| |
Collapse
|
47
|
Ni S, Zhai D, Huan Z, Zhang T, Chang J, Wu C. Nanosized concave pit/convex dot microarray for immunomodulatory osteogenesis and angiogenesis. NANOSCALE 2020; 12:16474-16488. [PMID: 32743625 DOI: 10.1039/d0nr03886e] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The immunomodulatory capability of biomaterials is of paramount importance for successful material-mediated bone regeneration. Particularly, the design of surface nano-topography can be leveraged to instruct immune reactions, yet the understanding of such "nano-morphology effect" is still very limited. Herein, highly ordered nano-concave pit (denoted as NCPit) and nano-convex dot (denoted as NCDot) microarrays with two different sizes were successfully constructed on a 316LSS surface via anodization and subsequently immersion-coating treatment, respectively. We, for the first time, comparatively investigated the interactions of NCPit and NCDot microarrays with RAW264.7 macrophages and their immunomodulatory impacts on osteogenesis and angiogenesis of human bone mesenchymal stem cells (hBMSCs) and human umbilical vein endothelial cells (HUVECs). NCDot microarrays induced macrophages towards M2 polarization with the higher expression level of anti-inflammatory markers (IL-10 and CD 206) and the lower level of pro-inflammatory markers (TNF-α, IL-1β, IL-6 and CD 86) than those of the corresponding NCPit microarrays. During the process, the expressions of osteogenesis-related genes (Runx2, OPN and OCN) of hBMSCs, and angiogenesis-related genes (eNOS, HIF-1α, KDR and VEGF) of HUVECs were significantly upregulated by the NCDot microarray-modulating immune microenvironment of macrophages, and finally stimulated osteogenesis and angiogenesis. Thus, the prepared NCDot arrays were able to significantly promote osteo-/angiogenic activity by generating a more suitable immune microenvironment than NCPit arrays, offering substantial evidence for designing immunomodulatory biomaterials with specific microstructures and optimal bioactivity.
Collapse
Affiliation(s)
- Siyu Ni
- College of Chemistry, Chemical Engineering and Biotechnology; Donghua University, North Renmin Road 2999, Shanghai 201620, P. R. China
| | | | | | | | | | | |
Collapse
|
48
|
Rahmati M, Silva EA, Reseland JE, A Heyward C, Haugen HJ. Biological responses to physicochemical properties of biomaterial surface. Chem Soc Rev 2020; 49:5178-5224. [PMID: 32642749 DOI: 10.1039/d0cs00103a] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biomedical scientists use chemistry-driven processes found in nature as an inspiration to design biomaterials as promising diagnostic tools, therapeutic solutions, or tissue substitutes. While substantial consideration is devoted to the design and validation of biomaterials, the nature of their interactions with the surrounding biological microenvironment is commonly neglected. This gap of knowledge could be owing to our poor understanding of biochemical signaling pathways, lack of reliable techniques for designing biomaterials with optimal physicochemical properties, and/or poor stability of biomaterial properties after implantation. The success of host responses to biomaterials, known as biocompatibility, depends on chemical principles as the root of both cell signaling pathways in the body and how the biomaterial surface is designed. Most of the current review papers have discussed chemical engineering and biological principles of designing biomaterials as separate topics, which has resulted in neglecting the main role of chemistry in this field. In this review, we discuss biocompatibility in the context of chemistry, what it is and how to assess it, while describing contributions from both biochemical cues and biomaterials as well as the means of harmonizing them. We address both biochemical signal-transduction pathways and engineering principles of designing a biomaterial with an emphasis on its surface physicochemistry. As we aim to show the role of chemistry in the crosstalk between the surface physicochemical properties and body responses, we concisely highlight the main biochemical signal-transduction pathways involved in the biocompatibility complex. Finally, we discuss the progress and challenges associated with the current strategies used for improving the chemical and physical interactions between cells and biomaterial surface.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway. h.j.haugen.odont.uio.no
| | | | | | | | | |
Collapse
|
49
|
Influence of Extracellular Mimicked Hierarchical Nano-Micro-Topography on the Bacteria/Abiotic Interface. Polymers (Basel) 2020; 12:polym12040828. [PMID: 32260531 PMCID: PMC7240582 DOI: 10.3390/polym12040828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 11/29/2022] Open
Abstract
The study of interfaces between engineered surfaces and prokaryotic cells is a subject whose actual relevance has been reinforced by the current outbreaks due to unknown viruses and antibiotic-resistant bacteria. Studies aiming at the development of antibacterial surfaces are based on two pillars: surface chemistry or topographical cues. This work reports the study of only the topographic aspect by the development of thin films of polyamide, which present attractive surface chemistry for bacterial adhesion. The same chemistry with only nano- or hierarchical nano- and micro-topography that mimics the extracellular matrix is obtained by sputter-depositing the thin films onto Si and polydimethylsiloxane (PDMS), respectively. The surface average roughness of the Si-modified surfaces was around 1 nm, while the hierarchical topography presented values from 750 to 1000 nm, with wavelengths and amplitudes ranging from 15–30 µm and 1–3 µm, respectively, depending on the deposition parameters. The surface topography, wettability, surface charge, and mechanical properties were determined and related to interface performance with two Gram+ and two Gram- bacterial strains. The overall results show that surfaces with only nano-topographic features present less density of bacteria, regardless of their cell wall composition or cell shape, if the appropriate surface chemistry is present.
Collapse
|
50
|
Bachhuka A, Madathiparambil Visalakshan R, Law CS, Santos A, Ebendorff-Heidepriem H, Karnati S, Vasilev K. Modulation of Macrophages Differentiation by Nanoscale-Engineered Geometric and Chemical Features. ACS APPLIED BIO MATERIALS 2020; 3:1496-1505. [DOI: 10.1021/acsabm.9b01125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- A. Bachhuka
- ARC Center of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia
- Institute for Photonics and Advanced Sensing (IPAS), The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - R. Madathiparambil Visalakshan
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Adelaide, South Australia 5095, Australia
| | - C. S. Law
- ARC Center of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia
- Institute for Photonics and Advanced Sensing (IPAS), The University of Adelaide, Adelaide, South Australia 5005, Australia
- School of Chemical Engineering, University of Adelaide, Engineering North Building, Adelaide, South Australia 5005, Australia
| | - A. Santos
- ARC Center of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia
- Institute for Photonics and Advanced Sensing (IPAS), The University of Adelaide, Adelaide, South Australia 5005, Australia
- School of Chemical Engineering, University of Adelaide, Engineering North Building, Adelaide, South Australia 5005, Australia
| | - H. Ebendorff-Heidepriem
- ARC Center of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia
- Institute for Photonics and Advanced Sensing (IPAS), The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - S. Karnati
- Institute for Anatomy and Cell Biology, Julius Maximilians University, Koellikerstrasse 6, Wuerzburg 97070, Germany
| | - K. Vasilev
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Adelaide, South Australia 5095, Australia
- School of Engineering, University of South Australia, Mawson Lakes Campus, Adelaide, South Australia 5095, Australia
| |
Collapse
|