101
|
Rahmati M, Mozafari M. Selective Contribution of Bioactive Glasses to Molecular and Cellular Pathways. ACS Biomater Sci Eng 2019; 6:4-20. [PMID: 33463236 DOI: 10.1021/acsbiomaterials.8b01078] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Over the past few decades, biomedical scientists and surgeons have given substantial attention to bioactive glasses as promising, long-lasting biomaterials that can make chemical connections with the neighboring hard and soft tissues. Several studies have examined the cellular and molecular responses to bioactive glasses to determine if they are suitable biomaterials for tissue engineering and regenerative medicine. In this regard, different ions and additives have been used recently to induce specific characteristics for selective cellular and molecular responses. This Review briefly describes foreign-body response mechanisms and the role of adsorbed proteins as the key players in starting interactions between cells and biomaterials. It then explains the physicochemical properties of the most common bioactive glasses, which have a significant impact on their cellular and molecular responses. It is expected that, with the development of novel strategies, the physiochemical properties of bioactive glasses can be engineered to precisely control proteins' adsorption and cellular functions after implantation.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran 14155-4777, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 144961-4535, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 144961-4535 Tehran, Iran
| |
Collapse
|
102
|
Olthof MGL, Tryfonidou MA, Dadsetan M, Dhert WJA, Yaszemski MJ, Kempen DHR, Lu L. In Vitro and In Vivo Correlation of Bone Morphogenetic Protein-2 Release Profiles from Complex Delivery Vehicles. Tissue Eng Part C Methods 2019; 24:379-390. [PMID: 29756545 DOI: 10.1089/ten.tec.2018.0024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Local sustained delivery of bioactive molecules from biomaterials is a promising strategy to enhance bone regeneration. To optimize delivery vehicles for bone formation, the design characteristics are tailored with consequential effect on bone morphogenetic protein-2 (BMP-2) release and bone regeneration. Complying with the 3R principles (Replacement, Reduction, and Refinement), the growth factor release is often investigated in vitro using several buffers to mimic the in vivo physiological environment. However, this remains an unmet need. Therefore, this study investigates the in vitro-in vivo correlation (IVIVC) of BMP-2 release from complex delivery vehicles in several commonly used in vitro buffers: cell culture model, phosphate buffered saline, and a strong desorption buffer. The results from this study showed that the release environment affected the BMP-2 release profiles, creating distinct relationships between release versus time and differences in extent of release. According to the guidance set by the U.S. Food and Drug Administration (FDA), IVIVC resulted in level A internal predictability for individual composites. Since the IVIVC was influenced by the BMP-2 loading method and composite surface chemistry, the external predictive value of the IVIVCs was limited. These results show that the IVIVCs can be used for predicting the release of an individual composite. However, the models cannot be used for predicting in vivo release for different composite formulations since they lack external predictability. Potential confounding effects of drug type, delivery vehicle formulations, and application site should be added to the equation to develop one single IVIVC applicable for complex delivery vehicles. Altogether, these results imply that more sophisticated in vitro systems should be used in bone regeneration to accurately discriminate and predict in vivo BMP-2 release from different complex delivery vehicles.
Collapse
Affiliation(s)
- Maurits G L Olthof
- 1 Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,2 Department of Orthopedic Surgery, Mayo Clinic College of Medicine , Rochester, Minnesota.,3 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands .,4 Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University , Utrecht, The Netherlands
| | - Marianna A Tryfonidou
- 4 Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University , Utrecht, The Netherlands
| | - Mahrokh Dadsetan
- 1 Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,2 Department of Orthopedic Surgery, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Wouter J A Dhert
- 4 Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University , Utrecht, The Netherlands
| | - Michael J Yaszemski
- 1 Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,2 Department of Orthopedic Surgery, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Diederik H R Kempen
- 5 Department of Orthopaedic Surgery, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Lichun Lu
- 1 Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,2 Department of Orthopedic Surgery, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
103
|
Zhang W, Lu X, Yuan Z, Shen M, Song Y, Liu H, Deng J, Zhong X, Zhang X. Establishing an osteoimmunomodulatory coating loaded with aspirin on the surface of titanium primed with phase-transited lysozyme. Int J Nanomedicine 2019; 14:977-991. [PMID: 30787611 PMCID: PMC6368129 DOI: 10.2147/ijn.s190766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND To improve osseointegration and enhance the success rate of implanted biomaterials, the surface modification technology of bone implants has developed rapidly. Intensive research on osteoimmunomodulation has shown that the surfaces of implants should possess favorable osteoimmunomodulation to facilitate osteogenesis. METHODS A novel, green and efficient phase-transited lysozyme (PTL) technique was used to prime titanium discs with a positive charge. In addition, sodium hyaluronate (HA) and self-assembled type I collagen containing aspirin (ASA) nanoparticles were decorated on PTL-primed Ti discs via electrostatic interaction. RESULTS The behaviors of bone marrow stromal cells (BMSCs) on the Ti disc surfaces containing ASA were analyzed in different conditioned media (CM) generated by macrophages. Additionally, the secretion of inflammation-related cytokines of macrophages on the surfaces of different Ti discs was investigated in in vitro experiments, which showed that the Ti surface containing ASA not only supported the migration, proliferation and differentiation of BMSCs but also reduced the inflammatory response of macrophages compared with Ti discs without surface modification. After implantation in vivo, the ASA-modified implant can significantly contribute to bone formation around the implant, which mirrors the evaluation in vitro. CONCLUSION This study highlights the significant effects of appropriate surface characteristics on the regulation of osteogenesis and osteoimmunomodulation around an implant. Implant modification with ASA potentially provides superior strategies for the surface modification of biomaterials.
Collapse
Affiliation(s)
- Wenxin Zhang
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Xin Lu
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Zuoying Yuan
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, People's Republic of China
| | - Minjuan Shen
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Yunjia Song
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Huanhuan Liu
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Jingjing Deng
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Xue Zhong
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| | - Xu Zhang
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People's Republic of China,
| |
Collapse
|
104
|
Sharifi F, Htwe SS, Righi M, Liu H, Pietralunga A, Yesil-Celiktas O, Maharjan S, Cha BH, Shin SR, Dokmeci MR, Vrana NE, Ghaemmaghami AM, Khademhosseini A, Zhang YS. A Foreign Body Response-on-a-Chip Platform. Adv Healthc Mater 2019; 8:e1801425. [PMID: 30694616 PMCID: PMC6398437 DOI: 10.1002/adhm.201801425] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/17/2018] [Indexed: 11/10/2022]
Abstract
Understanding the foreign body response (FBR) and desiging strategies to modulate such a response represent a grand challenge for implant devices and biomaterials. Here, the development of a microfluidic platform is reported, i.e., the FBR-on-a-chip (FBROC) for modeling the cascade of events during immune cell response to implants. The platform models the native implant microenvironment where the implants are interfaced directly with surrounding tissues, as well as vasculature with circulating immune cells. The study demonstrates that the release of cytokines such as monocyte chemoattractant protein 1 (MCP-1) from the extracellular matrix (ECM)-like hydrogels in the bottom tissue chamber induces trans-endothelial migration of circulating monocytes in the vascular channel toward the hydrogels, thus mimicking implant-induced inflammation. Data using patient-derived peripheral blood mononuclear cells further reveal inter-patient differences in FBR, highlighting the potential of this platform for monitoring FBR in a personalized manner. The prototype FBROC platform provides an enabling strategy to interrogate FBR on various implants, including biomaterials and engineered tissue constructs, in a physiologically relevant and individual-specific manner.
Collapse
Affiliation(s)
- Fatemeh Sharifi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- School of Mechanical Engineering, Sharif University of Technology, Tehran, 11155-8639, Iran
| | - Su Su Htwe
- Immunology and Immuno-bioengineering Group, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Martina Righi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Hua Liu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Anna Pietralunga
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ozlem Yesil-Celiktas
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Bioengineering, Faculty of Engineering Ege University, Bornova, 35100, Izmir, Turkey
| | - Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Byung-Hyun Cha
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Mehmet Remzi Dokmeci
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Piaggio, 56025, Italy
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioindustrial Technologies, Konkuk University, Seoul, 05029, Republic of Korea
| | - Nihal Engin Vrana
- Biomatériaux et Bioingénierie, Institut National de la Santé et de la Recherche Médicale (INSERM), 67085, Strasbourg, France
- Protip Medical, 67000, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg, Fédération des Matériaux et Nanoscience d'Alsace (FMNA), Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Amir M Ghaemmaghami
- Immunology and Immuno-bioengineering Group, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ali Khademhosseini
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Piaggio, 56025, Italy
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310012, P. R. China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310012, P. R. China
- Research Institute for Bioscience and Biotechnology, Lalitpur, 44600, Nepal
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| |
Collapse
|
105
|
Rousselle SD, Ramot Y, Nyska A, Jackson ND. Pathology of Bioabsorbable Implants in Preclinical Studies. Toxicol Pathol 2019; 47:358-378. [DOI: 10.1177/0192623318816681] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Bioabsorbable implants can be advantageous for certain surgical tissue bioengineering applications and implant-assisted tissue repair. They offer the obvious benefits of nonpermanence and eventual restoration of the native tissue’s biomechanical and immunological properties, while providing a structural scaffold for healing and a route for additional therapies (i.e., drug elution). They present unique developmental, imaging, and histopathological challenges in the conduct of preclinical animal studies and in interpretation of pathology data. The bioabsorption process is typically associated with a gradual decline (over months to years) in structural strength and integrity and may also be associated with cellular responses such as phagocytosis that may confound interpretation of efficacy and safety end points. Additionally, as these implants bioabsorb, they become increasingly difficult to isolate histologically and thus imaging modalities such as microCT become very valuable to determine the original location of the implants and to assess the remodeling response in tandem with histopathology. In this article, we will review different types of bioabsorbable implants and commonly used bioabsorbable materials; additionally, we will address some of the most common challenges and pitfalls confronting histologists and pathologists in collecting, handling, imaging, preparing tissues through histology, evaluating, and interpreting study data associated with bioabsorbable implants.
Collapse
Affiliation(s)
| | - Yuval Ramot
- Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | | | | |
Collapse
|
106
|
The inflammasome in host response to biomaterials: Bridging inflammation and tissue regeneration. Acta Biomater 2019; 83:1-12. [PMID: 30273748 DOI: 10.1016/j.actbio.2018.09.056] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/05/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
Abstract
The development of new biomaterials to be used in tissue engineering applications is creating new solutions for a range of healthcare problems. The trend in biomaterials research has shifted from biocompatible "immune-evasive" biomaterials to "immune-interactive" materials that modulate the inflammatory response supporting implant integration as well as improving healing and tissue regeneration. Inflammasomes are large intracellular multiprotein complexes that are key players in host defence during innate immune responses and assemble after recognition of pathogens or danger signals. The process of biomaterial implantation causes injury to tissues that will consequently release danger signals that could be sensed by the inflammasome. There are increasing evidences that the inflammasome has a role in several inflammatory processes, from pathogen clearance to chronic inflammation or tissue repair. Thus, modulation of the inflammasome activity appears as an important target in the development of effective approaches in regenerative medicine. In this review, we discuss the main points of the current understanding on the host response to implanted biomaterials and how the paradigm of "immune-evasive" biomaterials has shifted over the last years; the significance of the inflammasome in the inflammatory response to biomaterials; and the growing idea that the immune system is of key importance in an effective tissue repair and regeneration. STATEMENT OF SIGNIFICANCE: We herein discuss the main points of the current understanding on the host response to implanted biomaterials and how the paradigm of "immune-evasive" biomaterials has shifted to "immune-interactive" over the last years; the significance of the inflammasome in the inflammatory response to biomaterials; and the growing idea that the immune system is of key importance in an effective tissue repair and regeneration, supporting the emerging concept of Regenerative Immunology. The inflammasome is a recent and central concept in immunology research. Since the beginning of this century the inflammasome is viewed as key platform of the innate immune response. We believe that, successful modulation of the inflammasome activity will become a milestone in the fields of tissue engineering and regenerative medicine.
Collapse
|
107
|
Diaz-Rodriguez P, Chen H, Erndt-Marino JD, Liu F, Totsingan F, Gross RA, Hahn MS. Impact of Select Sophorolipid Derivatives on Macrophage Polarization and Viability. ACS APPLIED BIO MATERIALS 2018; 2:601-612. [DOI: 10.1021/acsabm.8b00799] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
108
|
Shin BH, Kim BH, Kim S, Lee K, Choy YB, Heo CY. Silicone breast implant modification review: overcoming capsular contracture. Biomater Res 2018; 22:37. [PMID: 30598837 PMCID: PMC6302391 DOI: 10.1186/s40824-018-0147-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/07/2018] [Indexed: 12/25/2022] Open
Abstract
Background Silicone implants are biomaterials that are frequently used in the medical industry due to their physiological inertness and low toxicity. However, capsular contracture remains a concern in long-term transplantation. To date, several studies have been conducted to overcome this problem. This review summarizes and explores these trends. Main body First, we examined the overall foreign body response from initial inflammation to fibrosis capsule formation in detail and introduced various studies to overcome capsular contracture. Secondly, we introduced that the main research approaches are to inhibit fibrosis with anti-inflammatory drugs or antibiotics, to control the topography of the surface of silicone implants, and to administer plasma treatment. Each study examined aspects of the various mechanisms by which capsular contracture could occur, and addressed the effects of inhibiting fibrosis. Conclusion This review introduces various silicone surface modification methods to date and examines their limitations. This review will help identify new directions in inhibiting the fibrosis of silicone implants.
Collapse
Affiliation(s)
- Byung Ho Shin
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Byung Hwi Kim
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Sujin Kim
- 2Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| | - Kangwon Lee
- 2Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea.,7Advanced Institutes of Convergence Technology, Suwon, Gyeonggi-do 16229 South Korea
| | - Young Bin Choy
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea.,3Interdisciplinary Program for Bioengineering, College of Engineering, Seoul National University, Seoul, 08826 Republic of Korea.,6Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080 Republic of Korea
| | - Chan Yeong Heo
- 3Interdisciplinary Program for Bioengineering, College of Engineering, Seoul National University, Seoul, 08826 Republic of Korea.,4Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul, 03080 Republic of Korea.,5Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, 13620 Republic of Korea
| |
Collapse
|
109
|
Lin W, Guo H, Wang L, Zhang D, Wu X, Li L, Li D, Tang R. Nitrite Enhances MC-LR-Induced Changes on Splenic Oxidation Resistance and Innate Immunity in Male Zebrafish. Toxins (Basel) 2018; 10:E512. [PMID: 30513985 PMCID: PMC6315824 DOI: 10.3390/toxins10120512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/24/2018] [Accepted: 11/28/2018] [Indexed: 01/14/2023] Open
Abstract
Hazardous contaminants, such as nitrite and microcystin-leucine arginine (MC-LR), are released into water bodies during cyanobacterial blooms and may adversely influence the normal physiological function of hydrobiontes. The combined effects of nitrite and MC-LR on the antioxidant defense and innate immunity were evaluated through an orthogonal experimental design (nitrite: 0, 29, 290 μM; MC-LR: 0, 3, 30 nM). Remarkable increases in malondialdehyde (MDA) levels have suggested that nitrite and/or MC-LR exposures induce oxidative stress in fish spleen, which were indirectly confirmed by significant downregulations of total antioxidant capacity (T-AOC), glutathione (GSH) contents, as well as transcriptional levels of antioxidant enzyme genes cat1, sod1 and gpx1a. Simultaneously, nitrite and MC-LR significantly decreased serum complement C3 levels as well as the transcriptional levels of splenic c3b, lyz, il1β, ifnγ and tnfα, and indicated that they could jointly impact the innate immunity of fish. The severity and extent of splenic lesions were aggravated by increased concentration of nitrite or MC-LR and became more serious in combined groups. The damages of mitochondria and pseudopodia in splenic macrophages suggest that oxidative stress exerted by nitrite and MC-LR aimed at the membrane structure of immune cells and ultimately disrupted immune function. Our results clearly demonstrate that nitrite and MC-LR exert synergistic suppressive effects on fish innate immunity via interfering antioxidant responses, and their joint toxicity should not be underestimated in eutrophic lakes.
Collapse
Affiliation(s)
- Wang Lin
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Honghui Guo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Lingkai Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Dandan Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xueyang Wu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Li Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China.
- National Demonstration Center for Experimental Aquaculture Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Dapeng Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China.
- National Demonstration Center for Experimental Aquaculture Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Rong Tang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China.
- National Demonstration Center for Experimental Aquaculture Education, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
110
|
Rashad A, Suliman S, Mustafa M, Pedersen TØ, Campodoni E, Sandri M, Syverud K, Mustafa K. Inflammatory responses and tissue reactions to wood-Based nanocellulose scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:208-221. [PMID: 30678905 DOI: 10.1016/j.msec.2018.11.068] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 01/18/2023]
Abstract
Two wood-derived cellulose nanofibril (CNF) porous scaffolds were prepared by TEMPO-oxidation and carboxymethylation. The effects of these scaffolds on the production of inflammatory cytokines by human macrophage-like cells (U937) was profiled in vitro after 1 and 3 days and in subcutaneous tissues of rats after 4 and 30 days, using PCR and Multiplex arrays. Tissue culture plates (TCP) and gelatin scaffolds served as controls in vitro and in vivo respectively. After 3 days in vitro, there was no significant difference between the effects of CNF scaffolds and TCP on the production of chemokines/growth factors and pro-inflammatory cytokines. At day 4 in vivo there was significantly higher gene expression of the anti-inflammatory IL-1Ra in the CNF scaffolds than the gelatin scaffold. Production of IL-1β, IL-6, MCP-1, MIP-1α CXCL-1 and M-CSF was significantly less than in the gelatin, demonstrating an early mild inflammatory response. At day 30, both CNF scaffolds significantly stimulated the production of the anti-inflammatory cytokine IL-10. Unlike gelatin, neither CNF scaffold had degraded 180 days post-implantation. The slow degradation of CNF scaffolds resulted in a foreign body reaction, with high production of IL-1β, IL-2, TNF-α, IFN-ϒ, MCP-1, MIP-1α, M-CSF, VEGF cytokines and expression of MMP-9 gene. The surface chemistry of the CNF scaffolds elicited a modest effect on cytokine production and did not shift the inflammatory profile in vitro or in vivo. The decisive role in development of the foreign body reaction was the slow degradation of the CNF scaffolds.
Collapse
Affiliation(s)
- Ahmad Rashad
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway.
| | - Salwa Suliman
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Manal Mustafa
- Oral Health Centre of Expertise in Western Norway, Bergen, Norway
| | | | - Elisabetta Campodoni
- Institute of Science and Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Monica Sandri
- Institute of Science and Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Kristin Syverud
- RISE PFI, Trondheim, Norway; Department of Chemical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway.
| |
Collapse
|
111
|
Radhakrishnan A, Sreekumaran S, Nair JA, Kurup MG. Immunocompatibility of poly(ethylene glycol)-penetrated alginate–chitosan hydrogels. BIOINSPIRED BIOMIMETIC AND NANOBIOMATERIALS 2018. [DOI: 10.1680/jbibn.18.00023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Scaffold-guided tissue regeneration using hydrogels has been emerging as an ideal alternative for the management of terminal-stage organ damage. Design of scaffolds for tissue regrowth mainly focuses on their immuno/biocompatibility as well as physiochemical characteristics. The focus of this study is on the biocompatibility evaluations of a panel of four hydrogel scaffolds fabricated using alginate, chitosan and hydroxyapatite reinforced with poly(ethylene glycol). The hydrogels were subsequently cross-linked with calcium ions and glutaraldehyde. Immunocompatibility was assessed by interacting Raw 264.7 cell lines with these hydrogels. MTT cell viability assay revealed the non-cytotoxic nature of the hydrogels, and the macrophages grown in contact with the hydrogels exhibited no alteration in their morphology and were similar to the untreated normal cells. The concentration of nitric oxide, activity of myeloperoxidase and the messenger ribonucleic acid transcripts of proinflammatory cytokines, interleukin 6 and tumour necrosis factor alpha exhibited no considerable increase in the macrophages cultured with the hydrogels when compared to lipopolysaccharide-stimulated cells. In short, the absence of macrophage activation on contact with hydrogels is a clear indication of their in vitro immunocompatibility, suggesting their potential application as tissue engineering templates.
Collapse
Affiliation(s)
- Anitha Radhakrishnan
- Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
| | | | - Jayakumaran A Nair
- Inter University Centre for Genomics and Gene Technology, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Muraleedhara G Kurup
- Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
| |
Collapse
|
112
|
Li X, Gao P, Tan J, Xiong K, Maitz MF, Pan C, Wu H, Chen Y, Yang Z, Huang N. Assembly of Metal-Phenolic/Catecholamine Networks for Synergistically Anti-Inflammatory, Antimicrobial, and Anticoagulant Coatings. ACS APPLIED MATERIALS & INTERFACES 2018; 10:40844-40853. [PMID: 30403339 DOI: 10.1021/acsami.8b14409] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The development of a facile and versatile strategy to endow surfaces with synergistically anti-inflammatory, antimicrobial, and anticoagulant functions is of particular significance for blood-contacting biomaterials and medical devices. In this work, we report a simple and environmentally friendly "one-pot" method inspired by byssal cuticle chemistry, namely, [Fe(dopa)3] coordination chemistry for assembly of copper ions (Cu2+) and plant polyphenol (tannic acid)/catecholamine (dopamine or norepinephrine) to form metal-phenolic/catecholamine network-based coatings. This one-pot method enabled us to easily develop a multifunctional surface based on the combination of the characteristic functions of metal ions and plant polyphenol or catecholamine. The residual phenolic hydroxyl groups on the coatings imparted the modified surface with excellent antioxidant and anti-inflammatory functions. The robust chelation of copper ions to the metal-phenolic/catecholamine networks provided not only durable antibacterial property but also glutathione peroxidase like catalytic capability to continuously and controllably produce antithrombotic nitric oxide by catalyzing endogenous S-nitrothiol. The biological functions of such coatings could be well regulated by adjusting the ratios of the feed concentration of Cu2+ ions to plant polyphenol or catecholamine. We envision that our simple, multifunctional, and bioinspired coating strategy can hold great application promise for bioengineering blood-contacting devices.
Collapse
Affiliation(s)
- Xiangyang Li
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| | - Peng Gao
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| | - Jianying Tan
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| | - Kaiqin Xiong
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials , Leibniz Institute of Polymer Research Dresden , Hohe Strasse 6 , Dresden 01069 , Germany
| | - Changjiang Pan
- Faculty of Mechanical and Materials Engineering , Huaiyin Institute of Technology , Huai'an 223003 , China
| | - Hongkai Wu
- Division of Biomedical Engineering , The Hong Kong University of Science and Technology , Hong Kong , China
| | - Yin Chen
- Division of Biomedical Engineering , The Hong Kong University of Science and Technology , Hong Kong , China
| | - Zhilu Yang
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| | - Nan Huang
- Key Laboratory of Advanced Technology for Materials of Education Ministry, School of Materials Science and Engineering , Southwest Jiaotong University , Chengdu 610031 , China
| |
Collapse
|
113
|
Yuan B, Raucci MG, Fan Y, Zhu X, Yang X, Zhang X, Santin M, Ambrosio L. Injectable strontium-doped hydroxyapatite integrated with phosphoserine-tethered poly(epsilon-lysine) dendrons for osteoporotic bone defect repair. J Mater Chem B 2018; 6:7974-7984. [PMID: 32255042 DOI: 10.1039/c8tb02526f] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The control of the inflammatory response induced by the implantation of foreign biomaterials is fundamental in determining tissue healing. It has been shown that the activation of specific macrophage pathways upon contact with a biomaterial can lead either to a chronic inflammation preventing a physiological tissue repair or to an improved tissue healing. In the case of bone repair, calcium phosphate cements with good osteoconductivity properties have been successfully applied in bone defect filling and repair, but poor handling properties, insufficient viscous flow and unmatched degradation rate are still problems that remain unsolved. In this study, a strontium-doped hydroxyapatite (HA) gel was modified by integrating branched poly(epsilon-lysine) dendrons with third-generation branches exposing phosphoserine (SrHA/G3-K PS). The interaction of this material with macrophages was investigated in vitro, focusing on the secretion and gene expression of specific pro-inflammatory cytokines. Our results showed that the addition of strontium and G3-K PS to HA sol-gel could down-regulate the gene expression of inflammatory factors such as IL-1β, TNF-α and MCP-1, while increasing the gene expression of IL-6, a cytokine known for its osteogenic effect. These results were further confirmed by ELISA test of the respective protein concentrations. When exposed to supernatants of macrophage culture in the presence of strontium and G3-K PS, osteoblast viability was promoted with elevated osteogenic gene markers, in terms of OPG, ALP, OCN and COL-I. In vivo implantation experiments using an osteoporotic rat model with bone defect further confirmed that the addition of G3-K PS to HA could dramatically promote new bone regeneration. Although the introduction of strontium improved the degradation properties of the injectable materials, no positive effect on promoting in vivo bone regeneration was observed.
Collapse
Affiliation(s)
- Bo Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | | | | | | | | | | | | | | |
Collapse
|
114
|
Mödinger Y, Teixeira GQ, Neidlinger-Wilke C, Ignatius A. Role of the Complement System in the Response to Orthopedic Biomaterials. Int J Mol Sci 2018; 19:ijms19113367. [PMID: 30373272 PMCID: PMC6274916 DOI: 10.3390/ijms19113367] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022] Open
Abstract
Various synthetic biomaterials are used to replace lost or damaged bone tissue that, more or less successfully, osseointegrate into the bone environment. Almost all biomaterials used in orthopedic medicine activate the host-immune system to a certain degree. The complement system, which is a crucial arm of innate immunity, is rapidly activated by an implanted foreign material into the human body, and it is intensely studied regarding blood-contacting medical devices. In contrast, much less is known regarding the role of the complement system in response to implanted bone biomaterials. However, given the increasing knowledge of the complement regulation of bone homeostasis, regeneration, and inflammation, complement involvement in the immune response following biomaterial implantation into bone appears very likely. Moreover, bone cells can produce complement factors and are target cells of activated complement. Therefore, new bone formation or bone resorption around the implant area might be greatly influenced by the complement system. This review aims to summarize the current knowledge on biomaterial-mediated complement activation, with a focus on materials primarily used in orthopedic medicine. In addition, methods to modify the interactions between the complement system and bone biomaterials are discussed, which might favor osseointegration and improve the functionality of the device.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Graciosa Q Teixeira
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Cornelia Neidlinger-Wilke
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| |
Collapse
|
115
|
Axl Involved in Mineral Trioxide Aggregate Induces Macrophage Polarization. J Endod 2018; 44:1542-1548. [PMID: 30170844 DOI: 10.1016/j.joen.2018.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/29/2018] [Accepted: 07/07/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In this study, we examined the effect of mineral trioxide aggregate (MTA) on macrophage polarization and the potential involvement of Axl/nuclear factor kappa B (NF-κB) signaling in mediating the effect of MTA. METHODS The human monocyte cell line THP-1 was cultured with MTA solution for 1, 2, or 3 days, and the population change of M2 macrophages was analyzed by flow cytometry. Expression of M2 cytokines was examined by enzyme-linked immunosorbent assay. Phagocytosis and angiogenesis-induction ability were also assayed. The involvement of Axl/NF-κB signaling in MTA-treated cells was examined by analyzing phosphorylation status of Axl, Akt, IKKα/β, and IκBα. Specific inhibitors for Axl/Akt/NF-κB signaling were added to MTA-treated THP-1 cells, and their cytokine expression change was examined. RESULTS Flow cytometry analysis showed that MTA treatment increased CD206+ cells in a time-dependent way. After MTA treatment, the expression of M2-related cytokines was up-regulated. MTA also enhanced phagocytic ability and the ability of THP-1 cells to induce angiogenesis. Treatment of MTA led to activate Axl/Akt/NF-kB signal axis by phosphorylation of Axl, Akt, IKKα/β, IκBα, and p65. In addition, MTA-induced interleukin 10, transforming growth factor beta, and vascular endothelial growth factor expression was suppressed as specific inhibitors were added. CONCLUSIONS Our findings indicate that MTA is able to induce macrophage polarization toward the M2 phenotype, with up-regulation of interleukin 10, transforming growth factor beta, and vascular endothelial growth factor, and that Axl/Akt/NF-κB signaling participates in this process. These results provide the cellular and molecular basis of MTA's anti-inflammatory action in clinical applications.
Collapse
|
116
|
Unveiling the fate of adhering bacteria to antimicrobial surfaces: expression of resistance-associated genes and macrophage-mediated phagocytosis. Acta Biomater 2018; 78:189-197. [PMID: 30071350 DOI: 10.1016/j.actbio.2018.07.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/16/2018] [Accepted: 07/29/2018] [Indexed: 12/31/2022]
Abstract
Since most antibacterial coatings reported to fight biomaterial-associated infections (BAI) fail in completely preventing bacterial colonization, it is crucial to know the impact of that small fraction of adhered bacteria in BAI recrudescence. This study aims to understand the fate of Staphylococcus aureus able to adhere to an antimicrobial coating previously developed, in terms of potential development of bacterial resistance and their macrophage-mediated phagocytosis. Antimicrobial coating comprised the co-immobilization of Palm peptide and DNase I onto polydimethylsiloxane. Expression of genes associated to resistance and virulence mechanisms showed that cells in contact with antimicrobial surfaces for a long period of 30 days, exhibit genes equally or less expressed, as compared to cells recovered from control surfaces. Recovered cells also exhibit the same susceptibility patterns, which strengthens the evidence of no resistance development. Remarkably, cells adhered to modified surfaces shows a reduced metabolic activity upon vancomycin treatment unlike the cells found on control surfaces, which can be identified as a clinical opportunity for prophylactically administration after implant surgery. Furthermore, results highlight that functionalization of PDMS with Palm and DNase I should not compromise the action of host immune cells. The overall results reinforce the potential of this antimicrobial strategy to fight BAI.
Collapse
|
117
|
Al-Maawi S, Vorakulpipat C, Orlowska A, Zrnc TA, Sader RA, Kirkpatrick CJ, Ghanaati S. In vivo Implantation of a Bovine-Derived Collagen Membrane Leads to Changes in the Physiological Cellular Pattern of Wound Healing by the Induction of Multinucleated Giant Cells: An Adverse Reaction? Front Bioeng Biotechnol 2018; 6:104. [PMID: 30155464 PMCID: PMC6102314 DOI: 10.3389/fbioe.2018.00104] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/05/2018] [Indexed: 11/13/2022] Open
Abstract
The present study evaluated the tissue response toward a resorbable collagen membrane derived from bovine achilles tendon (test group) in comparison to physiological wound healing (control group). After subcutaneous implantation in Wistar rats over 30 days, histochemical and immunohistochemical methods elucidated the cellular inflammatory response, vascularization pattern, membrane protein and cell absorbance capacity. After 30 days, the test-group induced two different inflammatory patterns. On the membrane surface, multinucleated giant cells (MNGCs) were formed after the accumulation of CD-68-positive cells (macrophages), whereas only mononuclear cells (MNCs) were found within the membrane central region. Peri-implant vascularization was significantly enhanced after the formation of MNGCs. No vessels were found within the central region of the membrane. Physiological wound healing revealed no MNGCs at any time point. These dynamic changes in the cellular reaction and vascularization within the test-group are related typical indications of a foreign body reaction. Due to the membrane-specific porosity, mononuclear cells migrated into the central region, and the membrane maintained its integrity over 30 days by showing no breakdown or disintegration. The ex vivo investigation analyzed the interaction between the membrane and a blood concentrate system, liquid platelet-rich fibrin (liquid PRF), derived from human peripheral blood and consisting of platelets, leukocytes and fibrin. PRF penetrated the membrane after just 15 min. The data question the role of biomaterial-induced MNGCs as a pathological reaction and whether this is acceptable to trigger vascularization or should be considered as an adverse reaction. Therefore, further pre-clinical and clinical studies are needed to identify the types of MNGCs that are induced by clinically approved biomaterials.
Collapse
Affiliation(s)
- Sarah Al-Maawi
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| | - Chakorn Vorakulpipat
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| | - Anna Orlowska
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| | - Tomislav A Zrnc
- Department of Oral and Maxillofacial Surgery, Medical University of Graz, Graz, Austria
| | - Robert A Sader
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| | - C James Kirkpatrick
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| | - Shahram Ghanaati
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Frankfurt Orofacial Regenerative Medicine Lab, University Hospital Frankfurt Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
118
|
Zhou G, Groth T. Host Responses to Biomaterials and Anti-Inflammatory Design-a Brief Review. Macromol Biosci 2018; 18:e1800112. [DOI: 10.1002/mabi.201800112] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/08/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Guoying Zhou
- Biomedical Materials Group; Institute of Pharmacy; Martin Luther University Halle-Wittenberg; 06099 Halle (Saale) Germany
| | - Thomas Groth
- Biomedical Materials Group; Institute of Pharmacy and, Interdisciplinary Center of Material Science and Interdisciplinary Center for Transfer-Oriented Research in Natural Sciences; Martin Luther University Halle-Wittenberg; 06099 Halle (Saale) Germany
| |
Collapse
|
119
|
Liu R, Lin Y, Lin J, Zhang L, Mao X, Huang B, Xiao Y, Chen Z, Chen Z. Blood Prefabrication Subcutaneous Small Animal Model for the Evaluation of Bone Substitute Materials. ACS Biomater Sci Eng 2018; 4:2516-2527. [PMID: 33435115 DOI: 10.1021/acsbiomaterials.8b00323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Runheng Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yixiong Lin
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Jinying Lin
- Xiamen Stomatological Hospital, Xiamen 361000, China
| | - Linjun Zhang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xueli Mao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Baoxin Huang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yin Xiao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Institute of Health and Biomedical Innovation and the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia
| | - Zhuofan Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zetao Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
120
|
Huang Q, Li X, Elkhooly TA, Xu S, Liu X, Feng Q, Wu H, Liu Y. The osteogenic, inflammatory and osteo-immunomodulatory performances of biomedical Ti-Ta metal-metal composite with Ca- and Si-containing bioceramic coatings. Colloids Surf B Biointerfaces 2018; 169:49-59. [PMID: 29747030 DOI: 10.1016/j.colsurfb.2018.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 02/01/2023]
Abstract
It is known that good mechanical properties, low modulus to reduce stress-shielding effect, favorable osteogenic activity and limited inflammatory response are critical factors for orthopedic implants to induce excellent osteointegration. In this study, Ti-20% Ta metal-metal composite (referred as Ti-Ta) which consisted of Ti- and Ta-rich phases was fabricated via the strategy of powder metallurgy. Micro-arc oxidation (MAO) was employed to modify the surface of Ti-Ta composite. The surfaces of Ti-Ta composite after MAO treatment at an applied voltage of 250 (referred as MAO-250 V) or 300 V (referred as MAO-300 V) exhibited three distinct zones with significantly different morphological features and surface chemistry. Osteoblast-like SaOS-2 cells were found to be preferential to attach on the Ta-rich phase and its surrounding areas, exhibiting an area-dependent adhesion tendency. However, the attachment of Raw 264.7 macrophages was found to be insensitive to the surface characteristics. The proliferation and differentiation of SaOS-2 cells cultured on various surfaces basically followed the trend: MAO-modified surfaces > Ti-Ta surface > Ti surface. The Ti-Ta and MAO-modified surfaces were found to inhibit the inflammatory response and polarize macrophages to anti-inflammatory M2 phenotype compared to Ti surface. Moreover, the microenvironments created by Ti-Ta, MAO-250 V and MAO-300 V/macrophage interactions promoted the proliferation and differentiation of SaOS-2 cells compared to that created by Ti/macrophage interactions. MAO-300 V surface exhibited further enhanced positive osteo-immunomodulatory effects compared to Ti-Ta surface. Together, the Ti-20% Ta metal-metal composite modified by MAO at an applied voltage of 300 V is considered as a promising implant material for orthopedic applications.
Collapse
Affiliation(s)
- Qianli Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Xuezhong Li
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Tarek A Elkhooly
- Department of ceramics, Inorganic Chemical Industries Division, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Shenghang Xu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Xujie Liu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, PR China
| | - Qingling Feng
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, PR China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China.
| | - Yong Liu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China.
| |
Collapse
|
121
|
Freitas de Lima F, Lescano CH, Arrigo JDS, Cardoso CAL, Coutinho JP, Moslaves ISB, Ximenes TVDN, Kadri MCT, Weber SS, Perdomo RT, Kassuya CAL, Vieira MDC, Sanjinez-Argandoña EJ. Anti-inflammatory, antiproliferative and cytoprotective potential of the Attalea phalerata Mart. ex Spreng. pulp oil. PLoS One 2018; 13:e0195678. [PMID: 29634766 PMCID: PMC5892912 DOI: 10.1371/journal.pone.0195678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/27/2018] [Indexed: 12/28/2022] Open
Abstract
The anti-inflammatory, antiproliferative and cytoprotective activity of the Attalea phalerata Mart. ex Spreng pulp oil was evaluated by in vitro and in vivo methods. As for the chemical profile, the antioxidant activity was performed by spectrophotometry, and the profile of carotenoids and amino acids by chromatography. Our data demonstrated that A. phalerata oil has high carotenoid content, antioxidant activity and the presence of 5 essential amino acids. In the in vitro models of inflammation, the oil demonstrated the capacity to inhibit COX1 and COX2 enzymes, the production of nitric oxide and also induces macrophages to spreading. In the in vivo models of inflammation, the oil inhibited edema and leukocyte migration in the Wistar rats. In the in vitro model of antiproliferative and cytoprotective activity, the oil was shown inactive against the kidney carcinoma and prostate carcinoma lineage cells and with cytoprotective capacity in murine fibroblast cells, inhibiting the cytotoxic action of doxorubicin. Therefore, it is concluded that A. phalerata pulp oil has anti-inflammatory effects with nutraceutical properties potential due to the rich composition. Moreover, the oil also has cytoprotective activity probably because of its ability to inhibit the action of free radicals.
Collapse
Affiliation(s)
- Fernando Freitas de Lima
- Federal University of Grande Dourados, Faculty of Health Sciences, Dourados, MS, Brazil
- * E-mail:
| | | | | | | | | | - Iluska Senna Bonfá Moslaves
- Federal University of Mato Grosso do Sul, Center of Biological and Health Sciences, Campo Grande, MS, Brazil
| | | | | | - Simone Schneider Weber
- Federal University of Mato Grosso do Sul, Center of Biological and Health Sciences, Campo Grande, MS, Brazil
| | - Renata Trentin Perdomo
- Federal University of Mato Grosso do Sul, Center of Biological and Health Sciences, Campo Grande, MS, Brazil
| | | | - Maria do Carmo Vieira
- Federal University of Grande Dourados, Faculty of Agricultural Sciences, Dourados, MS, Brazil
| | | |
Collapse
|
122
|
Fu F, Zhu X, Qin Z, Wang JJ, Xu C, Wang LN, Tu Y, Zhang S, Li RX, Li XH, Zhao ML. Differential degradation rate and underlying mechanism of a collagen/chitosan complex in subcutis, spinal cord and brain tissues of rat. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:35. [PMID: 29556804 DOI: 10.1007/s10856-018-6033-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 02/03/2018] [Indexed: 06/08/2023]
Abstract
Degradation rate is an important index for evaluating biomaterials. The authors' aim was to determine whether the degradation rate of biomaterials is different in distinct tissues and to clarify the underlying mechanism of degradation. The collagen-chitosan (CG-CS) composite scaffolds were prepared using freeze-drying technology. The porosity, water absorption and swelling ratio of the scaffolds were tested in vitro. The scaffolds were implanted into the subcutis, spinal cord and brain tissues of SD rats, the rate of degradation was assessed by continuous monitoring of weight loss, the pathological changes of target areas were observed by histological staining, and matrix metalloproteinase 9 (MMP-9) and lysozyme were detected at the rapid stage of degradation of the scaffolds. Physical and chemical property testing confirmed that CG-CS composite scaffold components can meet the biological requirements of in vivo transplantation. The in vivo experimental results showed that the scaffolds were completely absorbed in the subcutis at 12 days, the scaffolds in the spinal cord and brain groups exhibited progressive mass loss starting from the 3rd week, and a substantial fraction of the scaffold was degraded at 12 weeks. HE staining found that compared with the spinal cord and brain groups, macrophages and capillaries appeared earlier in the subcutis group, and the number was significantly higher (P < 0.05). Western blot analysis showed that the MMP-9 and lysozyme levels in the subcutis were higher than those in the spinal cord and brain (P < 0.05). The results of in vivo experiments demonstrated that the CG-CS scaffold has good biocompatibility and biodegradability, while the rate of degradation was significantly different between the three tissues at the same time point. Macrophage behavior and vascularization in different parts of the body may result in control over the balance of degradation and reconstruction.
Collapse
Affiliation(s)
- Feng Fu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Xiang Zhu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
- The First Traditional Chinese Medicine Hospital of Luoyang City, Luoyang, 471000, China
| | - Zhe Qin
- Chinese People' s Armed Police Forces General Hospital, Beijing, 100039, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Chao Xu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Li-Na Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China
| | - Rui-Xin Li
- Central Laboratory, Tianjin Stomatological Hospital, Tianjin, 300041, China.
| | - Xiao-Hong Li
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China.
| | - Ming-Liang Zhao
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Logistics University of PAP, Tianjin, 300162, China.
| |
Collapse
|
123
|
Zhang R, Liu X, Xiong Z, Huang Q, Yang X, Yan H, Ma J, Feng Q, Shen Z. The immunomodulatory effects of Zn-incorporated micro/nanostructured coating in inducing osteogenesis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018. [DOI: 10.1080/21691401.2018.1446442] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Ranran Zhang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Xujie Liu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zhiyuan Xiong
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Qianli Huang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Xing Yang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Hao Yan
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Jing Ma
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Qingling Feng
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, China
| | - Zhijian Shen
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
- Department of Materials and Environmental Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| |
Collapse
|
124
|
Construction of a Biocompatible and Antioxidant Multilayer Coating by Layer-by-Layer Assembly of κ-Carrageenan and Quercetin Nanoparticles. FOOD BIOPROCESS TECH 2018. [DOI: 10.1007/s11947-018-2077-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
125
|
Shi M, Wang C, Wang Y, Tang C, Miron RJ, Zhang Y. Deproteinized bovine bone matrix induces osteoblast differentiation via macrophage polarization. J Biomed Mater Res A 2018; 106:1236-1246. [PMID: 29280261 DOI: 10.1002/jbm.a.36321] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
Abstract
Bone grafts are widely used in bone regeneration to increase the speed and quality of new bone formation. While they are routinely characterized based on their biocompatible and bioactive properties, they also exert a profound impact on host immune responses, which in turn can display a significant effect on the healing and repair process. In this study, we investigated the role of macrophage behavior on deproteinized bovine bone matrix (DBBM, BioOss) to investigate their impact on creating either a pro- or anti-inflammatory microenvironment for tissue integration. RT-PCR and immunofluorescence staining results demonstrated the ability for RAW 264.7 cells to polarize toward M2 wound-healing macrophages in response to DBBM and positive control (IL-4). Interestingly, significantly higher expression of interleukin-10 and higher number of multinucleated giant cells (MNGCs) was observed in the DBBM group. Thereafter, conditioned media (CM) from macrophages cultured with DBBM seeded with MC3T3-E1 cells demonstrated a marked increase in osteoblast differentiation. Noteworthy, this effect was reversed by blocking IL10 with addition of IL10 antibody to CM from the DBBM macrophages. Furthermore, the use of dendritic cell specific transmembrane protein (DC-STAMP)-knockout to inhibit MNGC formation in the DBBM group resulted in a significant reduction in osteoblast differentiation, indication a pivotal role for MNGCs in biomaterials-induced osteogenesis. The results from this study indicate convincingly that the immune response of macrophages towards DBBM has a potent effect on osteoblast differentiation. Furthermore, DBBM promoted macrophage fusion and polarization towards an M2 wound-healing phenotype, further created a microenvironment favoring biomaterial-induced osteogenesis. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1236-1246, 2018.
Collapse
Affiliation(s)
- Miusi Shi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China.,Department of Dental Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China
| | - Can Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China
| | - Yulan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China
| | - Cuizhu Tang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China.,Department of Dental Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, People's Republic of China
| |
Collapse
|
126
|
Caires HR, Barros da Silva P, Barbosa MA, Almeida CR. A co-culture system with three different primary human cell populations reveals that biomaterials and MSC modulate macrophage-driven fibroblast recruitment. J Tissue Eng Regen Med 2017; 12:e1433-e1440. [PMID: 28865088 DOI: 10.1002/term.2560] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/28/2017] [Accepted: 08/25/2017] [Indexed: 01/24/2023]
Abstract
The biological response to implanted biomaterials is a complex and highly coordinated phenomenon involving many different cell types that interact within 3D microenvironments. Here, we increased the complexity of a 3D platform to include at least 3 cell types that play a role in the host response upon scaffold implantation. With this system, it was possible to address how immune responses triggered by 3D biomaterials mediate recruitment of stromal cells that promote tissue regeneration, mesenchymal stromal/stem cells (MSC), or a foreign body response, fibroblasts. Primary human macrophages yielded the highest fibroblast recruitment when interacting with chitosan scaffolds but not polylactic acid. Interestingly, when there were MSC and fibroblasts in the same environment, macrophages in chitosan scaffolds again promoted a significant increase on fibroblast recruitment, but not of MSC. However, macrophages that were firstly allowed to interact with MSC within the scaffolds were no longer able to recruit fibroblasts. This study illustrates the potential to use different scaffolds to regulate the dynamics of recruitment of proregenerative or fibrotic cell types through immunomodulation. Overall, this work strengths the idea that ex vivo predictive systems need to consider the different players involved in the biological response to biomaterials and that timing of arrival of specific cell types will affect the outcome.
Collapse
Affiliation(s)
- Hugo R Caires
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Patrícia Barros da Silva
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Porto, Portugal
| | - Mário A Barbosa
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina R Almeida
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Porto, Portugal.,Department of Medical Sciences and Institute for Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
127
|
Rennert K, Nitschke M, Wallert M, Keune N, Raasch M, Lorkowski S, Mosig AS. Thermo-responsive cell culture carrier: Effects on macrophage functionality and detachment efficiency. J Tissue Eng 2017; 8:2041731417726428. [PMID: 28890781 PMCID: PMC5574476 DOI: 10.1177/2041731417726428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
Harvesting cultivated macrophages for tissue engineering purposes by enzymatic digestion of cell adhesion molecules can potentially result in unintended activation, altered function, or behavior of these cells. Thermo-responsive polymer is a promising tool that allows for gentle macrophage detachment without artificial activation prior to subculture within engineered tissue constructs. We therefore characterized different species of thermo-responsive polymers for their suitability as cell substrate and to mediate gentle macrophage detachment by temperature shift. Primary human monocyte- and THP-1-derived macrophages were cultured on thermo-responsive polymers and characterized for phagocytosis and cytokine secretion in response to lipopolysaccharide stimulation. We found that both cell types differentially respond in dependence of culture and stimulation on thermo-responsive polymers. In contrast to THP-1 macrophages, primary monocyte-derived macrophages showed no signs of impaired viability, artificial activation, or altered functionality due to culture on thermo-responsive polymers compared to conventional cell culture. Our study demonstrates that along with commercially available UpCell carriers, two other thermo-responsive polymers based on poly(vinyl methyl ether) blends are attractive candidates for differentiation and gentle detachment of primary monocyte-derived macrophages. In summary, we observed similar functionality and viability of primary monocyte-derived macrophages cultured on thermo-responsive polymers compared to standard cell culture surfaces. While this first generation of custom-made thermo-responsive polymers does not yet outperform standard culture approaches, our results are very promising and provide the basis for exploiting the unique advantages offered by custom-made thermo-responsive polymers to further improve macrophage culture and recovery in the future, including the covalent binding of signaling molecules and the reduction of centrifugation and washing steps. Optimizing these and other benefits of thermo-responsive polymers could greatly improve the culture of macrophages for tissue engineering applications.
Collapse
Affiliation(s)
- Knut Rennert
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Germany
| | - Mirko Nitschke
- Leibniz Institute of Polymer Research Dresden, Institute of Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials Dresden, Dresden, Germany
| | - Maria Wallert
- Department of Nutritional Biochemistry and Physiology, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany
| | - Natalie Keune
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Martin Raasch
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Stefan Lorkowski
- Department of Nutritional Biochemistry and Physiology, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany.,Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Alexander S Mosig
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Germany
| |
Collapse
|
128
|
The role of insulin growth factor-1 on the vascular regenerative effect of MAA coated disks and macrophage-endothelial cell crosstalk. Biomaterials 2017; 144:199-210. [PMID: 28841464 DOI: 10.1016/j.biomaterials.2017.08.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/04/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022]
Abstract
The IGF-1 signaling pathway and IGF-1-dependent macrophage/endothelial cell crosstalk was found to be critical features of the vascular regenerative effect displayed by implanted methacrylic acid -co-isodecyl acrylate (MAA-co-IDA; 40% MAA) coated disks in CD1 mice. Inhibition of IGF-1 signaling using AG1024 an IGF1-R tyrosine kinase inhibitor abrogated vessel formation 14 days after disk implantation in a subcutaneous pocket. Explanted tissue had increased arginase 1 expression and reduced iNOS expression consistent with the greater shift from "M1" ("pro-inflammatory") macrophages to "M2" ("pro-angiogenic") macrophages for MAA coated disks relative to control MM (methyl methacrylate-co-IDA) disks; the latter did not generate a vascular response and the polarization shift was muted with AG1024. In vitro, medium conditioned by macrophages (both human dTHP1 cells and mouse bone marrow derived macrophages) had elevated IGF-1 mRNA and protein levels, while the cells had reduced IGF1-R but elevated IGFBP-3 mRNA levels. These cells also had reduced iNOS and elevated Arg1 expression, consistent with the in vivo polarization results, including the inhibitory effects of AG1024. On the other hand, HUVEC exposed to dTHP1 conditioned medium migrated and proliferated faster suggesting that the primary target of the macrophage released IGF-1 was endothelial cells. Although further investigation is warranted, IGF-1 appears to be a key feature underpinning the observed vascularization. Why MAA based materials have this effect remains to be defined, however.
Collapse
|
129
|
Luo T, David MA, Dunshee LC, Scott RA, Urello MA, Price C, Kiick KL. Thermoresponsive Elastin-b-Collagen-Like Peptide Bioconjugate Nanovesicles for Targeted Drug Delivery to Collagen-Containing Matrices. Biomacromolecules 2017; 18:2539-2551. [PMID: 28719196 PMCID: PMC5815509 DOI: 10.1021/acs.biomac.7b00686] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the past few decades, (poly)peptide block copolymers have been widely employed in generating well-defined nanostructures as vehicles for targeted drug delivery applications. We previously reported the assembly of thermoresponsive nanoscale vesicles from an elastin-b-collagen-like peptide (ELP-CLP). The vesicles were observed to dissociate at elevated temperatures, despite the LCST-like behavior of the tethered ELP domain, which is suggested to be triggered by the unfolding of the CLP domain. Here, the potential of using the vesicles as drug delivery vehicles for targeting collagen-containing matrices is evaluated. The sustained release of an encapsulated model drug was achieved over a period of 3 weeks, following which complete release could be triggered via heating. The ELP-CLP vesicles show strong retention on a collagen substrate, presumably through collagen triple helix interactions. Cell viability and proliferation studies using fibroblasts and chondrocytes suggest that the vesicles are highly cytocompatible. Additionally, essentially no activation of a macrophage-like cell line is observed, suggesting that the vesicles do not initiate an inflammatory response. Endowed with thermally controlled delivery, the ability to bind collagen, and excellent cytocompatibility, these ELP-CLP nanovesicles are suggested to have significant potential in the controlled delivery of drugs to collagen-containing matrices and tissues.
Collapse
Affiliation(s)
- Tianzhi Luo
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Michael A. David
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Lucas C. Dunshee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Rebecca A. Scott
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, Newark, DE, 19711, USA
| | - Morgan A. Urello
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Kristi L. Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, Newark, DE, 19711, USA
| |
Collapse
|
130
|
A Role for CD154, the CD40 Ligand, in Granulomatous Inflammation. Mediators Inflamm 2017; 2017:2982879. [PMID: 28785137 PMCID: PMC5529663 DOI: 10.1155/2017/2982879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 06/10/2017] [Accepted: 06/15/2017] [Indexed: 01/08/2023] Open
Abstract
Granulomatous inflammation is a distinctive form of chronic inflammation in which predominant cells include macrophages, epithelioid cells, and multinucleated giant cells. Mechanisms regulating granulomatous inflammation remain ill-understood. CD154, the ligand of CD40, is a key mediator of inflammation. CD154 confers a proinflammatory phenotype to macrophages and controls several macrophagic functions. Here, we studied the contribution of CD154 in a mouse model of toxic liver injury with carbon tetrachloride and a model of absorbable suture graft. In both models, granulomas are triggered in response to endogenous persistent liver calcified necrotic lesions or by grafted sutures. CD154-deficient mice showed delayed clearance of carbon tetrachloride-induced liver calcified necrotic lesions and impaired progression of suture-induced granuloma. In vitro, CD154 stimulated phagocytosis of opsonized erythrocytes by macrophages, suggesting a potential mechanism for the altered granulomatous inflammation in CD154KO mice. These results suggest that CD154 may contribute to the natural history of granulomatous inflammation.
Collapse
|
131
|
Dasgupta Q, Movva S, Chatterjee K, Madras G. Controlled release from aspirin based linear biodegradable poly(anhydride esters) for anti-inflammatory activity. Int J Pharm 2017. [DOI: 10.1016/j.ijpharm.2017.06.065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
132
|
A Cell-Adhesive Plasma Polymerized Allylamine Coating Reduces the In Vivo Inflammatory Response Induced by Ti6Al4V Modified with Plasma Immersion Ion Implantation of Copper. J Funct Biomater 2017; 8:jfb8030030. [PMID: 28726761 PMCID: PMC5618281 DOI: 10.3390/jfb8030030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 01/16/2023] Open
Abstract
Copper (Cu) could be suitable to create anti-infective implants based on Titanium (Ti), for example by incorporating Cu into the implant surface using plasma immersion ion implantation (Cu-PIII). The cytotoxicity of Cu might be circumvented by an additional cell-adhesive plasma polymerized allylamine film (PPAAm). Thus, this study aimed to examine in vivo local inflammatory reactions for Ti6Al4V implants treated with Cu-PIII (Ti-Cu), alone or with an additional PPAAm film (Ti-Cu-PPAAm), compared to untreated implants (Ti). Successful Cu-PIII and PPAAm treatment was confirmed with X-ray Photoelectron Spectroscopy. Storage of Ti-Cu and Ti-Cu-PPAAm samples in double-distilled water for five days revealed a reduction of Cu release by PPAAm. Subsequently, Ti, Ti-Cu and Ti-Cu-PPAAm samples were simultaneously implanted into the neck musculature of 24 rats. After 7, 14 and 56 days, peri-implant tissue was retrieved from 8 rats/day for morphometric immunohistochemistry of different inflammatory cells. On day 56, Ti-Cu induced significantly stronger reactions compared to Ti (tissue macrophages, antigen-presenting cells, T lymphocytes) and to Ti-Cu-PPAAm (tissue macrophages, T lymphocytes, mast cells). The response for Ti-Cu-PPAAm was comparable with Ti. In conclusion, PPAAm reduced the inflammatory reactions caused by Cu-PIII. Combining both plasma processes could be useful to create antibacterial and tissue compatible Ti-based implants.
Collapse
|
133
|
Goonoo N. Modulating Immunological Responses of Electrospun Fibers for Tissue Engineering. ACTA ACUST UNITED AC 2017; 1:e1700093. [PMID: 32646177 DOI: 10.1002/adbi.201700093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Indexed: 12/28/2022]
Abstract
The promise of tissue engineering is to improve or restore functions of impaired tissues or organs. However, one of the biggest challenges to its translation to clinical applications is the lack of tissue integration and functionality. The plethora of cellular and molecular events occurring following scaffold implantation is a major bottleneck. Recent studies confirmed that inflammation is a crucial component influencing tissue regeneration. Immuno-modulation or immune-engineering has been proposed as a potential solution to overcome this key challenge in regenerative medicine. In this review, strategies to modify scaffold physicochemical properties through the use of the electrospinning technique to modulate host response and improve scaffold integration will be discussed. Electrospinning, being highly versatile allows the fabrication of ECM-mimicking scaffolds and also offers the possibility to control scaffold properties for instance, tailoring of fiber properties, chemical conjugation or physical adsorption of non-immunogenic materials on the scaffold surface, encapsulating cells or anti-inflammatory molecules within the scaffold. Such electrospun scaffold-based immune-engineering strategies can significantly improve the resulting outcomes of tissue engineering scaffolds.
Collapse
Affiliation(s)
- Nowsheen Goonoo
- Physical Chemistry I, Department of Chemistry and Biology & Research Center of Micro and Nanochemistry and Engineering (Cµ), University of Siegen, 57076, Siegen, Germany.,Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical and Biomaterials Research, MSIRI Building, University of Mauritius, Réduit, Mauritius
| |
Collapse
|
134
|
Calcium supplementation decreases BCP-induced inflammatory processes in blood cells through the NLRP3 inflammasome down-regulation. Acta Biomater 2017; 57:462-471. [PMID: 28528118 DOI: 10.1016/j.actbio.2017.05.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
Interaction of host blood with biomaterials is the first event occurring after implantation in a bone defect. This study aimed at investigating the cellular and molecular consequences arising at the interface between whole blood and biphasic calcium phosphate (BCP) particles. We observed that, due to calcium capture, BCP inhibited blood coagulation, and that this inhibition was reversed by calcium supplementation. Therefore, we studied the impact of calcium supplementation on BCP effects on blood cells. Comparative analysis of BCP and calcium supplemented-BCP (BCP/Ca) effects on blood cells showed that BCP as well as BCP/Ca induced monocyte proliferation, as well as a weak but significant hemolysis. Our data showed for the first time that calcium supplementation of BCP microparticles had anti-inflammatory properties compared to BCP alone that induced an inflammatory response in blood cells. Our results strongly suggest that the anti-inflammatory property of calcium supplemented-BCP results from its down-modulating effect on P2X7R gene expression and its capacity to inhibit ATP/P2X7R interactions, decreasing the NLRP3 inflammasome activation. Considering that monocytes have a vast regenerative potential, and since the excessive inflammation often observed after bone substitutes implantation limits their performance, our results might have great implications in terms of understanding the mechanisms leading to an efficient bone reconstruction. STATEMENT OF SIGNIFICANCE Although scaffolds and biomaterials unavoidably come into direct contact with blood during bone defect filling, whole blood-biomaterials interactions have been poorly explored. By studying in 3D the interactions between biphasic calcium phosphate (BCP) in microparticulate form and blood, we showed for the first time that calcium supplementation of BCP microparticles (BCP/Ca) has anti-inflammatory properties compared to BCP-induced inflammation in whole blood cells and provided information related to the molecular mechanisms involved. The present study also showed that BCP, as well as BCP/Ca particles stimulate monocyte proliferation. As monocytes represent a powerful target for regenerative therapies and as an excessive inflammation limits the performance of biomaterials in bone tissue engineering, our results might have great implications to improve bone reconstruction.
Collapse
|
135
|
Pasold J, Markhoff J, Tillmann J, Krogull M, Pisowocki P, Bader R. Direct influence of titanium and zirconia particles on the morphology and functionality of mature human osteoclasts. J Biomed Mater Res A 2017; 105:2608-2615. [PMID: 28544592 DOI: 10.1002/jbm.a.36114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 04/10/2017] [Accepted: 05/12/2017] [Indexed: 11/06/2022]
Abstract
Within the last ten years of biomedical implants, the focus is increasingly on bioceramics, specifically on zirconia (ZrO2 ). Hence, we analyzed the impact of ZrO2 particles in comparison to titanium particles on mature human osteoclasts (OCs) as little is known about the direct effect of wear particles on mature OCs and their role in the osteolytic process during aseptic endoprosthesis loosening. Changes in cell morphology and functionality of OCs incubated with particles in different concentrations were investigated in vitro. OCs tend to be enlarged after three days of cultivation with both types of particles, especially with high concentrations of ZrO2 , suggesting increased cell fusion. Further, we identified significantly increased expression of OC specific and bone matrix related genes: VNR, RANK, TRAP, and CTSK pointing on a direct stimulatory particle effect on the functionality of mature OCs. In completion, we quantified the bone resorption activity of particle treated mature OCs but could not detect a significant difference in bone resorption compared to OCs cultivated without particles. However, we could identify significantly higher gene expression of MMP-1 in particle treated OCs compared to untreated control OCs after three days of incubation. We also detected an impaired production of the tissue inhibitor of metalloproteinase, especially for OCs treated with high ZrO2 concentrations. In conclusion, our in vitro data show that abrasion particles could have a direct influence on mature OCs and therefore could promote increased OC-mediated bone resorption during aseptic loosening of total joint replacements. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2608-2615, 2017.
Collapse
Affiliation(s)
- Juliane Pasold
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| | - Jana Markhoff
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| | - Jenny Tillmann
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| | - Martin Krogull
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| | - Phillip Pisowocki
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| | - Rainer Bader
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Doberaner Strasse 142, Rostock, 18057, Germany
| |
Collapse
|
136
|
Kianoush F, Nematollahi M, Waterfield JD, Brunette DM. Regulation of RAW264.7 macrophage polarization on smooth and rough surface topographies by galectin-3. J Biomed Mater Res A 2017; 105:2499-2509. [PMID: 28498622 DOI: 10.1002/jbm.a.36107] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 11/05/2022]
Abstract
Recognition of topographical features induces phenotypic changes in macrophages although the receptors and signaling pathways are not completely characterized. As integrin molecules in focal adhesions/podosomes are in intimate contact with topography and topography modulates the NFkB pathway through cholesterol enriched raft-associated adhesive signaling structures we hypothesized that a cell-surface signaling complex comprised of galectin-3 together with its ligand CD98 and integrinβ1 is important for topography-directed lineage determination. This study used polished, sand blasted and acid etched (SLA) surfaces and two novel grooved topographies (G1 and G2) produced by anisotropic etching of Si <1 1 0> to evaluate the role of galectin-3 in macrophage polarization in RAW 264.7 macrophages, as determined by gene expression and morphology. In the presence of the galectin-3 inhibitor, lactose, the M2 marker (mannose receptor) was down-regulated while the M1 marker (iNOS) was up-regulated on smooth and rough surfaces. This skewing of phenotype suggests a role for galectin-3 in macrophage polarization towards the M2 phenotype. Additionally, we evaluated the role of PI3K on polarization using PI3K inhibitor LY294002. We found that the M2 marker was down-regulated on both PO (surface polished) and G1 surfaces implicating PI3K in lineage determination. We also found that surface topography altered cell morphology; macrophages had a larger area on G2 surfaces. Lactose treatment significantly reduced the cell area on all topographies suggesting that the galectin-3 is also involved in signaling complexes triggering the rearrangement of the actin cytoskeleton. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2499-2509, 2017.
Collapse
Affiliation(s)
- F Kianoush
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - M Nematollahi
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - J D Waterfield
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - D M Brunette
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
137
|
Lamichhane S, Anderson JA, Vierhout T, Remund T, Sun H, Kelly P. Polytetrafluoroethylene topographies determine the adhesion, activation, and foreign body giant cell formation of macrophages. J Biomed Mater Res A 2017; 105:2441-2450. [DOI: 10.1002/jbm.a.36099] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/06/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Sujan Lamichhane
- Biomedical Engineering; The University of South Dakota; 4800 N. Career Avenue Sioux Falls South Dakota 57107
| | - Jordan A. Anderson
- Biomedical Engineering; The University of South Dakota; 4800 N. Career Avenue Sioux Falls South Dakota 57107
| | - Thomas Vierhout
- Biomedical Engineering; The University of South Dakota; 4800 N. Career Avenue Sioux Falls South Dakota 57107
| | - Tyler Remund
- Sanford Research; 2301 East 60 Street North Sioux Falls South Dakota 57104
| | - Hongli Sun
- Biomedical Engineering; The University of South Dakota; 4800 N. Career Avenue Sioux Falls South Dakota 57107
| | - Patrick Kelly
- Sanford Health; 1305 West 18 Street Sioux Falls South Dakota 57105
| |
Collapse
|
138
|
Doty AC, Weinstein DG, Hirota K, Olsen KF, Ackermann R, Wang Y, Choi S, Schwendeman SP. Mechanisms of in vivo release of triamcinolone acetonide from PLGA microspheres. J Control Release 2017; 256:19-25. [DOI: 10.1016/j.jconrel.2017.03.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 03/18/2017] [Indexed: 02/05/2023]
|
139
|
Han S, Chen Z, Han P, Hu Q, Xiao Y. Activation of Macrophages by Lipopolysaccharide for Assessing the Immunomodulatory Property of Biomaterials<sup/>. Tissue Eng Part A 2017; 23:1100-1109. [PMID: 28346799 DOI: 10.1089/ten.tea.2016.0501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The design paradigm of biomaterials has been changed to ones with favorable immunomodulatory effects, indicating the importance of accurately evaluating the immunomodulatory properties of biomaterials. Among all the immune cells macrophages receive most attention, due to their plasticity and multiple roles in the materials and host interactions, and thereby become model immune cells for the evaluation of immunomodulatory properties of biomaterials in many studies. Lipopolysaccharides (LPS), a polysaccharide in the outer membrane of Gram-negative bacteria, elicit strong immune responses, which was often applied to activate macrophages, resulting in a proinflammatory M1 phenotype, and the release of proinflammatory cytokines, including tumor necrosis factor alpha (TNFα), interleukin (IL)-1, and IL-6. However, there is no consensus on how to apply macrophages and LPS to detect the immunomodulatory properties of biomaterials. The lack of scientific consideration of this issue has led to some inaccurate and insufficient conclusions on the immunomodulatory properties of biomaterials, and inconsistences between different research groups. In this study, we carried out a systemic study to investigate the stimulatory effects of LPS with different times, doses, and conditions on the activation of macrophages. An experimental pathway was proposed accordingly for the activation of macrophages using LPS for assessing the immunomodulatory property of biomaterials.
Collapse
Affiliation(s)
- Shengwei Han
- 1 Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China .,3 Institute of Health and Biomedical Innovation, the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia
| | - Zetao Chen
- 2 Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China .,3 Institute of Health and Biomedical Innovation, the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia
| | - Pingping Han
- 3 Institute of Health and Biomedical Innovation, the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia
| | - Qingang Hu
- 1 Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Yin Xiao
- 3 Institute of Health and Biomedical Innovation, the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
140
|
Wang J, Liu D, Guo B, Yang X, Chen X, Zhu X, Fan Y, Zhang X. Role of biphasic calcium phosphate ceramic-mediated secretion of signaling molecules by macrophages in migration and osteoblastic differentiation of MSCs. Acta Biomater 2017; 51:447-460. [PMID: 28126596 DOI: 10.1016/j.actbio.2017.01.059] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/22/2017] [Indexed: 02/05/2023]
Abstract
The inflammatory reaction initiates fracture healing and could play a role in the osteoinductive effect of calcium phosphate (CaP) ceramics, which has been widely confirmed; however, the underlying mechanism has not been fully elucidated. In this study, various signaling molecules from macrophages under the stimulation of osteoinductive biphasic calcium phosphate (BCP) ceramic and its degradation products were examined and evaluated for their influence on the migration and osteoblastic differentiation of mesenchymal stem cells (MSCs). The results of cellular experiments confirmed that the gene expression of most inflammatory factors (IL-1, IL-6 and MCP-1) and growth factors (VEGF, PDGF and EGF) by macrophages were up-regulated to varying degrees by BCP ceramic and its degradation products. Cell migration tests demonstrated that the conditioned media (CMs), which contained abundant signaling molecules secreted by macrophages cultured on BCP ceramic and its degradation products, promoted the migration of MSCs. qRT-PCR analysis indicated that CMs promoted the gene expression of osteogenic markers (ALP, COL-I, OSX, BSP and OPN) in MSCs. ALP activity and mineralization staining further confirmed that CMs promoted the osteoblastic differentiation of MSCs. The present study confirmed the correlation between the inflammatory reaction and osteoinductive capacity of BCP ceramic. The ceramic itself and its degradation products can induce macrophages to express and secrete various signaling molecules, which then recruit and promote the MSCs to differentiate into osteoblasts. Compared with BCP conditioned media, degradation particles played a more substantial role in this process. Thus, inflammation initiated by BCP ceramic and its degradation products could be necessary for osteoinduction by the ceramic. STATEMENT OF SIGNIFICANCE It is known that the inflammatory reaction initiates fracture healing. The aim of this study was to examine whether osteoinductive BCP ceramics could cause macrophages to change their secretion patterns and whether the secreted cytokines could affect migration and osteoblastic differentiation of MSCs. Moreover, the duration of inflammation could be influenced by the local ionic environment and the degradation products of the implant. Our experimental results revealed the correlation between the inflammatory reaction and osteoinductive capacity of BCP ceramic. The ceramic itself and its degradation products can induce macrophages to express and secrete various signaling molecules, which then recruit and promote the MSCs to differentiate into osteoblasts. Compared with ionic microenvironment, degradation particles played a more substantial role in this process. Therefore, the appropriate inflammation initiated by BCP ceramic and its degradation products could be essential for osteoinduction by the ceramic. We believe that the present study improves the understanding of the effect of biomaterial-mediated inflammation on MSC migration and differentiation and established a preliminary correlation between the immune system and osteoinduction by biomaterials.
Collapse
|
141
|
Morris AH, Stamer DK, Kyriakides TR. The host response to naturally-derived extracellular matrix biomaterials. Semin Immunol 2017; 29:72-91. [PMID: 28274693 DOI: 10.1016/j.smim.2017.01.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 12/13/2022]
Abstract
Biomaterials based on natural materials including decellularized tissues and tissue-derived hydrogels are becoming more widely used for clinical applications. Because of their native composition and structure, these biomaterials induce a distinct form of the foreign body response that differs from that of non-native biomaterials. Differences include direct interactions with cells via preserved moieties as well as the ability to undergo remodeling. Moreover, these biomaterials could elicit adaptive immune responses due to the presence of modified native molecules. Therefore, these biomaterials present unique challenges in terms of understanding the progression of the foreign body response. This review covers this response to natural materials including natural polymers, decellularized tissues, cell-derived matrix, tissue derived hydrogels, and biohybrid materials. With the expansion of the fields of regenerative medicine and tissue engineering, the current repertoire of biomaterials has also expanded and requires continuous investigation of the responses they elicit.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States
| | - D K Stamer
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - T R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Department of Pathology, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.
| |
Collapse
|
142
|
Zhang J, Hiromoto S, Yamazaki T, Huang H, Jia G, Li H, Yuan G. Macrophage phagocytosis of biomedical Mg alloy degradation products prepared by electrochemical method. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 75:1178-1183. [PMID: 28415404 DOI: 10.1016/j.msec.2017.02.126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/23/2016] [Accepted: 02/24/2017] [Indexed: 11/28/2022]
Abstract
Biomedical Mg alloy is promising for its widespread use clinically. In vitro and in vivo studies showed that the degradation products of biomedical Mg alloy were composed of O, P, Ca, Mg and other alloying elements. However, little is known about the metabolism of the degradation products. In this study, the in vitro macrophage phagocytosis of the degradation products of a biomedical Mg-Nd-Zn-Zr alloy was directly observed. This result affirms the necessity to investigate the long-term fate of Mg alloy degradation products in physiological environments. Besides, an electrochemical method was proposed to prepare enough amount of degradation products in vitro efficiently.
Collapse
Affiliation(s)
- Jian Zhang
- National Engineering Research Center of Light Alloy Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai 200240, China; Biomaterials Unit, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science, Tsukuba 305-0047, Japan
| | - Sachiko Hiromoto
- Biomaterials Unit, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science, Tsukuba 305-0047, Japan.
| | - Tomohiko Yamazaki
- Biomaterials Unit, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science, Tsukuba 305-0047, Japan
| | - Hua Huang
- National Engineering Research Center of Light Alloy Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gaozhi Jia
- National Engineering Research Center of Light Alloy Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haiyan Li
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guangyin Yuan
- National Engineering Research Center of Light Alloy Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
143
|
Wang WC, Xia YM, Yang B, Su XN, Chen JK, Li W, Jiang T. Protective Effects of Tyrosol against LPS-Induced Acute Lung Injury via Inhibiting NF-κB and AP-1 Activation and Activating the HO-1/Nrf2 Pathways. Biol Pharm Bull 2017; 40:583-593. [PMID: 28190857 DOI: 10.1248/bpb.b16-00756] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tyrosol (Tyr) is a natural antioxidant that displays anti-oxidant and anti-inflammatory properties. The present study aimed to investigate the effect and mechanism of Tyr on lipopolysaccharide (LPS)-induced acute lung injury (ALI). In a mouse model, we found that pretreatment with Tyr significantly improved survival rate, attenuated lung permeability, ameliorated histopathological alterations, reduced expression of the inflammatory mediators and improved expression of the antioxidant enzyme. Further study revealed that Tyr markedly inhibited nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) activation at both in vivo and in vitro levels. To investigate the underlying mechanism, we examined the impact of Tyr on the heme oxygenase (HO)-1/nuclear factor erythroid-2 related factor 2 (Nrf2) pathway in vivo and in vitro. The results showed that Tyr significantly improved the expression of HO-1 and the activation of Nrf2. This study offers novel evidence to support the efficacy of Tyr against ALI, which helps to clarify the underlying causes of the therapeutic effects behind Tyr.
Collapse
Affiliation(s)
- Wen-Chen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University
| | - Yan-Min Xia
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Center Hospital
| | - Xiang-Ni Su
- Department of Nursing, Fourth Military Medical University
| | - Jia-Kuan Chen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University
| | - Wei Li
- Department of Histology and Embryology, Fourth Military Medical University
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University
| |
Collapse
|
144
|
Brochhausen C, Schmitt VH, Mamilos A, Schmitt C, Planck CNE, Rajab TK, Hierlemann H, Kirkpatrick CJ. Expression of CD68 positive macrophages in the use of different barrier materials to prevent peritoneal adhesions-an animal study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:15. [PMID: 27995493 PMCID: PMC5167770 DOI: 10.1007/s10856-016-5821-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/26/2016] [Indexed: 05/19/2023]
Abstract
In preventing postoperative adhesion formation the optimal barrier material has still not been found. It is therefore imperative to assess the biocompatibility of potential barrier devices. Macrophages play a decisive role in the regulation of wound healing, tissue regeneration and foreign body reaction. Since the number of CD68-positive macrophages represents an important parameter within biomaterial testing, in the present study it was analysed whether a correlation exists between the total number of CD68-positive macrophages and the extent of fibrosis or inflammation in peritoneal adhesion prevention using biomaterials. After standardized peritoneal wounding, Wistar rats were treated with five adhesion barriers or remained untreated as a control. After 14 days, animals were sacrificed and the treated areas were evaluated histomorphologically and immunohistologically. A heterogeneous pattern of macrophage count in relation to fibrosis or inflammation was found. While some groups described a moderate macrophage infiltration without fibrosis, others showed similar numbers of macrophages, but accompanied by moderate fibrosis. Moreover, a minimal number of macrophages was associated with minimal fibrosis. Mild inflammation was seen both with minimal and moderate macrophage infiltration. Altogether, no correlation could be established between the tissue response and the count of CD68-positive macrophages. With a view to macrophage heterogeneity further studies are required to determine the different macrophage subpopulations and clarify the role of these in the tissue responses to barrier materials.
Collapse
Affiliation(s)
| | - Volker H Schmitt
- Cardiology I, Centre for Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Andreas Mamilos
- REPAIR-lab, Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Christine Schmitt
- Department of Internal Medicine, St. Vincenz and Elisabeth Hospital of Mainz (KKM), Mainz, Germany
| | - Constanze N E Planck
- Department of Gynaecology and Obstetrics, University of Tuebingen, Tuebingen, Germany
- German Centre of Biomaterials and Artificial Organs e.V. Denkendorf, Denkendorf, Germany
| | - Taufiek K Rajab
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Helmut Hierlemann
- German Centre of Biomaterials and Artificial Organs e.V. Denkendorf, Denkendorf, Germany
- Institute of Textile Technology and Process Engineering, Denkendorf, Germany
| | - C James Kirkpatrick
- Department of Biomaterials, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
145
|
Hayrapetyan A, Surjandi S, Lemsom EEPJ, Wolters MMMW, Jansen JA, van den Beucken JJJP. Coculture effects on the osteogenic differentiation of human mesenchymal stromal cells. Tissue Eng Regen Med 2016; 13:713-723. [PMID: 30603452 DOI: 10.1007/s13770-016-0008-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/04/2016] [Accepted: 03/10/2016] [Indexed: 12/16/2022] Open
Abstract
Cell-based bone regeneration is generally pursued based on single cell type approaches, for which human adipose tissue-derived mesenchymal stromal cells (AT-MSCs) are frequently used, owing to their easy accessibility and relatively large yield. In view of multiple cell types involved in physiological bone regeneration, this study aimed to evaluate the osteogenic differentiation of AT-MSCs upon co-culture with endothelial cells or macrophages in a direct or indirect in vitro co-culture set-up. Our hypotheses were that 1) endothelial cells and macrophages stimulate AT-MSCs proliferation and osteogenic differentiation and that 2) these two cell types will more profoundly affect osteogenic differentiation of AT-MSCs in a direct compared to an indirect co-culture set-up, because of the possibility for both cell-cell interactions and effects of secreted soluble factors in the former. Osteogenic differentiation of AT-MSCs was stimulated by endothelial cells, particularly in direct co-cultures. Although initial numbers of AT-MSCs in co-culture with endothelial cells were 50% compared to monoculture controls, equal levels of mineralization were achieved. Macrophages showed a variable effect on AT-MSCs behavior for indirect co-cultures and a negative effect on osteogenic differentiation of AT-MSCs in direct co-cultures, the latter likely due to species differences of the cell types used. The results of this study demonstrate potential for cell combination strategies in bone regenerative therapies.
Collapse
Affiliation(s)
| | - Soraya Surjandi
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | | | | | - John A Jansen
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | - Jeroen J J P van den Beucken
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands.,2Department of Biomaterials (309), Radboudumc, Ph van Leijdenlaan 25, Nijmegen, 6525 the Netherlands
| |
Collapse
|
146
|
Bygd HC, Bratlie KM. Investigating the Synergistic Effects of Combined Modified Alginates on Macrophage Phenotype. Polymers (Basel) 2016; 8:E422. [PMID: 30974698 PMCID: PMC6432444 DOI: 10.3390/polym8120422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/17/2016] [Accepted: 12/01/2016] [Indexed: 01/06/2023] Open
Abstract
Understanding macrophage responses to biomaterials is crucial to the success of implanted medical devices, tissue engineering scaffolds, and drug delivery vehicles. Cellular responses to materials may depend synergistically on multiple surface chemistries, due to the polyvalent nature of cell⁻ligand interactions. Previous work in our lab found that different surface functionalities of chemically modified alginate could sway macrophage phenotype toward either the pro-inflammatory or pro-angiogenic phenotype. Using these findings, this research aims to understand the relationship between combined material surface chemistries and macrophage phenotype. Tumor necrosis factor-α (TNF-α) secretion, nitrite production, and arginase activity were measured and used to determine the ability of the materials to alter macrophage phenotype. Cooperative relationships between pairwise modifications of alginate were determined by calculating synergy values for the aforementioned molecules. Several materials appeared to improve M1 to M2 macrophage reprogramming capabilities, giving valuable insight into the complexity of surface chemistries needed for optimal incorporation and survival of implanted biomaterials.
Collapse
Affiliation(s)
- Hannah C Bygd
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA.
- Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA.
- Division of Materials Science & Engineering, Ames National Laboratory, Ames, IA 50011, USA.
| |
Collapse
|
147
|
Guillory RJ, Bowen PK, Hopkins SP, Shearier ER, Earley EJ, Gillette AA, Aghion E, Bocks M, Drelich JW, Goldman J. Corrosion Characteristics Dictate the Long-Term Inflammatory Profile of Degradable Zinc Arterial Implants. ACS Biomater Sci Eng 2016; 2:2355-2364. [DOI: 10.1021/acsbiomaterials.6b00591] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | | | - Eli Aghion
- Department
of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Martin Bocks
- Division
of Pediatric Cardiology, University of Michigan Congenital Heart Center, Ann Arbor, Michigan 48109, United States
| | | | | |
Collapse
|
148
|
The impact of surface chemistry modification on macrophage polarisation. Immunobiology 2016; 221:1237-46. [DOI: 10.1016/j.imbio.2016.06.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/01/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
149
|
Kuo YL, Wang SG, Wu CY, Lee KC, Jao CJ, Chou SH, Chen YC. Functional gold nanoparticle-based antibacterial agents for nosocomial and antibiotic-resistant bacteria. Nanomedicine (Lond) 2016; 11:2497-510. [PMID: 27622499 DOI: 10.2217/nnm-2016-0232] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Medical treatments for bacterial-infections have become challenging because of the emergence of antibiotic-resistant bacterial strains. Thus, new therapeutics and antibiotics must be developed. MATERIALS & METHODS Arginine and tryptophan can target negatively-charged bacteria and penetrate bacterial cell membrane, respectively. Synthetic-peptides containing arginine, tryptophan and cysteine termini, in other words, (DVFLG)2REEW4C and (DVFLG)2REEW2C, as starting materials were mixed with aqueous tetrachloroauric acid to generate peptide-immobilized gold nanoparticles (i.e., [DVFLG]2REEW4C-AuNPs and [DVFLG]2REEW2C-AuNPs) through one-pot reactions. RESULTS & DISCUSSION The peptide immobilized AuNPs exhibit targeting capacity and antibacterial activity. Furthermore, (DVFLG)2REEW4C-AuNPs immobilized with a higher number of tryptophan molecules possess more effective antibacterial capacity than (DVFLG)2REEW2C-AuNPs. Nevertheless, they are not harmful for animal cells. The feasibility of using the peptide-AuNPs to inhibit the cell growth of bacterium-infected macrophages was demonstrated. CONCLUSION These results suggested that the proposed antibacterial AuNPs are effective antibacterial agents for Staphylococci, Enterococci and antibiotic-resistant bacterial strains. [Formula: see text].
Collapse
Affiliation(s)
- Yen-Ling Kuo
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Sin-Ge Wang
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Ching-Yi Wu
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Kai-Chieh Lee
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Chan-Jung Jao
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Shiu-Huey Chou
- Department of Life Science, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Chie Chen
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
150
|
Fonseca-García A, Pérez-Alvarez J, Barrera C, Medina J, Almaguer-Flores A, Sánchez RB, Rodil SE. The effect of simulated inflammatory conditions on the surface properties of titanium and stainless steel and their importance as biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 66:119-129. [DOI: 10.1016/j.msec.2016.04.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/29/2016] [Accepted: 04/11/2016] [Indexed: 12/26/2022]
|